Next Article in Journal
CCL2 Inhibition of Pro-Resolving Mediators Potentiates Neuroinflammation in Astrocytes
Next Article in Special Issue
Natural Alkaloids as Multi-Target Compounds towards Factors Implicated in Alzheimer’s Disease
Previous Article in Journal
Using ELP Repeats as a Scaffold for De Novo Construction of Gadolinium-Binding Domains within Multifunctional Recombinant Proteins for Targeted Delivery of Gadolinium to Tumour Cells
Previous Article in Special Issue
A Search for Cyclin-Dependent Kinase 4/6 Inhibitors by Pharmacophore-Based Virtual Screening, Molecular Docking, and Molecular Dynamic Simulations
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Tricyclodecan-9-yl-Xanthogenate (D609): Mechanism of Action and Pharmacological Applications

1
Department of Clinical Biochemistry, University of Kashmir, Srinagar 190006, India
2
Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
3
Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
4
Department of Pharmacy Practice, College of Pharmacy, AlMaarefa University, Ad Diriyah 13713, Saudi Arabia
5
Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(6), 3305; https://doi.org/10.3390/ijms23063305
Submission received: 21 February 2022 / Revised: 14 March 2022 / Accepted: 16 March 2022 / Published: 18 March 2022
(This article belongs to the Special Issue Multi-Target Directed Ligands in Drug Development)

Abstract

:
Tricyclodecan-9-yl xanthogenate (D609) is a synthetic tricyclic compound possessing a xanthate group. This xanthogenate compound is known for its diverse pharmacological properties. Over the last three decades, many studies have reported the biological activities of D609, including antioxidant, antiapoptotic, anticholinergic, anti-tumor, anti-inflammatory, anti-viral, anti-proliferative, and neuroprotective activities. Its mechanism of action is extensively attributed to its ability to cause the competitive inhibition of phosphatidylcholine (PC)-specific phospholipase C (PC-PLC) and sphingomyelin synthase (SMS). The inhibition of PCPLC or SMS affects secondary messengers with a lipidic nature, i.e., 1,2-diacylglycerol (DAG) and ceramide. Various in vitro/in vivo studies suggest that PCPLC and SMS inhibition regulate the cell cycle, block cellular proliferation, and induce differentiation. D609 acts as a pro-inflammatory cytokine antagonist and diminishes Aβ-stimulated toxicity. PCPLC enzymatic activity essentially requires Zn2+, and D609 might act as a potential chelator of Zn2+, thereby blocking PCPLC enzymatic activity. D609 also demonstrates promising results in reducing atherosclerotic plaque formation, post-stroke cerebral infarction, and cancer progression. The present compilation provides a comprehensive mechanistic insight into D609, including its chemistry, mechanism of action, and regulation of various pharmacological activities.

1. Introduction

Tricyclodecan-9-yl xanthogenate (D609) is a synthetic tricyclic compound featuring a xanthate group, known as a phosphate group analog (Figure 1). The first antiviral biological experiments did not commence until 1984, over fifty years after the initial synthesis was published [1,2]. The discovery of the various biological mechanisms that followed led to the detection of many other biological mechanisms, including anti-tumoral, antiviral, anti-apoptotic, and anti-inflammatory effects [3,4,5]. The D609 suppressed both acidic sphingomyelinase and phosphatidylcholine-specific phospholipase C (PC-PLC) activity due to its unique competitive inhibitory action on both these enzymes [6,7].
Recently, D609 was observed to be the single recorded sphingomyelin synthase (SMS) inhibitor that correlates with the clearance of amyloid-β peptide as well as metabolic syndrome [8]. Over the last 30 years, more than 700 reports on the biological activity of D609 have been published, predominantly for its lipid biology [9]. Essentially, D609 is an extremely sturdy regulator of lipid biology and lipid chemical biology [10]. There are eight stereoisomers and their enantiomers paired by three asymmetric centers in D609 [11,12]. The biological activities of D609 are typically stimulated by phosphatidylcholine (PC)-specific inhibition of phospholipase C (PC-PLC) [13,14]. The suppression of PC-PLC and/or SMS by D609 can be averted by lipid second messengers; ceramide and/or 1,2-diacylglycerol (DAG) [15,16]. According to reports, the inhibition of either SMS or PC-PLC regulated the cell cycle in various in vitro and in vivo experiments and suppressed proliferation while promoting differentiation. Xanthogenate compounds, including D609, possess potent antioxidant properties, and D609 also diminishes the Aß-stimulated toxicity [17,18,19]. Zn2+ is necessary for the enzymatic activity of PC-PLC, while chelation with Zn2+ leads to the suppression of D609 [20]. D609 also has a role in suppressing acidic sphingomyelinase [21,22] or down-regulating hypoxia-inducible factor-1a, even though these are downstream actions imperative to the PC-PLC suppression [23]. PC-PLC mammals′ characterization is restricted to blocking enzymatic activity (commonly calculated with bacterial PC-PLC as a standard, called Amplex red assay) [6]. Until now, no cloning of PC-PLC mammals has occurred, and no systemic and temporal specifics are yet available. D609 exhibited promising results for decreasing atherosclerotic plaque formation (PC-PLC inhibition), as well as stroke infarction [24,25]. As a pro-inflammatory cytokine receptor, PC-PLC is responsible for the action of D609. Following oxidation, D609 possesses a free thiol group that releases a disulfide that mimics glutathione. The resulting molecule, which regenerates D609, is a glutathione reductase substratum [18]. D609 defends the brain and cultures of neurons from potential triggers of AD (Alzheimer′s disease) and oxidative stress caused by Aβ cytotoxicity, according to recent reports [18,26]. Mitochondria are vital organelles containing pro and anti-apoptotic protein factors. Under in vitro conditions, the administration of D609 is believed to mediate neuroprotection against apoptosis and free-radical-induced mitochondrial damage [18]. In vitro oxidants, Fe2+/H2O2 (hydroxyl free radicals), and AD relevant peptides 1–42 A (1–42) and 2,2-azobis (2-amidinopropane) dihydrochloride were used to recover brain mitochondria from gerbils 1 h after the intraperitoneal (i.p.) administration of D609 (AAPH, alkoxyl, and peroxyl free radicals).
Protein carbonyl, protein-bound hydroxynonenal (HNE) (a lipid peroxidation product), 3-nitrotyrosine (3-NT), and cytochrome-c release were all significantly lower in oxidant-treated brain mitochondria isolated from saline-injected gerbils [18]. Moreover, D609 therapy successfully maintained the (reduced glutathione/oxidized glutathione) GSH/GSSG ratio of oxidant-treated mitochondria. D609-treated glutathione reductase (GR), glutathione peroxidase (GPx), and glutathione S-transferase (GST) gerbils improved brain growth, which reinforced the concept that D609 works as GSH. The anti-apoptotic properties of this molecule are studied as possible therapeutics for oxidative stress-induced neurodegenerative diseases, such as AD [27].
In this review, a comprehensive analysis of the mechanism of D609 and its pharmacological action in different serious ailments and its role is discussed in detail. A large proportion of the study is devoted to the mechanism of D609 as an antiviral, antioxidant, anti-inflammatory, anti-tumoral, antiapoptotic, anticholinergic activity which are comprehensively documented.

2. Chemistry of D609

D609 comprises a total of eight stereoisomers in addition to their enantiomers, which are allocated among three asymmetric centers in D609 [2,28]. The initial reports of D609 date back to the year 2002 [29], and, currently, the drug is also available from several commercial sources. Until now, no comprehensive study has been carried out to examine the impact on the biological behavior of D609s. In fact, due to the enormous difficulties of assigning proper stereochemistry to modern chemical analytics, no chiral or relative stereochemistry information is generally present in commercially available D609s. Vibrational circular dichroism (VCD) spectroscopy with ab initio theoretical computations has been developed during the last decade to determine the underlying structure of D609. Through the cellular vibrational transition, VCD estimates the differential absorption of circularly polarized IR radiation on the chiral molecule [11].

3. Mechanism of Action of D609

3.1. PC Specific-PC-PLC

PC-PLC (66 kDa) hydrolyzes phosphocholine and 1,2-diacylglycerol PCs to produce DAG. Although scientists have purified bacterial PC-PLC, mammalian PC-PLC is yet to be cloned. Its sequence is still undetermined, thereby restricting the identification of D609 actions by a mammalian enzyme [30]. Rabbit polyclonal antibodies demonstrated cross-reactivity with mammalian PC-PLC to Bacillus cereus PC-PLCI in the occurrence or lack of an essential fibroblast growth factor (ßFGF). The suppression of PC-PLC by D609 blocked replication and permitted different cell systems to be distinguished (Figure 2). PC-PLC cells also have a role in vascular endothelial apoptosis and senesis. In a cell-cycle-dependent manner, the PC-PLC expression varied inversely with cell division cycle protein 20 (CDC20) homolog and CDC20 overexpression stimulated ubiquitin-proteasome pathway-mediated PC-PLC degradation [31]. A highly glycosylated transmembrane protein, cluster of differentiation-16 (CD16), also mediated PC-PLC expression in (natural killer) NK cells. D609 treatment caused a dramatic reduction in the expression of PC-PLC and CD16 receptor [32]. Acts of D609 due to PC-PLC inhibition include the inhibited post-stroke production of hypoxia-inducible factor 1-alpha (HIF-1 α), the protection of tumor necrosis factor (TNF-α) or lethal shock mediated by LPS in mice, diminished expression of cytokines in macrophages induced by lipopolysaccharide (LPS) and safeguarding immature neurons (non-expressed glutamate receptors) against oxidative toxic glutamate [33]. According to reports, inhibiting PC-PLC with D609 promotes phospholipase D (PLD) action in UMR-106 osteoblastic cells, which could be due to counterbalancing effects of D609 or direct enhancement of PLD [34].
Derivatives of xanthate fit into the active core of PC-PLC with a lipophilic chain, and the dithiocarbonate group possibly functions as a phosphate replacement and binds to the active site of Zn2+ ions [3,14,35,36]. Generally, Amplex Red assay is employed to monitor the operation of PC-PLC. This assay works on the theory that PC-PLC hydrolyzes DAG-PC and phosphocholine-PC. The enzyme alkaline phosphatase converts phosphocholine to choline. Choline oxidase is converted to betaine, which produces H2O2. The Amplex Red reagent is stoichiometrically oxidized by hydrogen peroxide in the presence of horseradish peroxidase (HRPO) to produce fluorescent resorufin, which can be measured spectrophotometrically or fluorometrically. [37,38].

3.2. Sphingolipid Metabolism

In de novo ceramide biosynthesis, serine palmitoyltransferase (SPT) catalyzes the first and rate-limiting step. In the form of SM and DAG, the phosphocholine group from PC is converted to ceramide in the PC-ceramide membrane framework. Golgi apparatus has SMS1, while plasma membrane has SMS2. These two types of SMS are inhibited by D609 [6,39,40,41,42]. D609′s effects on PC-PLC inhibition appear to include SMS inhibition, according to studies [43]. De novo ceramide production was similarly boosted by D609, which might be explained by SPT stimulation [44]. In one rat stroke model, D609′s neuroprotection was due to the inhibition of SMS, which induced the accumulation of ceramide and influenced cell-cycle events. Lipid rafts/microdomains and the transport of fatty acids to the cell are associated with scavenger CD36/fatty acid translocase. The translocation and function of CD36 can be impacted by the ceramide [6,45]. Levels of ceramide may be critical for its signaling and may induce retinoblastoma dephosphorylation, triggering the arrest of the cell cycle (Rb) [46,47,48]. D609 suppressed ßFGF-stimulated astrocyte spread, probably due to SMS inhibition and an elevation in levels of ceramide [49]. By inhibiting SMS and upregulating the cyclin-dependent kinase (Cdk) inhibitors p27 and p21, D609 can cause the cell-cycle arrest and increase ceramide levels [50,51,52]. Ceramide may stimulate p27 and p21 expression by activating c-myc regulating protein phosphatase 2A (PP2A), which suppresses p21 and p27 expression [53]. Since the expression of cellular myelocytomatosis oncogene (c-myc) is not blocked by D609, these downstream effects are likely to increase ceramide levels.

4. Pharmacological Properties of D609

4.1. Antioxidant

D609 is an effective antioxidant in numerous studies. As with Alzheimer’s disease (AD), many neurological disorders are linked to oxidative stress. Intracellular neurofibrillary tangles (NFTs), extracellular amyloid protein deposits (primarily consisting of hyperphosphorylated tau protein), mitochondrial malfunction, synapse degradation, and apoptosis or cell death are all symptoms of AD [18,54]. The reduced energy metabolism in AD could be due to the oxidative failure of some essential metabolic or mitochondrial enzymes, which would lead to increased ROS [55]. Mitochondria is well-known as a key cellular energy-producing organelle. The production of ROS, in turn, leads to neuronal oxidative damage, which is dependent on these organelles [56,57]. In mitochondria, specific anti-apoptotic signals and pro-apoptotic defenses unite. Protein factors such as the second mitochondria-derived activator of caspase (SMAC) protein, and direct inhibitor of apoptosis-binding protein with low pi (DIABLO), cytochrome-C, apoptosis-inducing factor (AIF), and apoptotic protease activating factor 1 (Apaf1) produced during mitochondrial oxidative stress trigger caspase-independent and caspase-dependent pathways, leading to programmed cell death [58]. Mitochondria regulate intracellular Ca2+ homeostasis, generate ATP, and produce endogenous ROS. A higher concentration of mitochondrial calcium leads to superoxide formation and pro-apoptotic mitochondrial protein release, culminating in cell death [59].
DNA, RNA, protein, and lipid peroxidation, as well as neuronal malfunction or death, all occur in the AD brains [60]. Current research suggests that lipid peroxidation and protein oxidation in brains with mild cognitive impairment indicate that oxidative stress occurs early in the AD pathogenesis [61]. Understanding oxidative stress and its associated diseases, necessitates a comprehensive understanding of mitochondrial ROS activity and its effect on neuronal processes. The transport of mitochondrial electrons may be a source of ROS [62]. ROS such as hydroxyl radical (OH), hydrogen peroxide (H2O2), peroxynitrite (ONOO), and superoxide anion (O2−) have long been known to cause neurodegeneration [63]. The mitochondrial membrane potential promotes the release of cytochrome-C into the cytoplasm and enhances the activity of caspase-3, signifying that mitochondrial mtDNA-derived malfunction contributes to Alzheimer′s disease [64]. Many studies confirm D609 to be helpful in a variety of central nervous system (CNS) and other neurological disorders [18].
D609 exhibits antioxidant and glutathione mimic properties due to its thiol content. Reduced glutathione (GSH) has been studied for its mimetic/antioxidant activity in the Aß toxicity trials [16]. D609′s xanthate group can quickly oxidize into a disulfide, which is a glutathione reductase substrate for the D609 regeneration [26]. In a variety of neurological disorders and CNS traumas, ROS and lipid peroxidation/oxidation have been found to play a crucial role in tissue pathophysiology. D609′s antioxidant/glutathione mimetic properties reduced ROS and oxidized PC (OxPC) growth, thereby benefiting these CNS pathologies [18]; D609 is a compound with anti-inflammatory and antiviral properties [4]. The majority of these occurrences are related to D609′s inhibition of PC-PLC [65]. This inhibition prevents secondary messenger diacylglycerol (DAG) synthesis, which in turn inhibits PKC and acidic sphingomyelinase (aSMase) [66]. Although it is unclear which ROS species D609 can effectively scavenge, this xanthate can scavenge hydroxyl radicals. Since xanthates have the good reductive ability, they are likely to react with other ROS [18]. As with pyrrolidine dithiocarbamate, a well-known antioxidant; D609 also inhibits dihydrorhodamine Fenton-reaction-induced oxidation in a concentration-dependent manner. The free thiol moiety in xanthate anions and protonated xanthic acid makes them highly reductive [6,17]. D609 also prevents alpha-phenyl-tert-butylnitron free-radical spin adducts and synaptosomal membrane lipid peroxidation, when Fenton reactions are used [67]. D609 has been shown to protect intracellular GSH, a key intracellular defense molecule in neurons, from oxidative stress and radiation [17,68]. D609, mimetic glutathione, has recently been shown to protect primary neuronal cultures from amyloid-peptide (1–42) induced oxidative stress and neurotoxicity in vitro and in vivo synaptosomes [69,70].

4.2. Anti-Viral

D609 was created with the intention of turning it into an antiviral agent. D609 and other Xanthates were first described as broad-spectrum antiviral compounds [1]. D609 exists as an enantiomeric pair of four diastereomers, with unknown variable mixtures of these eight isomers in industrial preparations. In antiviral and PC-PLC inhibition assays, isomers have varying biochemical efficacy [65]. Lumavita (Basel, Switzerland) is working on an antiviral drug called Letermovir-601 (LMV-601), which is a pure enantiomeric isomer of D609. Over time, LMV-601, a PC-PLC inhibitor, decreased the high-risk Human Papillomavirus (HPV) expression in types 16, 18, and 31, and exacerbated defects of pre-cancer cervical cell [6]. Tricyclodecan-9-yl-xanthogenate, an antiviral xanthate compound, can inhibit RNA and DNA viruses in vitro [71]. Infectivity assays and Western blot analysis have shown that it can prevent infectious HIV from spreading to chronically infected lymphoma cell (KE37-III) tissue-culture medium [72]. HIV-specific proteins, on the other hand, have accumulated intracellularly. At D609 doses that allowed mitotic cell divisions, de novo HIV replication commencement after infection with permissive KE37-1 cells was entirely stopped. Furthermore, the loss of HIV replicative intermediate DNA demonstrated the xanthate compound′s selective anti-viral efficacy. Within these cells, the cellular gene expression of c-myc remains unchanged [72].
The human respiratory syncytial virus (RSV) is a cytoplasmic, enveloped virus with negative RNA polarity. In newborns and young children, paramyxovirus is among the common causes of respiratory tract infections [73]. In human epithelial cells, the antiviral compound D609 prevents the development of the respiratory syncytial (RS) virus (Hep 2) [74]. Viral protein aggregation, the viral phosphorylation of phosphoprotein, infectious particles levels, and extracellular antigens all decreased after treatment with D609. When the polarity was positive or negative, the viral proteins’ relative accumulation was also unbalanced, but there were no variations in viral RNA volume. Furthermore, nucleocapsid formation was not hampered [75]. HSV-1 (Herpes simplex virus-1) replication was prevented by D609 without causing cytotoxicity. It inhibited HSV-1 encoded protein kinase and decreased virus-infected cell polypeptide phosphorylation (US3 PK). At concentrations greater than 3.8 µM, virus production decreased by D609, with absolute inhibition at 75.2 µM at or below 1 PFU/cell MOI. D609 can be given up to 7 h after infection and still prevent virus replication. These findings indicate that D609′s antiviral activity is mediated by the protein kinase inhibition and phosphorylation of protein, which influences late HSV replication. As a result of the above research, it is clear that D609 prevents the development of many virus-related diseases [76]. The role of D609 in various diseases and its mechanisms are summarized in Figure 3.

4.3. Anti-Inflammatory

D609 has also demonstrated significant potential anti-inflammatory activities [77]. Immunization of uveal melanin protein causes experimental melanin–protein induced uveitis (EMIU), which is a form of chronic autoimmune uveitis. The induction of inductive nitric oxide synthase (iNOS) is prevented by D609, a basic inhibitor of phosphatidylcholine-specific phospholipase C [78,79]. Thirty-five (Lewis) rats with EMIU were given either PBS or D609 in two separate experiments. D609 had a significant inhibitory effect on EMIU during this study [80]. D609-treated eyes had lower peroxide and nitrite levels, higher SOD levels, and lower iNOS mRNA expressions than PBS-treated eyes. The Fas and FasL levels in D609-injected animals′ eyes and lymph nodes increased significantly. D609-treated rats had increased DNA fragmentation in their lymph nodes. D609 inhibited EMIU by quenching NO and initiating programmed cell death, which had formerly been inhibited by NO, as well as displaying anti-inflammatory properties [79]. D609, a NO scavenger, was recently shown to be effective at preventing allergic encephalomyelitis in animals [81,82]. D609, in addition to inhibiting NO, can also inhibit other inflammatory mediators. By preventing the PC-PLC pathway, D609 inhibits the development of NOS activity. In response to inflammation, the PC-PLC pathway, which plays an important role, is activated [83,84]. D609 reduces IL-1α, IL-6, and NO release in endotoxin shock mice, according to Tschaikowsky et al. D609′s inhibition of NO synthesis has a much greater protective effect than D609′s inhibition of other inflammatory mediators. It has also been shown that D609, a PC-PLC and NOS inhibitor, inhibited NO expression in EMIU [85]. N9 and BV-2 microglia, RAW 264.7 macrophages, and DITNC1 astrocytes were all significantly inhibited by 100-μM D609 treatment without affecting cell viability [50]. D609 may perform an anti-inflammatory function by preventing the proliferation of macrophages/microglia, which are the principal sources of IL-1 and TNF and other pro-inflammatory cytokines. TNF-induced PAR formation is inhibited in D609-treated cultures, according to studies. D609 reduced microglial morphological change, NF-kB transcriptional activation, and PARP-1 activation [86].

4.4. Anti-Tumor and Anti-Proliferative

Ceramide and DAG, linked by the sphingomyelin (SM) synthase pathway, are two-second messengers that control cell proliferation and growth arrest in opposing ways [87]. In SM and DAG, SMS transports the phosphocholine group from the PC to the ceramide [88]. SMS comes in two varieties: SMS1 is located in the Golgi apparatus, while SMS2 is found in the plasma membrane [89,90]. D609 inhibits both forms of SMS. By preventing ceramide injection into SM, inhibiting SMS elevates ceramide levels. Thanks to SMS inhibition, D609 prevented the proliferation of ßFGF-stimulated astrocyte [16,91]. According to one study, D609 decreased non-neuronal cell-line proliferation without triggering cell death [67]. In BV-2 microglia, D609 therapy elevated the expression of ceramide and p21 levels [50,92]. D609 also hypophosphorylated Rb, causing cell-cycle inhibition in the G0/G1 step, and a decrease in proportion of cells in the S-Phase [50]. Ceramide′s significance in D609-induced cell-cycle arrests is supported by the exogenous C8-ceramide investigations [50].
D609 regulates ceramide formation and cell death caused by death receptors [93,94]. Nontoxic concentrations of D609 inhibit sphingomyelin synthase and glucosylceramide synthase in Jurkat cells [95]. FasL-induced caspase activation and apoptosis were significantly enhanced by D609 [96,97]. Since the authors claim that Bcl-xL overexpression caused D609 outcomes, mitochondrial events were likely involved. The authors conclude that D609 causes cell death in T lymphocyte cells by acting downstream of caspases 8. They believe that in Fasl-induced cell death, a rise occurs due to D609 inhibition of ceramide transfer to complex sphingolipids [98]. Anti-HER2 medicines, which block the tumorigenic actions of HER2, do not significantly affect the treatment of HER2-positive EOC (epithelial ovarian cancer) [99,100]. Preclinical trial models can be utilized to assess the molecular processes causing HER2 overexpression and oncogenicity, giving rise to new EOC treatments. The enhanced HER2 expression in breast cancer cells is regulated by PC-PLC [14]. Researchers found that the inhibition of PC-PLC may be a good target for the tumorigenic effects of increased HER2 expression in EOC41 cells [44].

4.5. Neuroprotective

Evidence suggests that oxidative stress has a role in the pathogenesis of Alzheimer′s disease [101]. Amyloid-peptide accumulation causes a cascade of oxidative neuron damage and eventually leads to neuronal death, one of the hallmarks of Alzheimer′s disease [102,103]. Amyloid-peptide is senile plaque’s major component. It causes damage to nucleic acids, proteins, and membrane lipids in neurons by generating free radicals [104,105]. As a result, interest is growing in antioxidant chemicals as they play a protective role in the treatment of AD and other disorders of oxidative stress [106,107]. Amid the various antioxidant medications, “thiol-delivering” compounds have received much attention. PC-PLC is inhibited by D609, a compound that mimics the action of glutathione. Recent studies have shown that it can inhibit phosphatidylcholine-specific phospholipase C [108,109]. Another study assesses the efficiency of D609 to protect synaptosomes in vivo from amyloid-peptide caused by oxidative stress [110,111]. After gerbils were fed D609 or saline solution, synaptosomes were extracted from their brains. The ex vivo treatment of gerbils with synaptosomal preparations derived from D609-injected gerbils with amyloid peptide (1–42) significantly decreased oxidative stress parameters, such as reactive oxygen species, lipid peroxidation (4-hydroxy-2-nonenal), protein oxidation (carbonyl and 3-nitrotyrosine protein), and levels [110]. These findings support the idea that amyloid-peptide-induced free-radical control may be a useful therapeutic tool for treating AD and other related oxidative stress diseases [112]. Based on the above evidence, D609 is a potent antioxidant that may help cure AD and other related oxidative stress diseases [108,110,113,114].
Tricyclodecan-9-yl-Xanthogenate has anti-proliferative, antioxidant, and anti-inflammatory properties. In mitochondria, cellular oxidation of the substrate releases energy in the ATP form [27,115]. This process involves the release of reactive oxygen species (ROS), causing damage to membrane phospholipids, DNA, and proteins, eventually leading to cell death and disease [116,117]. On the other hand, endogenous antioxidants scavenge ROS and prevent cell death [118,119]. D609 was discovered to be a powerful inhibitor of the PCPLC, which is essential for proliferation and cell survival [120,121]. D609 is an effective antioxidant and glutathione mimetic drug in previous studies.
Similarly, there is a substantial amount of literature explaining D609′s anti-proliferative effects in various cells. D609 has been shown in studies to inhibit the proliferation of neural progenitor cells [122,123]. Despite numerous studies, the mechanism of D609-induced cell-cycle arrest remains unclear. Furthermore, only a limited amount of knowledge is available on D609′s antioxidant impact on neural progenitor cells. Researchers could use D609′s neuroprotective and anti-proliferative properties to treat diseases such as stroke and cancer, which both need ATP for survival and proliferation if they could determine how it works on its potential targets. In studies on the function of the ATP content and cytochrome c oxidase of neural progenitor cells, D609′s antioxidant properties were also highlighted. In this research, an in vitro cell culture model of neural progenitor cells obtained from adult rat brains was used [110,124,125]. D609 possesses antioxidant properties, with its neuroprotective and antiproliferative properties encompassing a wide range of cells. D609 was previously found to reduce neural progenitor cell distribution. The antioxidant properties of D609 have been used to assess cell oxidation and calculate cells′ ATP content in the neural progenitor cells extracted from rat’s brains in the subventricular area. D609 reduces the neural progenitor cells’ ATP content by about 40%, which may inhibit cellular metabolic capability [124,126]. COX, also called ETC complex IV, is a terminal enzyme that causes the oxidation of substrates to generate energy for cellular activity [127]. Modulating COX activity can thus interfere with ATP production, influencing cell proliferation. After the incubation of D609 neural progenitor cells, cytochrome C oxidase activity was found to decrease, supporting this hypothesis [128,129]. According to these findings, D609 can inhibit cytochrome C oxidase activity and, as a result, ATP synthesis in neural progenitor cells [129].

4.6. Cholinergic Neuron Differentiation

A cholinergic neuron is a nerve cell that sends signals primarily via acetylcholine (ACh). Cholinergic neurons, which primarily use the neurotransmitter acetylcholine (Ach) for message transmission, play a crucial role in memory, locomotion, and behavioral response [130,131]. Cholinergic neuron loss causes a decline in choline acetyltransferase (ChAT) function, which causes motor nerve degeneration and cognitive dysfunction, as seen in AD [132,133]. A new treatment for cholinergic neuron loss is yet to be found, despite the use of cell-based therapies and nerve transplants to treat a variety of neurological disorders [134]. Several stem-cell-based therapies, including motor nerve disorders and Alzheimer′s disease, have recently been proposed as experimental therapies to alleviate the pathophysiology of cholinergic nerve disorders [134,135]. The tricyclodecane-9-yl-xanthogenate (D609) neuronal induction approach was used to successfully differentiate hDPSCs-cryo into cholinergic neurons (DF-chN) [136]. Motor nerve regeneration on an unprecedented scale was observed when DF-chN was inserted in vivo into experimental rats with sciatic nerve defects [136]. The simple inhibitor of phosphatidylcholine-specific C phospholipase, D609, has previously been shown to distinguish bone marrow MSCs (BMSCs) from neuron-like cells [137]. In BMSCs, D609 induces neuron-like cells with cholinergic neuronal properties; however, the mechanism underlying D609 neurogenic induction is unknown [138]. There is another belief that the treatment of D609 in BMSCs blocks PC-PLC activity, while elevated levels of HSP70 trigger the activation of transcription regulator B-cell translocation gene 2 (BTG2), resulting in cholinergic neuronal differentiation based on the number of responsive neuronal-specific genes [139].
D609 therapy has also been shown to reduce mesodermal and endodermal differentiation gene expression while increasing neuroprotection, neuronal differentiation, and cholesterol synthesis gene expression [136]. The formation and maintenance of the myelin sheath require cholesterol. In addition, differentiated cholinergic neuron relocation increases functional neuron regeneration and protection in animals with spinal cord injuries in vivo models. As a result, treating D609 with BMSCs is a quick and easy way to induce a cholinergic neuron induction [138]. According to Soomi Jang et al. (2018), treatment with D609 caused stem cells (derived from cryopreserved dental pulp) to appropriately transform into cholinergic neurons [138]. These cholinergic neurons, differentiated from dental pulp stem cells, had morphological properties similar to those of neurons, such as a neuron body and axonal fiber, as well as positive mRNA and protein expression of cholinergic neuronal markers [136].
Since bone marrow stromal cells (BMSCs) have a low degree of neuronal differentiation in vivo, raising the number of BMSC-derived neurons to treat neurological disorders is critical. According to Chunhui Sun et al., D609, an inhibitor of PC-PLC, stimulated BMSCs to differentiate between neuron-like cells in vitro. The neuronal form, on the other hand, was not exact. It is still uncertain whether these neuron-like cells restore neuronal dysfunction by exhibiting functional neuronal physiological activities. Chunhui Sun et al. assessed their properties by noting the neurotransmitters involved in neuron function and calcium to answer these questions. In vitro, both cells had to function cholinergic neurons, according to the findings. The regeneration of mice with injured spinal cords was aided by the transplantation of such cholinergic neuron-like cells, which were more effective than BMSCs. When cholinergic neuron-like cells derived from BMSC were injected, the proportion of cholinergic neurons increased, indicating a high degree of in vivo differentiation. The cholinergic neuron percentage in host cells rose, acetylcholine synthesis increased, and neurofilament preservation was seen in the lesions of animals injected with BMSC-derived neurons, implying neuronal protection. The results of the study provide a clear method for converting BMSCs into cholinergic neuron-like cells, as well as a potential treatment plan for spinal injuries [138]. In spinal-cord-injured mice, D609 allows BMSCs to differentiate into cholinergic neuron-like cells, and their distribution promotes the functional regeneration and defense of the neurons. This study revealed a new approach for BMSCs to obtain cholinergic neuron-like cells, as well as novel treatment options for neurological disorders. An overview of the pharmacological properties of D609 is presented in Table 1.

5. Conclusions

This review focuses on the multiple and diverse pharmacological activities of D609, including its inhibition of cancer proliferation, inflammatory cascade, oxidation stress, and neuroprotective effects. It seems that D609 regulates multiple pathways by targeting SMS/PC-PLC. It promotes the de novo synthesis of ceramide by stimulating SPT and has mimetic antioxidant/glutathione effects due to the presence of the thiol feature. Moreover, D609 attenuates oxidized PC (OxPC) and ROS development via its glutathione/antioxidant mimetic properties, thereby ameliorating CNS pathologies. Recently, it has been shown that D609 defends primary neuronal cultures against amyloid β-peptide-induced neurotoxicity and oxidative stress in vitro/in vivo synaptosomes.
Furthermore, researchers believe that D609 treatment increases HSP70, which stimulates the transcription regulator B-cell translocation gene 2 (BTG2), triggering cholinergic neuronal differentiation by increasing the level of receptive neuronal-specific genes. By suppressing the proliferation of microglia/macrophages, D609 can reduce free radical formation, especially ROS and oxidized PC (OxPC). This also inhibits the replication of herpes simplex virus type 1 without causing cytotoxicity, and has a diminishing effect on nitrite and peroxide expression, while it increases superoxide dismutase and lowers iNOS expressions. D609 also activates NF-kB transcription, blocks the stimulation of PARP-1, and causes microglial morphological transformation, significantly improving the caspase activation and apoptosis triggered by FasL.
Hence, through various pharmacological activities, this compound can be used as a multitarget approach to combat various diseases. Further studies are needed to determine the exact preventive mechanism of action of D609 in various diseases; this review will help academics to conduct more detailed research on this topic.

Author Contributions

Conceptualization, A.K.; methodology, A.H.B., K.B.D. and S.A.; software, M.M.G.; validation, F.S., S.M.A., and M.M.G.; formal analysis, P.A.; investigation, F.S., and P.A.; resources, S.M.A.; data curation, P.A.; writing—original draft preparation, A.H.B., K.B.D.; writing—review and editing, A.K., F.S., S.M.A., and P.A.; visualization, M.M.G.; supervision, A.K.; project administration, A.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors are thankful to Jazan University and AlMaarefa University for encouraging authors to write this review.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Shugar, D.J.P. Viral and host-cell protein kinases: Enticing antiviral targets and relevance of nucleoside, and viral thymidine, kinases. Pharmacol. Ther. 1999, 82, 315–335. [Google Scholar] [CrossRef]
  2. Eurtivong, C.; Pilkington, L.I.; van Rensburg, M.; White, R.M.; Bar, H.K.; Rees, S.; Paulin, E.K.; Xu, C.S.; Sharma, N.; Leung, I.K.H.; et al. Discovery of novel phosphatidylcholine-specific phospholipase C drug-like inhibitors as potential anticancer agents. Eur. J. Med. Chem. 2020, 187, 111919. [Google Scholar] [CrossRef] [PubMed]
  3. Podo, F.; Paris, L.; Cecchetti, S.; Spadaro, F.; Abalsamo, L.; Ramoni, C.; Ricci, A.; Pisanu, M.E.; Sardanelli, F.; Canese, R.; et al. Activation of phosphatidylcholine-specific phospholipase C in breast and ovarian cancer: Impact on MRS-detected choline metabolic profile and perspectives for targeted therapy. Front. Oncol. 2016, 6, 171. [Google Scholar] [CrossRef] [PubMed]
  4. Milhas, D.; Andrieu-Abadie, N.; Levade, T.; Benoist, H.; Ségui, B. The tricyclodecan-9-yl-xanthogenate D609 triggers ceramide increase and enhances FasL-induced caspase-dependent and-independent cell death in T lymphocytes. Int. J. Mol. Sci. 2012, 13, 8834–8852. [Google Scholar] [CrossRef]
  5. Amtmann, E.; Sauer, G. Tumor necrosis factor induces necrosis of human carcinoma xenografts in the presence of tricyclodecan-9-yl-xanthogenate and lauric acid. Int. J. Cancer 1990, 45, 1113–1118. [Google Scholar] [CrossRef] [PubMed]
  6. Adibhatla, R.M.; Hatcher, J.F.; Gusain, A. Tricyclodecan-9-yl-xanthogenate (D609) mechanism of actions: A mini-review of literature. Neurochem. Res. 2012, 37, 671–679. [Google Scholar] [CrossRef] [Green Version]
  7. Paris, L.; Cecchetti, S.; Spadaro, F.; Abalsamo, L.; Lugini, L.; Pisanu, M.E.; Iorio, E.; Natali, P.G.; Ramoni, C.; Podo, F. Inhibition of phosphatidylcholine-specific phospholipase C downregulates HER2 overexpression on plasma membrane of breast cancer cells. Breast Cancer Res. 2010, 12, R27. [Google Scholar] [CrossRef] [Green Version]
  8. Lu, M.-H.; Ji, W.-L.; Xu, D.-E.; Yao, P.-P.; Zhao, X.-Y.; Wang, Z.-T.; Fang, L.-P.; Huang, R.; Lan, L.-L.; Chen, J.-B.; et al. Inhibition of sphingomyelin synthase 1 ameliorates alzheimer-like pathology in APP/PS1 transgenic mice through promoting lysosomal degradation of BACE1. Exp. Neurol. 2019, 311, 67–79. [Google Scholar] [CrossRef]
  9. Adibhatla, R.M.; Hatcher, J.F. Lipid oxidation and peroxidation in CNS health and disease: From molecular mechanisms to therapeutic opportunities. Antioxid. Redox Signal 2010, 12, 125–169. [Google Scholar] [CrossRef]
  10. Vaughan, E.M.; You, J.-S.; Elsie Yu, H.-Y.E.; Lasek, A.; Vitale, N.; Hornberger, T.A.; Bement, W.M. Lipid domain–dependent regulation of single-cell wound repair. Mol. Biol. Cell 2014, 25, 1867–1876. [Google Scholar] [CrossRef] [Green Version]
  11. Kato, M.; Hammam, M.A.; Taniguchi, T.; Suga, Y.; Monde, K. What is the true structure of D609, a widely used lipid related enzyme inhibitor? Org. Lett. 2016, 18, 768–771. [Google Scholar] [CrossRef] [PubMed]
  12. Pilkington, L.I.; Sparrow, K.; Rees, S.W.P.; Paulin, E.K.; van Rensburg, M.; Xu, C.S.; Langley, R.J.; Leung, I.K.H.; Reynisson, J.; Leung, E.; et al. Development, synthesis and biological investigation of a novel class of potent PC-PLC inhibitors. Eur. J. Med. Chem. 2020, 191, 112162. [Google Scholar] [CrossRef] [PubMed]
  13. Schütze, S.; Potthoff, K.; Machleidt, T.; Berkovic, D.; Wiegmann, K.; Krönke, M. TNF activates NF-κB by phosphatidylcholine-specific phospholipase C-induced “acidic” sphingomyelin breakdown. Cell 1992, 71, 765–776. [Google Scholar] [CrossRef]
  14. Abalsamo, L.; Spadaro, F.; Bozzuto, G.; Paris, L.; Cecchetti, S.; Lugini, L.; Iorio, E.; Molinari, A.; Ramoni, C.; Podo, F. Inhibition of phosphatidylcholine-specific phospholipase C results in loss of mesenchymal traits in metastatic breast cancer cells. Breast Cancer Res. 2012, 14, R50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Ballou, L.R.; Laulederkind, S.J.; Rosloniec, E.F.; Raghow, R. Ceramide signalling and the immune response. Biochim. Biophys. Acta 1996, 1301, 273–287. [Google Scholar] [CrossRef]
  16. Adibhatla, R.M.; Hatcher, J.F. Protection by D609 through cell-cycle regulation after stroke. Mol. Neurobiol. 2010, 41, 206–217. [Google Scholar] [CrossRef]
  17. Zhou, D.; Lauderback, C.M.; Yu, T.; Brown, S.A.; Butterfield, D.A.; Thompson, J.S. D609 inhibits ionizing radiation-induced oxidative damage by acting as a potent antioxidant. J. Pharmacol. Exp. Ther. 2001, 298, 103–109. [Google Scholar]
  18. Ansari, M.A.; Joshi, G.; Huang, Q.; Opii, W.O.; Abdul, H.M.; Sultana, R.; Butterfield, D.A. In vivo administration of D609 leads to protection of subsequently isolated gerbil brain mitochondria subjected to in vitro oxidative stress induced by amyloid beta-peptide and other oxidative stressors: Relevance to Alzheimer’s disease and other oxidative stress-related neurodegenerative disorders. Free Radic. Biol. Med. 2006, 41, 1694–1703. [Google Scholar]
  19. Wang, B.; Wang, L.; Gu, S.; Yu, Y.; Huang, H.; Mo, K.; Xu, H.; Zeng, F.; Xiao, Y.; Peng, L.; et al. D609 protects retinal pigmented epithelium as a potential therapy for age-related macular degeneration. Signal Transduct. Target. Ther. 2020, 5, 20. [Google Scholar] [CrossRef] [Green Version]
  20. González-Bulnes, P.; González-Roura, A.; Canals, D.; Delgado, A.; Casas, J.; Llebaria, A. 2-Aminohydroxamic acid derivatives as inhibitors of Bacillus cereus phosphatidylcholine preferred phospholipase C PC-PLC(Bc). Bioorg. Med. Chem. 2010, 18, 8549–8555. [Google Scholar] [CrossRef]
  21. Yu, Z.F.; Nikolova-Karakashian, M.; Zhou, D.; Cheng, G.; Schuchman, E.H.; Mattson, M.P. Pivotal role for acidic sphingomyelinase in cerebral ischemia-induced ceramide and cytokine production, and neuronal apoptosis. J. Mol. Neurosci. 2000, 15, 85–97. [Google Scholar] [CrossRef]
  22. Koishi, R.; Yoshimura, C.; Kohama, T.; Serizawa, N. Leustroducsin B activates nuclear factor-κ B via the acidic sphingomyelinase pathway in human bone marrow-derived stromal cell line KM-102. J. Interferon Cytokine Res. 2002, 22, 343–350. [Google Scholar] [CrossRef] [PubMed]
  23. Hannun, Y.A.; Obeid, L.M.; Dbaibo, G.S. Ceramide: A novel second messenger and lipid mediator. Lipid Second. Messengers 1996, 8, 177–204. [Google Scholar]
  24. Miao, J.; Zang, X.; Cui, X.; Zhang, J. Autophagy, hyperlipidemia, and atherosclerosis. Adv. Exp. Med. Biol. 2020, 1207, 237–264. [Google Scholar] [PubMed]
  25. Hopewell, S. Effects of Phosphatidylinositol on ApoA-I Metabolism: Implications in HDL Metabolism. Master’s Thesis, University of Ottawa, Ottawa, ON, Canada, 2008. [Google Scholar]
  26. Sultana, R.; Newman, S.; Mohmmad-Abdul, H.; Keller, J.N.; Butterfield, D.A. Protective effect of the xanthate, D609, on Alzheimer’s amyloid β-peptide (1–42)-induced oxidative stress in primary neuronal cells. Free Radic. Res. 2004, 38, 449–458. [Google Scholar] [CrossRef]
  27. Cai, Z.; Zhao, B.; Ratka, A. Oxidative stress and β-amyloid protein in Alzheimer’s disease. Neuromol. Med. 2011, 13, 223–250. [Google Scholar] [CrossRef]
  28. Bai, A.; Meier, G.P.; Wang, Y.; Luberto, C.; Hannun, Y.A.; Zhou, D. Prodrug modification increases potassium tricyclo [5.2. 1.02, 6]-decan-8-yl dithiocarbonate (D609) chemical stability and cytotoxicity against U937 leukemia cells. J. Pharmacol. Exp. Ther. 2004, 309, 1051–1059. [Google Scholar] [CrossRef] [Green Version]
  29. Nofer, J.-R.; Junker, R.; Seedorf, U.; Assmann, G.; Zidek, W.; Tepel, M. D609–phosphatidylcholine-specific phospholipase C inhibitor attenuates thapsigargin-induced sodium influx in human lymphocytes. Cell Signal 2000, 12, 289–296. [Google Scholar] [CrossRef]
  30. Pomerantsev, A.P.; Kalnin, K.V.; Osorio, M.; Leppla, S.H. Phosphatidylcholine-specific phospholipase C and sphingomyelinase activities in bacteria of the Bacillus cereus group. Infect. Immun. 2003, 71, 6591–6606. [Google Scholar] [CrossRef] [Green Version]
  31. Malavasi, F.; Deaglio, S.; Funaro, A.; Ferrero, E.; Horenstein, A.L.; Ortolan, E.; Vaisitti, T.; Aydin, S. Evolution and function of the ADP ribosyl cyclase/CD38 gene family in physiology and pathology. Physiol. Rev. 2008, 88, 841–886. [Google Scholar] [CrossRef] [Green Version]
  32. Mercurio, L.; Cecchetti, S.; Ricci, A.; Pacella, A.; Cigliana, G.; Bozzuto, G.; Podo, F.; Iorio, E.; Carpinelli, G. Phosphatidylcholine-specific phospholipase C inhibition down-regulates CXCR4 expression and interferes with proliferation, invasion and glycolysis in glioma cells. PLoS ONE 2017, 12, e0176108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Vasil, M.L. Pseudomonas aeruginosa phospholipases and phospholipids. In Pseudomonas; Springer: Berlin, Germany, 2006; pp. 69–97. [Google Scholar]
  34. Su, W.; Chen, Q.; Frohman, M.A. Targeting phospholipase D with small-molecule inhibitors as a potential therapeutic approach for cancer metastasis. Future Oncol. 2009, 5, 1477–1486. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Iorio, E.; Caramujo, M.J.; Cecchetti, S.; Spadaro, F.; Carpinelli, G.; Canese, R.; Podo, F. Key players in choline metabolic reprograming in triple-negative breast cancer. Front. Oncol. 2016, 6, 205. [Google Scholar] [CrossRef] [Green Version]
  36. He, X.; Chen, F.; McGovern, M.M.; Schuchman, E.H. A fluorescence-based, high-throughput sphingomyelin assay for the analysis of Niemann–Pick disease and other disorders of sphingomyelin metabolism. Anal. Biochem. 2002, 306, 115–123. [Google Scholar] [CrossRef] [PubMed]
  37. Diaz-Meco, M.; Dominguez, I.; Sanz, L.; Municio, M.M.; Berra, E.; Cornet, M.E.; de Herreros, A.G.; Johansen, T.; Moscat, J. Phospholipase C-mediated hydrolysis of phosphatidylcholine is a target of transforming growth factor beta 1 inhibitory signals. Mol. Cell Biol. 1992, 12, 302–308. [Google Scholar] [PubMed] [Green Version]
  38. Abelson, J.N.; Simon, M.I.; Merrill Jr, A.H.; Hannun, Y.A. Sphingolipid Metabolism and Cell Signaling, Part A; Elsevier: Amsterdam, The Netherlands, 1999. [Google Scholar]
  39. Zheng, W.; Kollmeyer, J.; Symolon, H.; Momin, A.; Munter, E.; Wang, E.; Kelly, S.; Allegood, J.C.; Liu, Y.; Peng, Q.; et al. Ceramides and other bioactive sphingolipid backbones in health and disease: Lipidomic analysis, metabolism and roles in membrane structure, dynamics, signaling and autophagy. Biochim. Biophys. Acta 2006, 1758, 1864–1884. [Google Scholar] [CrossRef] [Green Version]
  40. Ji, R.; Akashi, H.; Drosatos, K.; Liao, X.; Jiang, H.; Kennel, P.J.; Brunjes, D.L.; Castillero, E.; Zhang, X.; Deng, L.Y.; et al. Increased de novo ceramide synthesis and accumulation in failing myocardium. JCI Insight 2017, 2, e82922. [Google Scholar] [CrossRef] [Green Version]
  41. Li, Z.; Hailemariam, T.K.; Zhou, H.; Li, Y.; Duckworth, D.C.; Peake, D.A.; Zhang, Y.; Kuo, M.-S.; Cao, G.; Jiang, X.-C. Inhibition of sphingomyelin synthase (SMS) affects intracellular sphingomyelin accumulation and plasma membrane lipid organization. Biochim. Biophys. Acta 2007, 1771, 1186–1194. [Google Scholar] [CrossRef] [Green Version]
  42. Paris, L.; Podo, F.; Spadaro, F.; Abalsamo, L.; Pisanu, M.E.; Ricci, A.; Cecchetti, S.; Altabella, L.; Bouncervello, M.; Lozneanu, L.; et al. Phosphatidylcholine-specific phospholipase C inhibition reduces HER2-overexpression, cell proliferation and in vivo tumor growth in a highly tumorigenic ovarian cancer model. Oncotarget 2017, 8, 55022–55038. [Google Scholar] [CrossRef] [Green Version]
  43. Lou, B.; Dong, J.; Li, Y.; Ding, T.; Bi, T.; Li, Y.; Deng, X.; Ye, D.; Jiang, X.-C. Pharmacologic inhibition of sphingomyelin synthase (SMS) activity reduces apolipoprotein-B secretion from hepatocytes and attenuates endotoxin-mediated macrophage inflammation. PLoS ONE 2014, 9, e102641. [Google Scholar] [CrossRef] [Green Version]
  44. Meng, A.; Luberto, C.; Meier, P.; Bai, A.; Yang, X.; Hannun, Y.A.; Zhou, D. Sphingomyelin synthase as a potential target for D609-induced apoptosis in U937 human monocytic leukemia cells. Exp. Cell Res. 2004, 292, 385–392. [Google Scholar] [CrossRef] [PubMed]
  45. Pucci, B.; Kasten, M.; Giordano, A. Cell cycle and apoptosis. Neoplasia 2000, 2, 291–299. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Linke, S.P.; Clarkin, K.C.; Di Leonardo, A.; Tsou, A.; Wahl, G.M. A reversible, p53-dependent G0/G1 cell cycle arrest induced by ribonucleotide depletion in the absence of detectable DNA damage. Genes. Dev. 1996, 10, 934–947. [Google Scholar] [CrossRef] [Green Version]
  47. Trayssac, M.; Clarke, C.J.; Stith, J.L.; Snider, J.M.; Newen, N.; Gault, C.R.; Hannun, Y.A.; Obeid, L.M. Targeting sphingosine kinase 1 (SK1) enhances oncogene-induced senescence through ceramide synthase 2 (CerS2)-mediated generation of very-long-chain ceramides. Cell Death Dis. 2021, 12, 27. [Google Scholar] [CrossRef] [PubMed]
  48. Ye, L.; Yang, Y.; Zhang, X.; Cai, P.; Li, R.; Chen, D.; Wei, X.; Zhang, X.; Xu, H.; Xiao, J.; et al. The role of bFGF in the excessive activation of astrocytes is related to the inhibition of TLR4/NFκB signals. Int. J. Mol. Sci. 2016, 17, 37. [Google Scholar] [CrossRef] [PubMed]
  49. Gusain, A.; Hatcher, J.F.; Adibhatla, R.M.; Wesley, U.V.; Dempsey, R.J. Anti-proliferative effects of tricyclodecan-9-yl-xanthogenate (D609) involve ceramide and cell cycle inhibition. Mol. Neurobiol. 2012, 45, 455–464. [Google Scholar] [CrossRef] [Green Version]
  50. D’Angelo, G.; Moorthi, S.; Luberto, C. Role and function of sphingomyelin biosynthesis in the development of cancer. Adv. Cancer Res. 2018, 140, 61–96. [Google Scholar]
  51. Davis, C.K.; Jain, S.A.; Bae, O.-N.; Majid, A.; Rajanikant, G.K. Hypoxia mimetic agents for ischemic stroke. Front. Cell Dev. Biol. 2019, 6, 175. [Google Scholar]
  52. Lee, D.H.; Kim, G.W.; Yoo, J.; Lee, S.W.; Jeon, Y.H.; Kim, S.Y.; Kang, H.G.; Kim, D.-H.; Chun, K.-H.; Choi, J.; et al. Histone demethylase KDM4C controls tumorigenesis of glioblastoma by epigenetically regulating p53 and c-Myc. Cell Death Dis. 2021, 12, 89. [Google Scholar] [CrossRef]
  53. Hatzmann, F.M.; Ejaz, A.; Wiegers, G.J.; Mandl, M.; Brucker, C.; Lechner, S.; Rauchenwald, T.; Zweirzina, M.; Baumgarten, S.; Wagner, S.; et al. Quiescence, stemness and adipogenic differentiation capacity in human DLK1/CD34+/CD24+ adipose stem/progenitor cells. Cells 2021, 10, 214. [Google Scholar] [CrossRef]
  54. Alesse, E.; Zazzeroni, F.; Angelucci, A.; Giannini, G.; Marcotullio, L.D.; Gulino, A. The growth arrest and downregulation of c-myc transcription induced by ceramide are related events dependent on p21 induction, Rb underphosphorylation and E2F sequestering. Cell Death Differ. 1998, 5, 381–389. [Google Scholar] [CrossRef] [PubMed]
  55. Patwardhan, G.A.; Liu, Y.-Y. Sphingolipids and expression regulation of genes in cancer. Prog. Lipid Res. 2011, 50, 104–114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Chauhan, V.; Chauhan, A. Oxidative stress in Alzheimer’s disease. Pathophysiology 2006, 13, 195–208. [Google Scholar] [CrossRef] [PubMed]
  57. Zorov, D.B.; Juhaszova, M.; Sollott, S.J. Mitochondrial reactive oxygen species (ROS) and ROS-induced ROS release. Physiol. Rev. 2014, 94, 909–950. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Zhu, X.; Smith, M.A.; Perry, G.; Aliev, G. Mitochondrial failures in Alzheimer’s disease. Am. J. Alzheimers Dis. Other Demen. 2004, 19, 345–352. [Google Scholar] [CrossRef]
  59. Aliev, G.; Li, Y.; Palacios, H.H.; Obrenovich, M.E. Oxidative stress induced mitochondrial DNA deletion as a hallmark for the drug development in the context of the cerebrovascular diseases. Recent Pat. Cardiovasc. Drug Discov. 2011, 6, 222–241. [Google Scholar] [CrossRef]
  60. Wang, C.; Youle, R.J. The role of mitochondria in apoptosis. Annu. Rev. Genet. 2009, 43, 95–118. [Google Scholar] [CrossRef] [Green Version]
  61. Dai, S.-H.; Chen, T.; Wang, Y.-H.; Zhu, J.; Luo, P.; Rao, W.; Yang, Y.-F.; Fei, Z.; Xiang, X.-F. Sirt3 protects cortical neurons against oxidative stress via regulating mitochondrial Ca2+ and mitochondrial biogenesis. Int. J. Mol. Sci. 2014, 15, 14591–14609. [Google Scholar] [CrossRef] [Green Version]
  62. Zhao, Y.; Zhao, B. Oxidative stress and the pathogenesis of Alzheimer’s disease. Neurodeg. Neurogen. Oxid. Stress 2013, 2013, 316523. [Google Scholar] [CrossRef] [Green Version]
  63. Santos, C.X.C.; Tanaka, L.Y.; Wosniak, J.; Laurindo, F.R.M. Mechanisms and implications of reactive oxygen species generation during the unfolded protein response: Roles of endoplasmic reticulum oxidoreductases, mitochondrial electron transport, and NADPH oxidase. Antioxid. Redox Signal 2009, 11, 2409–2427. [Google Scholar] [CrossRef]
  64. Stadtman, E.R.; Berlett, B.S. Reactive oxygen-mediated protein oxidation in aging and disease. Drug Metab. Rev. 1997, 10, 485–494. [Google Scholar] [CrossRef] [PubMed]
  65. Cardoso, S.M.; Santana, I.; Swerdlow, R.H.; Oliveira, C.R. Mitochondria dysfunction of Alzheimer’s disease cybrids enhances Aβ toxicity. J. Neurochem. 2004, 89, 1417–1426. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Amtmann, E. The antiviral, antitumoural xanthate D609 is a competitive inhibitor of phosphatidylcholine-specific phospholipase C. Drugs Exp. Clin. Res. 1996, 22, 287–294. [Google Scholar] [PubMed]
  67. Turban, S.; Hajduch, E. Protein kinase C isoforms: Mediators of reactive lipid metabolites in the development of insulin resistance. FEBS Lett. 2011, 585, 269–274. [Google Scholar] [CrossRef] [Green Version]
  68. Cioffi, F.; Adam, R.H.I.; Broersen, K. Molecular mechanisms and genetics of oxidative stress in Alzheimer’s disease. J. Alzeimers Dis. 2019, 72, 981–1017. [Google Scholar] [CrossRef] [Green Version]
  69. Aquaro, S.; Scopelliti, F.; Pollicita, M.; Perno, C.F. Oxidative stress and HIV infection: Target pathways for novel therapies? Fut. HIV Ther. 2008, 2, 327–338. [Google Scholar] [CrossRef]
  70. Mansor, N.I.; Ntimi, C.M.; Abdul-Aziz, N.M.; Ling, K.-H.; Adam, A.; Rosli, R.; Hassan, Z.; Nordin, N. Asymptomatic neurotoxicity of amyloid β-peptides (Aβ1-42 and Aβ25-35) on mouse embryonic stem cell-derived neural cells. Bosn. J. Basic Med. Sci. 2021, 21, 98–110. [Google Scholar] [CrossRef] [Green Version]
  71. Yang, H.; Xie, Z.; Wei, L.; Ding, M.; Wang, P.; Bi, J.Z. Glutathione-mimetic D609 alleviates memory deficits and reduces amyloid-β deposition in an AβPP/PS1 transgenic mouse model. Neuroreport 2018, 29, 833–838. [Google Scholar] [CrossRef]
  72. Sauer, G.; Amtmann, E.; Melber, K.; Knapp, A.; Muller, K.; Hummel, K.; Scherm, A. DNA and RNA virus species are inhibited by xanthates, a class of antiviral compounds with unique properties. Proc. Natl. Acad. Sci. USA 1984, 81, 3263–3267. [Google Scholar] [CrossRef] [Green Version]
  73. Mellert, W.; Amtmanne, E.; Erfle, V.; Sauer, G. Inhibition of HIV-1 replication by an antiviral xanthate compound in vitro. AIDS Res. Hum. Retroviruses 1988, 4, 71–81. [Google Scholar] [CrossRef]
  74. Villanueva, N.; Navarro, J.; Cubero, E. Antiviral effects of xanthate D609 on the human respiratory syncytial virus growth cycle. Virology 1991, 181, 101–108. [Google Scholar] [CrossRef]
  75. Walro, D.G.; Rosenthal, K.S. The antiviral xanthate compound D609 inhibits herpes simplex virus type 1 replication and protein phosphorylation. Antiviral Res. 1997, 36, 63–72. [Google Scholar] [CrossRef]
  76. Bali, P.; Lahiri, D.K.; Banik, A.; Nehru, B.; Anand, A. Potential for stem cells therapy in Alzheimer’s disease: Do neurotrophic factors play critical role? Curr. Alzeimer Res. 2017, 14, 208–220. [Google Scholar] [CrossRef] [PubMed]
  77. Rashidi, S.; Fernandez-Rubio, C.; Manzano-Román, R.; Mansouri, R.; Shafiei, R.; Ali-Hassanzadeh, M.; Barazesh, A.; Karimazar, M.; Hatam, G.; Nguewa, P. Potential therapeutic targets shared between leishmaniasis and cancer. Parasitology 2021, 148, 655–671. [Google Scholar] [CrossRef] [PubMed]
  78. Smith, J.R.; Rosenbaum, J.T.; Williams, K.A. Experimental melanin-induced uveitis: Experimental model of human acute anterior uveitis. Ophthalmic Res. 2008, 40, 136–140. [Google Scholar] [CrossRef]
  79. Matteson, D.M.; Shen, D.F.; Chan, C.C. Inhibition of experimental melanin protein-induced uveitis (EMIU) by targeting nitric oxide via phosphatidylcholine-specific phospholipase C. J. Autoimmun. 1999, 13, 197–204. [Google Scholar] [CrossRef]
  80. Hooper, D.C.; Bagasra, O.; Marini, J.C.; Zborek, A.; Ohnishi, S.T.; Kean, R.; Champion, J.M.; Sarker, A.B.; Bobroski, L.; Farber, J.L.; et al. Prevention of experimental allergic encephalomyelitis by targeting nitric oxide and peroxynitrite: Implications for the treatment of multiple sclerosis. Proc. Natl. Acad. Sci. USA 1997, 94, 2528–2533. [Google Scholar] [CrossRef] [Green Version]
  81. Willenborg, D.O.; Staykova, M.A.; Cowden, W.B. Our shifting understanding of the role of nitric oxide in autoimmune encephalomyelitis: A review. J. Neuroimmunol. 1999, 100, 21–35. [Google Scholar] [CrossRef]
  82. Liu, H.; Zhang, H.; Forman, H.J. Silica induces macrophage cytokines through phosphatidylcholine-specific phospholipase C with hydrogen peroxide. Am. Respir. Cell Mol. Biol. 2007, 36, 594–599. [Google Scholar] [CrossRef] [Green Version]
  83. Goebeler, M.; Gillitzer, R.; Kilian, K.; Utzel, K.; Brocker, E.B.; Rapp, U.R.; Ludwig, S. Multiple signaling pathways regulate NF-κB–dependent transcription of the monocyte chemoattractant protein-1 gene in primary endothelial cells. Blood 2001, 97, 46–55. [Google Scholar] [CrossRef] [Green Version]
  84. Daun, J.M.; Fenton, M.J. Interleukin-1/Toll receptor family members: Receptor structure and signal transduction pathways. J. Interferon Cytokine Res. 2000, 20, 843–855. [Google Scholar] [CrossRef] [PubMed]
  85. Vuong, B.; Hogan-Cann, A.D.J.; Alano, C.C.; Stevenson, M.; Chan, W.Y.; Anderson, C.M.; Swanson, R.A.; Kauppinen, T.M. NF-κB transcriptional activation by TNFα requires phospholipase C, extracellular signal-regulated kinase 2 and poly (ADP-ribose) polymerase-1. J. Neuroinflammation 2015, 12, E229. [Google Scholar] [CrossRef] [Green Version]
  86. Flores, I.; Jones, D.R.; Mérida, I. Changes in the balance between mitogenic and antimitogenic lipid second messengers during proliferation, cell arrest, and apoptosis in T-lymphocytes. FASEB J. 2000, 14, 1873–1875. [Google Scholar] [CrossRef] [Green Version]
  87. Yeang, C.; Varshney, S.; Wang, R.; Zhang, Y.; Ye, D.; Jiang, X.-C. The domain responsible for sphingomyelin synthase (SMS) activity. Biochim. Biophys. Acta 2008, 1781, 610–617. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Villani, M.; Subathra, M.; Im, Y.-B.; Choi, Y.; Signorelli, P.; Poeta, M.D.; Lubreto, C. Sphingomyelin synthases regulate production of diacylglycerol at the Golgi. Biochem. J. 2008, 414, 31–41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Fernández-García, P.; Rosselló, C.A.; Rodríguez-Lorca, R.; Beteta-Gobel, R.; Fernandez-Diaz, J.; Llado, V.; Busquets, X.; Escriba, P.V. The opposing contribution of SMS1 and SMS2 to glioma progression and their value in the therapeutic response to 2OHOA. Cancers 2019, 11, 88. [Google Scholar] [CrossRef] [Green Version]
  90. Chen, Y.Q.; Yurek, D.A.; Yu, L.; Wang, H.; Ehsani, M.E.; Qian, Y.-W.; Konrad, R.J.; Jiang, X.-C.; Kuo, M.-S.; Cao, G.; et al. Development of a quantitative biochemical and cellular sphingomyelin synthase assay using mass spectrometry. Anal. Biocehm. 2013, 438, 61–66. [Google Scholar] [CrossRef]
  91. Moorthi, S. Role and Regulation of Sphingomyelin Synthase 1 in Leukemia. Ph.D. Thesis, Stony Brook University, Stony Brook, NY, USA, 2017. [Google Scholar]
  92. Ouasti, S.; Matarrese, P.; Paddon, R.; Khosravi-Far, R.; Sorice, M.; Tinari, A.; Malorni, W.; Esposti, M.D. Death receptor ligation triggers membrane scrambling between Golgi and mitochondria. Cell Death Differ. 2007, 14, 453–461. [Google Scholar] [CrossRef] [Green Version]
  93. Morad, S.A.F.; Cabot, M.C. Ceramide-orchestrated signalling in cancer cells. Nat. Rev. Cancer 2013, 13, 51–65. [Google Scholar] [CrossRef]
  94. Radin, N.S. Killing tumours by ceramide-induced apoptosis: A critique of available drugs. Biochem. J. 2003, 371, 243–256. [Google Scholar] [CrossRef]
  95. Moulin, M.; Carpentier, S.; Levade, T.; Arrigo, A.-P. Potential roles of membrane fluidity and ceramide in hyperthermia and alcohol stimulation of TRAIL apoptosis. Apoptosis 2007, 12, 1703–1720. [Google Scholar] [CrossRef] [PubMed]
  96. Argaud, L.; Prigent, A.-F.; Chalabreysse, L.; Loufouat, J.; Lagrade, M.; Ovize, M. Ceramide in the antiapoptotic effect of ischemic preconditioning. Am. J. Physiol. Heart Circ. Physiol. 2004, 286, H246–H251. [Google Scholar] [CrossRef] [PubMed]
  97. Thon, L.; Möhlig, H.; Mathieu, S.; Lange, A.; Bulanova, E.; Winoto-Morbach, S.; Schutze, S.; Bulfone-Paus, S.; Adam, D. Ceramide mediates caspase-independent programmed cell death. FASEB J. 2005, 19, 1945–1956. [Google Scholar] [CrossRef] [PubMed]
  98. English, D.P.; Roque, D.M.; Santin, A.D. HER2 expression beyond breast cancer: Therapeutic implications for gynecologic malignancies. Mol. Diagn. Ther. 2013, 17, 85–99. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Coward, J.I.; Middleton, K.; Murphy, F. New perspectives on targeted therapy in ovarian cancer. Int. J. Womens Health 2015, 7, 189–203. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Misrani, A.; Tabassum, S.; Yang, L. Mitochondrial dysfunction and oxidative stress in Alzheimer’s disease. Front. Aging Neurosci. 2021, 13, 617588. [Google Scholar] [CrossRef]
  101. Uddin, M.S.; Tewari, D.; Sharma, G.; Kabir, M.T.; Barreto, G.E.; Bin-Jumah, M.N.; Perveen, A.; Abdel-Daim, M.M.; Ashraf, G.M. Molecular mechanisms of ER stress and UPR in the pathogenesis of Alzheimer’s disease. Mol. Neurobiol. 2020, 57, 2902–2919. [Google Scholar] [CrossRef]
  102. Guo, J.; Yang, G.; He, Y.; Xu, H.; Fan, H.; An, J.; Zhang, L.; Zhang, R.; Cao, G.; Hao, D.; et al. Involvement of α7nAChR in the protective effects of genistein against β-amyloid-induced oxidative stress in neurons via a PI3K/Akt/Nrf2 pathway-related mechanism. Cell Mol. Neurobiol. 2021, 41, 377–393. [Google Scholar] [CrossRef]
  103. Picone, P.; Nuzzo, D.; Giacomazza, D.; Di Carlo, M. β-Amyloid peptide: The cell compartment multi-faceted interaction in Alzheimer’s disease. Neurotox. Res. 2020, 37, 250–263. [Google Scholar] [CrossRef]
  104. Leal, M.; Catarino, R.; Pimenta, A.; Souto, M.R.S. Roles of metal microelements in neurodegenerative diseases. Neurophysiology 2020, 52, 80–88. [Google Scholar] [CrossRef]
  105. Salehi, B.; Azzini, E.; Zucca, P.; Varoni, E.M.; Kumra, N.V.A.; Dini, L.; Panzarini, E.; Rajkovic, J.; Fokou, P.V.T.; Peluso, I.; et al. Plant-derived bioactives and oxidative stress-related disorders: A key trend towards healthy aging and longevity promotion. Appl. Sci. 2020, 10, 947. [Google Scholar] [CrossRef] [Green Version]
  106. Simioni, C.; Zauli, G.; Martelli, A.M.; Vitale, M.; Sacchetti, G.; Gonelli, A.; Neri, L.M. Oxidative stress: Role of physical exercise and antioxidant nutraceuticals in adulthood and aging. Oncotarget 2018, 9, 17181–17198. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Perluigi, M.; Joshi, G.; Sultana, R.; Calabrese, V.; Marco, C.D.; Coccia, R.; Butterfield, D.A. In vivo protection by the xanthate tricyclodecan-9-yl-xanthogenate against amyloid β-peptide (1–42)-induced oxidative stress. Neuroscience 2006, 138, 1161–1170. [Google Scholar] [CrossRef] [PubMed]
  108. Anjum, F.; Joshi, K.; Grinkina, N.; Gowda, S.; Cutaia, M.; Wadgaonkar, R. Role of sphingomyelin synthesis in pulmonary endothelial cell cytoskeletal activation and endotoxin-induced lung injury. Am. J. Respir. Cell Mol. Biol. 2012, 47, 94–103. [Google Scholar] [CrossRef] [PubMed]
  109. Di Domenico, F.; Barone, E.; Perluigi, M.; Butterfield, D.A. Strategy to reduce free radical species in Alzheimer’s disease: An update of selected antioxidants. Expert Rev. Neurother. 2015, 15, 19–40. [Google Scholar] [CrossRef]
  110. Chassaing, S.; Collin, F.; Dorlet, P.; Gout, J.; Hureau, C.; Faller, P. Copper and heme-mediated Abeta toxicity: Redox chemistry, Abeta oxidations and anti-ROS compounds. Curr. Top. Med. Chem. 2012, 12, 2573–2595. [Google Scholar] [CrossRef]
  111. Brochier, C.; Langley, B. Chromatin modifications associated with DNA double-strand breaks repair as potential targets for neurological diseases. Neurotherapeutics 2013, 10, 817–830. [Google Scholar] [CrossRef] [Green Version]
  112. Sultana, R.; Perluigi, M.; Butterfield, D.A. Oxidatively modified proteins in Alzheimer’s disease (AD), mild cognitive impairment and animal models of AD: Role of Abeta in pathogenesis. Acta Neuropathol. 2009, 118, 131–150. [Google Scholar] [CrossRef] [Green Version]
  113. Espinet, C.; Gonzalo, H.; Fleitas, C.; Menal, M.J.; Egea, J. Oxidative stress and neurodegenerative diseases: A neurotrophic approach. Curr. Drug Targets 2015, 16, 20–30. [Google Scholar] [CrossRef]
  114. Mir, F.A.; Rizvi, Z.A. Neurobiological mechanisms involved in the pathogenesis of Alzheimer’s disease. In Biological, Diagnostic and Therapeutic Advances in Alzheimer’s Disease; Springer: Berlin, Germany, 2019; pp. 235–269. [Google Scholar]
  115. Bandyopadhyay, U.; Das, D.; Banerjee, R.K. Reactive oxygen species: Oxidative damage and pathogenesis. Curr. Sci. 1999, 77, 658–666. [Google Scholar]
  116. Ott, M.; Gogvadze, V.; Orrenius, S.; Zhivotovsky, B. Mitochondria, oxidative stress and cell death. Apoptosis 2007, 12, 913–922. [Google Scholar] [CrossRef] [PubMed]
  117. Majid, A.S.A.; Yin, Z.Q.; Ji, D. Sulphur antioxidants inhibit oxidative stress induced retinal ganglion cell death by scavenging reactive oxygen species but influence nuclear factor (erythroid-derived 2)-like 2 signalling pathway differently. Biol. Pharm. Bull. 2013, 36, 1095–1110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Zhao, K.; Zhao, G.-M.; Wu, D.; Soong, Y.; Birk, A.V.; Schiller, P.W.; Szeto, H.H. Cell-permeable peptide antioxidants targeted to inner mitochondrial membrane inhibit mitochondrial swelling, oxidative cell death, and reperfusion injury. J. Biol. Chem. 2004, 279, 34682–34690. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Tamiya-Koizumi, K. Nuclear lipid metabolism and signaling. J. Biochem. 2002, 132, 13–22. [Google Scholar] [CrossRef]
  120. Martelli, A.M.; Sang, N.; Borgatti, P.; Capitani, S.; Neri, L.M. Multiple biological responses activated by nuclear protein kinase C. J. Cell Biochem. 1999, 74, 499–521. [Google Scholar] [CrossRef]
  121. Wang, N.; Sun, C.; Huo, S.; Zhang, Y.; Zhao, J.; Zhang, S.; Miao, J. Cooperation of phosphatidylcholine-specific phospholipase C and basic fibroblast growth factor in the neural differentiation of mesenchymal stem cells in vitro. Int. J. Biochem. Cell Biol. 2008, 40, 294–306. [Google Scholar] [CrossRef] [PubMed]
  122. Vieira, H.L.A.; Alves, P.M.; Vercelli, A. Modulation of neuronal stem cell differentiation by hypoxia and reactive oxygen species. Prog. Neurobiol. 2011, 93, 444–455. [Google Scholar] [CrossRef]
  123. Strom, J.O.; Theodorsson, A.; Theodorsson, E. Mechanisms of estrogens’ dose-dependent neuroprotective and neurodamaging effects in experimental models of cerebral ischemia. Int. J. Mol. Sci. 2011, 12, 1533–1562. [Google Scholar] [CrossRef]
  124. Calabrese, V.; Boyd-Kimball, D.; Scapagnini, G.; Butterfield, D.A. Nitric oxide and cellular stress response in brain aging and neurodegenerative disorders: The role of vitagenes. In Vivo 2004, 18, 245–268. [Google Scholar]
  125. Farooqui, A.A.; Antony, P.; Ong, W.-Y.; Horrocks, L.A.; Freysz, L. Retinoic acid-mediated phospholipase A2 signaling in the nucleus. Brain Res. Brain Res. Rev. 2004, 45, 179–195. [Google Scholar] [CrossRef]
  126. Hüttemann, M.; Helling, S.; Sanderson, T.H.; Sinkler, C.; Samavati, L.; Mahapatra, G.; Varughese, A.; Lu, G.; Liu, J.; Ramza, R.; et al. Regulation of mitochondrial respiration and apoptosis through cell signaling: Cytochrome c oxidase and cytochrome c in ischemia/reperfusion injury and inflammation. Biochim. Biophys. Acta 2012, 1817, 598–609. [Google Scholar] [CrossRef] [PubMed]
  127. Li, Y.; Park, J.-S.; Deng, J.-H.; Bai, Y. Cytochrome c oxidase subunit IV is essential for assembly and respiratory function of the enzyme complex. J. Bioeng. Biomembr. 2006, 38, 283–291. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Kalluri, H.S.G.; Dempsey, R.J. D609-mediated inhibition of ATP synthesis in neural progenitor cells. Cellular Mol. Dev. Neurosci. 2014, 25, 777–781. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Warner-Schmidt, J.L.; Schmidt, E.F.; Marshall, J.J.; Rubin, A.J.; Arango-Lievano, M.; Kaplitt, M.G.; Ibanez-Tallon, I.; Heintz, N.; Greengard, P. Cholinergic interneurons in the nucleus accumbens regulate depression-like behavior. Proc. Natl. Acad. Sci. USA 2012, 109, 11360–11365. [Google Scholar] [CrossRef] [Green Version]
  130. Kljakic, O.; Janickova, H.; Prado, V.F.; Prado, M.A.M. Cholinergic/glutamatergic co-transmission in striatal cholinergic interneurons: New mechanisms regulating striatal computation. J. Neurochem. 2017, 142, 90–102. [Google Scholar] [CrossRef]
  131. Tuszynski, M.H. Nerve growth factor gene therapy in Alzheimer disease. Alzheimer Dis. Assoc. Disord. 2007, 21, 179–189. [Google Scholar] [CrossRef]
  132. Dunnett, S.B.; Everitt, B.J.; Robbins, T.W. The basal forebrain-cortical cholinergic system: Interpreting the functional consequences of excitotoxic lesions. Trends Neurosci. 1991, 14, 494–501. [Google Scholar] [CrossRef]
  133. Kim, S.U.; de Vellis, J. Stem cell-based cell therapy in neurological diseases: A review. J. Neurosci. Res. 2009, 87, 2183–2200. [Google Scholar] [CrossRef]
  134. Kim, S.U.; Lee, H.J.; Kim, Y.B. Neural stem cell-based treatment for neurodegenerative diseases. Neuropathology 2013, 33, 491–504. [Google Scholar] [CrossRef]
  135. Jang, S.; Kang, Y.-H.; Ullah, I.; Shivakumar, S.B.; Rho, G.-J.; Cho, C.-Y.; Sung, I.-Y.; Park, B.-W. Cholinergic nerve differentiation of mesenchymal stem cells derived from long-term cryopreserved human dental pulp in vitro and analysis of their motor nerve regeneration potential in vivo. Int. J. Mol. Sci. 2018, 19, 2434. [Google Scholar] [CrossRef] [Green Version]
  136. Kang, Y.-H.; Shivakumar, S.B.; Son, Y.-B.; Bharti, D.; Jang, S.-J.; Heo, K.-S.; Park, W.-U.; Byun, J.-H.; Park, B.-W.; Rho, G.-J. Comparative analysis of three different protocols for cholinergic neuron differentiation in vitro using mesenchymal stem cells from human dental pulp. Anim. Cells Syst. 2019, 23, 275–287. [Google Scholar] [CrossRef] [Green Version]
  137. Sun, C.; Shao, J.; Su, L.; Zhao, J.; Bi, J.; Yang, S.; Zhang, S.; Gao, J.; Miao, J. Cholinergic neuron-like cells derived from bone marrow stromal cells induced by tricyclodecane-9-yl-xanthogenate promote functional recovery and neural protection after spinal cord injury. Cell Transplant. 2013, 22, 961–975. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Shao, J.; Sun, C.; Su, L.; Zhao, J.; Zhang, S.; Miao, J. Phosphatidylcholine-specific phospholipase C/heat shock protein 70 (Hsp70)/transcription factor B-cell translocation gene 2 signaling in rat bone marrow stromal cell differentiation to cholinergic neuron-like cells. Int. J. Biochem. Cell Biol. 2012, 44, 2253–2260. [Google Scholar] [CrossRef] [PubMed]
  139. Sultana, R.; Newman, S.F.; Abdul, H.M.; Cai, J.; Pierce, W.M.; Klein, J.B.; Merchant, M.; Butterfield, D.A. Protective effect of D609 against amyloid-beta1–42-induced oxidative modification of neuronal proteins: Redox proteomics study. J. Neurosci. Res. 2006, 84, 409–417. [Google Scholar] [CrossRef] [PubMed]
  140. Girón-Calle, J.; Srivatsa, K.; Forman, H.J. Priming of alveolar macrophage respiratory burst by H2O2 is prevented by phosphatidylcholine-specific phospholipase C inhibitor tricyclodecan-9-yl-xanthate (D609). J. Pharmacol. Exp. Ther. 2002, 301, 87–94. [Google Scholar] [CrossRef] [Green Version]
  141. Abdul, H.M.; Butterfield, D.A. Protection against amyloid beta-peptide (1–42)-induced loss of phospholipid asymmetry in synaptosomal membranes by tricyclodecan-9-xanthogenate (D609) and ferulic acid ethyl ester: Implications for Alzheimer’s disease. Biochem. Biophys. Acta 2005, 1741, 140–148. [Google Scholar]
  142. Lauderback, C.M.; Drake, J.; Zhou, D.; Hackett, J.M.; Castenga, A.; Kanski, J.; Tsoras, M.; Varadarajan, S.; Butterfield, D.A. Derivatives of xanthic acid are novel antioxidants: Application to synaptosomes. Free Radic. Res. 2003, 37, 355–365. [Google Scholar] [CrossRef]
  143. Müller-Decker, K.; Amtmann, E.; Sauer, G. Inhibition of the phosphorylation of the regulatory non-structural protein of vesicular stomatitis virus by an antiviral xanthate compound. J. Gen. Virol. 1987, 68, 3045–3056. [Google Scholar] [CrossRef]
  144. Waldeck, W. Antiviral xanthate causes conformational changes in simian virus 40 DNA and chromatin. Oncology 1990, 47, 191–198. [Google Scholar] [CrossRef]
  145. Zhang, L.; Li, H.Y.; Li, H.; Zhao, J.; Su, L.; Zhang, Y.; Zhang, S.L.; Miao, J.Y. Lipopolysaccharide activated phosphatidylcholine-specific phospholipase C and induced IL-8 and MCP-1 production in vascular endothelial cells. J. Cell Physiol. 2011, 226, 1694–1701. [Google Scholar] [CrossRef]
  146. Monick, M.M.; Carter, A.B.; Gudmundsson, G.; Mallampalli, R.; Powers, L.S.; Hunninghake, G.W. A phosphatidylcholine-specific phospholipase C regulates activation of p42/44 mitogen-activated protein kinases in lipopolysaccharide-stimulated human alveolar macrophages. J. Immunol. 1999, 162, 3005–3012. [Google Scholar] [PubMed]
  147. Monick, M.M.; Mallampalli, R.K.; Carter, A.B.; Flaherty, D.M.; Peterson, M.W.; Hunninghake, G.W. Ceramide regulates lipopolysaccharide-induced phosphatidylinositol 3-kinase and Akt activity in human alveolar macrophages. J. Immunol. 2001, 167, 5977–5985. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Zhang, F.; Zhao, G.; Dong, Z. Phosphatidylcholine-specific phospholipase C and D in stimulation of RAW264. 7 mouse macrophage-like cells by lipopolysaccharide. Int. Immunopharmacol. 2001, 1, 1375–1384. [Google Scholar] [CrossRef]
  149. Göggel, R.; Winoto-Morbach, S.; Vielhaber, G.; Imai, Y.; Linder, K.; Brade, L.; Brade, H.; Ehlers, S.; Slutsky, A.S.; Schutze, S.; et al. PAF-mediated pulmonary edema: A new role for acid sphingomyelinase and ceramide. Nat. Med. 2004, 10, 155–160. [Google Scholar] [CrossRef]
  150. Iorio, E.; Ricci, A.; Bagnoli, M.; Pisanu, M.E.; Castellano, G.; Vito, M.D.; Venturini, E.; Glunde, K.; Bhujwalla, Z.M.; Mezzanzanica, D.; et al. Activation of phosphatidylcholine cycle enzymes in human epithelial ovarian cancer cells. Cancer Res. 2010, 70, 2126–2135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. Spadaro, F.; Ramoni, C.; Mezzanzanica, D.; Miotti, S.; Alberti, P.; Cecchetti, S.; Iorio, E.; Dolo, V.; Canevari, S.; Podo, F. Phosphatidylcholine-specific phospholipase C activation in epithelial ovarian cancer cells. Cancer Res. 2008, 68, 6541–6549. [Google Scholar] [CrossRef] [Green Version]
  152. Machleidt, T.; Krämer, B.; Adam, D.; Neumann, B.; Schutze, S.; Wiegmann, K.; Kronke, M. Function of the p55 tumor necrosis factor receptor “death domain” mediated by phosphatidylcholine-specific phospholipase C. J. Exp. Med. 1996, 184, 725–733. [Google Scholar] [CrossRef] [Green Version]
  153. Kalluri, H.S.G.; Gusain, A.; Dempsey, R.J. Regulation of neural progenitor cell proliferation by D609: Potential role for ERK. Mol. Neurobiol. 2013, 47, 782–789. [Google Scholar] [CrossRef]
  154. Barbin, G.; Roisin, M.; Zalc, B. Tumor necrosis factor α activates the phosphorylation of ERK, SAPK/JNK, and P38 kinase in primary cultures of neurons. Neurochem. Res. 2001, 26, 107–112. [Google Scholar] [CrossRef]
  155. Wang, N.; Du, C.-Q.; Wang, S.-S.; Xie, K.; Zhang, S.-L.; Miao, J.-Y. D609 induces vascular endothelial cells and marrow stromal cells differentiation into neuron-like cells. Acta Pharmacol. Sin. 2004, 25, 442–446. [Google Scholar]
Figure 1. Structure of tricyclodecan-9-yl xanthogenate (D609).
Figure 1. Structure of tricyclodecan-9-yl xanthogenate (D609).
Ijms 23 03305 g001
Figure 2. D609 blocks PCPLC and SMS and regulates cell cycle and apoptosis.
Figure 2. D609 blocks PCPLC and SMS and regulates cell cycle and apoptosis.
Ijms 23 03305 g002
Figure 3. Role of D609 in human diseases and mechanisms involved.
Figure 3. Role of D609 in human diseases and mechanisms involved.
Ijms 23 03305 g003
Table 1. Pharmacological properties of D609.
Table 1. Pharmacological properties of D609.
Pharmacological PropertiesTarget DiseaseModel UsedMechanism of ActionReference
Antioxidant activityAlzheimer′s disease (AD)Isolated
gerbil brain mitochondria
(50 mg/kg body wt.)
Increased activity of glutathione S-transferase, glutathione peroxidase, and glutathione reductase[18]
Oxidative stress/ionizing radiation-induced oxidative damageIn vitroD609 inhibited the Fenton reaction-induced oxidation of dihydrorhodamine 123.
(1) Production of reactive oxygen species; (2) decrease in intracellular reduced glutathione; (3) oxidative damage to proteins and lipids; and (4) activation of nuclear factor-κB.
[17]
Oxidative stress induced by Ionizing radiationsIn vivo/mouse model
(50 mg/kg i.v)
Inhibited IR-induced cellular oxidative stress.[17]
Aβ(1–42)-induced cytotoxicity/Alzehimer’s diseaseAβ(1–42)-induced oxidative cell toxicity in cultured neuronsD609 significantly attenuated Aβ(1–42)-induced cytotoxicity, intracellular ROS accumulation, protein oxidation, lipid peroxidation, and apoptosis.[26,108,140]
Respiratory burst induced by H2O2Alveolar macrophage respiratory burst
(100 µM D609)
D609 has potential as an antioxidant due to its dithiocarbonate functional group, which allows it to slowly react with H2O2 and rapidly reduce cytochrome c, which interferes with a common assay for the respiratory burst.[141]
Alzheimer′s disease (AD)Abeta (1–42) induced modulation in phospholipid asymmetry in the synaptosomal membranes
(50 mg/kg body wt.)
Aß induced loss of phospholipid asymmetry[142]
Age-related macular degenerationSodium Iodide induced AMD mouse modelIncreased expression of metallothionein[19]
Synaptosomal lipid peroxidation (TBARs), protein oxidation (protein carbonyls), and protein conformationSynaptosomesXanthates scavenge hydroxyl radicals and hydrogen peroxide, form disulfide bonds (dixanthogens), and react with electrophilic products of lipid oxidation (acrolein) in a manner similar to GSH[143]
Antiviral activityRespiratory syncytial (RS) virus growthHuman epithelial (Hep 2) cellsCompound affects the relative proportion of viral proteins and the phosphorylation of P protein[75]
Herpes diseasesHerpes simplex virus type 1It inhibits the protein kinases and protein phosphorylation affecting a late step in HSV replication[76]
HIVHIV-infected KE37-1 cell lineInhibition of HIV-1 Replication[72]
Various DNA and RNA virusHerpes simplex virus types 1 and 2,
Bovine papilloma virus
Replication blocked at the DNA and RNA level both early and late after infection.
Episomal bovine papilloma virus DNA replication and transcription are also inhibited in transformed cells
[71]
Vesicular StomatitisVesicular Stomatitis VirusInhibition of the phosphorylation of the regulatory non-structural protein[144]
Transformation of cells in cancer cellsSimian Virus 40Inhibition of topoisomerase I by more than 1000-fold was detected[145]
Anti-inflammatory activityInflammatory diseases, such as atherosclerosis and septic shockLPS-induced inflammation in vascular endothelial cells
(10 mg/kg)
PC-PLC
inhibition.
Inhibited LPS induced IL-8 and MCP-1
[146]
InflammationLipopolysaccharide (LPS)-stimulated
macrophages
LPS-induced ERK kinase activation was inhibited,
LPS-induced PIP3 kinase activation reduced cytokine expression
[147,148,149]
Autoimmune uveitisExperimental melanin protein-induced uveitis (EMIU)Inhibits inducible nitric oxide synthase (iNOS) induction,
a specific inhibitor of phosphatidylcholine-specific phospholipase C
[80]
Pulmonary edemaASM-deficient or wild-type
control mice with PAF and assessed for the development of pulmonary
edema
Acid sphingomyelinase (ASM)-dependent production of ceramid,
activation of the cyclooxygenase pathway
[150]
Anti-tumour and anti-proliferative activityGlial cell proliferationMurine BV-2 microglia cell lineCeramide and cell-cycle inhibition, inhibiting SMS can increase ceramide levels, which can inhibit cell proliferation.[50]
Metastatic breast cancer cellsHuman MDA-MB-231 cellsInhibition of phosphatidylcholine-specific phospholipase C,
downregulates HER2 overexpression on plasma membrane of breast cancer cells
[7,14]
Human epithelial ovarian cancer cellsOVCAR3 and SKOV3 cancer cellsPC-PLC inhibition[151,152]
FibrosarcomaMouse fibrosarcoma cells L929, Wehi164Through PC-PLC, Anti-inflammatory action, TNF antagonist[44]
LeukemiaU937 human monocytic leukemia cellsSphingomyelin synthase inhibition[43]
Cellular proliferation of neural progenitor cellsNeural progenitor cellsDecreasing the ERK-mediated expression of cyclin D1,
Decreased the phosphorylation of extracellular signal-regulated kinase (ERK) but not Akt
[153]
Neuroprotective activityAge-related macular degeneration
Retinal pigmented epithelium (RPE) cell death
SI-induced AMD mouse modelAttenuated excessive reactive oxygen species (ROS) and prevented severe mitochondrial loss,
increased the expression of metallothionein.
[19]
Alzheimer’s disease (AD)AβPP/PS1 transgenic mouse modelReduced β-secretase 1 level and decreased amyloidogenic processing of AβPP, consequently reducing Aβ deposition in the mice.
Reduced oxidative stress.
[70]
Brain injuryWistar rats, cultures
Obtained from striatum and hippocampus
Inhibitor of phosphoinositide phospholipase C suggesting that TNFαsignaling in neurons involved the acidic sphingomyelinase[154]
Neurodegenerative disordersGerbil brain mitochondriaIncreased activity of glutathione S-transferase, glutathione peroxidase, and glutathione reductase in brain[18]
StrokeTransient middle-cerebral-artery occlusion (tMCAO)D609 provides benefits after stroke through inhibition of SMS, increased ceramide levels, and induction of cell-cycle arrest by up-regulating p21 and causing hypophosphorylation of Rb[16]
Neuroprotective effectneural progenitor cells isolated from the subventricular zone of the rat brainD609 could inhibit the activity of cytochrome C oxidase and subsequent ATP synthesis in neural progenitor cells.[129]
Cholinergic-neuron differentiation activityDifferentiation of cellsCell differentiation in vascular endothelial cells (VECs) and marrow stromal cells (MSCs).D609 induces VECs and MSCs differentiation into neuron-like cells.[155]
Protocols for cholinergic neuron differentiationDental pulp derived MSCs (DPSCs) were used,
DPSCs were cultured in serum-free ADMEM containing 15 µg/mL of D609 (tricyclodecan-9-yl-xanthogenate) for 4 days
Neuron-like morphologies with upregulated cholinergic neuron-specific markers, such as ChAT, HB9, ISL1, BETA-3, and MAP2, both at mRNA and protein levels, were observed[137]
Bone marrow stromal cells (BMSCs) differentiate into neuron-like cellsBone-marrow stromal cells (BMSCs)Inhibition of phosphatidylcholine-specific phospholipase C (PC-PLC) by D609 leads to BMSCs’ differentiation into cholinergic neuron-like cells,
Hsp70 participated in the neural differentiation of BMSCs directly through Btg2.
[139]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Bhat, A.H.; Dar, K.B.; Khan, A.; Alshahrani, S.; Alshehri, S.M.; Ghoneim, M.M.; Alam, P.; Shakeel, F. Tricyclodecan-9-yl-Xanthogenate (D609): Mechanism of Action and Pharmacological Applications. Int. J. Mol. Sci. 2022, 23, 3305. https://doi.org/10.3390/ijms23063305

AMA Style

Bhat AH, Dar KB, Khan A, Alshahrani S, Alshehri SM, Ghoneim MM, Alam P, Shakeel F. Tricyclodecan-9-yl-Xanthogenate (D609): Mechanism of Action and Pharmacological Applications. International Journal of Molecular Sciences. 2022; 23(6):3305. https://doi.org/10.3390/ijms23063305

Chicago/Turabian Style

Bhat, Aashiq Hussain, Khalid Bashir Dar, Andleeb Khan, Saeed Alshahrani, Sultan M. Alshehri, Mohammed M. Ghoneim, Prawez Alam, and Faiyaz Shakeel. 2022. "Tricyclodecan-9-yl-Xanthogenate (D609): Mechanism of Action and Pharmacological Applications" International Journal of Molecular Sciences 23, no. 6: 3305. https://doi.org/10.3390/ijms23063305

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