Next Article in Journal
Combination of Docking-Based and Pharmacophore-Based Virtual Screening Identifies Novel Agonists That Target the Urotensin Receptor
Next Article in Special Issue
Meroterpenoids and Steroids from the Marine-Derived Fungus Trametes sp. ZYX-Z-16
Previous Article in Journal
An Analysis of the Plant- and Animal-Based Hydrocolloids as Byproducts of the Food Industry
Previous Article in Special Issue
Cardioprotective Effects of 6-Gingerol against Alcohol-Induced ROS-Mediated Tissue Injury and Apoptosis in Rats
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Biomedical Importance of the Missing Pathway for Farnesol and Geranylgeraniol Salvage

by
Ignasi Bofill Verdaguer
1,
Marcell Crispim
1,
Agustín Hernández
2 and
Alejandro Miguel Katzin
1,*
1
Department of Parasitology, Institute of Biomedical Sciences of the University of São Paulo, Av. Lineu Prestes 1374, São Paulo 05508-000, Brazil
2
Integrated Unit for Research in Biodiversity (BIOTROP-CCBS), Center for Biological and Health Sciences, Federal University of São Carlos, São Carlos 13565-905, Brazil
*
Author to whom correspondence should be addressed.
Molecules 2022, 27(24), 8691; https://doi.org/10.3390/molecules27248691
Submission received: 10 November 2022 / Revised: 30 November 2022 / Accepted: 5 December 2022 / Published: 8 December 2022

Abstract

:
Isoprenoids are the output of the polymerization of five-carbon, branched isoprenic chains derived from isopentenyl pyrophosphate (IPP) and its isomer, dimethylallyl pyrophosphate (DMAPP). Isoprene units are consecutively condensed to form longer structures such as farnesyl and geranylgeranyl pyrophosphate (FPP and GGPP, respectively), necessary for the biosynthesis of several metabolites. Polyprenyl transferases and synthases use polyprenyl pyrophosphates as their natural substrates; however, it is known that free polyprenols, such as farnesol (FOH), and geranylgeraniol (GGOH) can be incorporated into prenylated proteins, ubiquinone, cholesterol, and dolichols. Furthermore, FOH and GGOH have been shown to block the effects of isoprenoid biosynthesis inhibitors such as fosmidomycin, bisphosphonates, or statins in several organisms. This phenomenon is the consequence of a short pathway, which was observed for the first time more than 25 years ago: the polyprenol salvage pathway, which works via the phosphorylation of FOH and GGOH. Biochemical studies in bacteria, animals, and plants suggest that this pathway can be carried out by two enzymes: a polyprenol kinase and a polyprenyl-phosphate kinase. However, to date, only a few genes have been unequivocally identified to encode these enzymes in photosynthetic organisms. Nevertheless, pieces of evidence for the importance of this pathway abound in studies related to infectious diseases, cancer, dyslipidemias, and nutrition, and to the mitigation of the secondary effects of several drugs. Furthermore, nowadays it is known that both FOH and GGOH can be incorporated via dietary sources that produce various biological effects. This review presents, in a simplified but comprehensive manner, the most important data on the FOH and GGOH salvage pathway, stressing its biomedical importance The main objective of this review is to bring to light the need to discover and characterize the kinases associated with the isoprenoid salvage pathway in animals and pathogens.

1. Isoprenoid Biosynthesis and Distribution

Isoprenoids are the most widespread and diverse group of compounds in nature. They are produced by all organisms and are also thought to be some of the most ancient lipids, originating as constituents of the primitive membranes in the first living organisms [1]. All isoprenoids are made up of five carbons; branched isoprenic chains, derived from isopentenyl pyrophosphate (IPP); and its isomer, dimethylallyl pyrophosphate (DMAPP). Isoprenoids may be synthesized by the mevalonate pathway (MVA pathway) or the non-MVA pathway, also known as the methyl erythritol phosphate pathway (MEP pathway). These are the only well-characterized pathways for IPP/DMAPP production [2,3]. However, there are pieces of evidence for some leucine-dependent pathways which are still not well-characterized [4,5]. These lie outside the scope of this review and, therefore, will be dealt with no further.
The MVA pathway was the first isoprenoid biosynthesis pathway discovered. It is active in plant cytosol, fungi, archaebacteria, eubacteria, and some protozoa groups [2,6]. This pathway starts with the condensation of an acetyl-coenzyme A (acetyl-CoA) molecule with acetoacetyl-CoA to yield 3-hydroxy-3-methyl-glutaryl-CoA (HMG-CoA), a reaction catalyzed by HMG-CoA synthase. Then, the enzyme HMG-CoA reductase (HMGR), produces MVA. This enzymatic step can be inhibited by statins (e.g., simvastatin, lovastatin, atorvastatin, or pitavastatin) [7]. Finally, MVA is phosphorylated twice and decarboxylated to yield IPP/DMAPP. Alternatively, the MEP pathway is present in plant plastids, bacteria, and some protozoa groups, including apicomplexan parasites [2,6]. The MEP pathway starts with the condensation of glyceraldehyde 3-phosphate with pyruvate to form 1-deoxy-D-xylulose-5-phosphate (DOXP). This step is catalyzed by the 1-deoxy-D-xylulose 5-phosphate synthase (DXS). DOXP is next transformed to MEP by the enzyme 1-deoxy-D-xylulose 5-phosphate reductase (DXR). Importantly, DXR can be inhibited by fosmidomycin, which could thus be considered to be the homolog of statins on the MEP pathway. Remarkably, fosmidomycin has been proposed as an antibiotic for several protozoa and bacterial diseases [8,9]. Next in the pathway, MEP suffers modifications, including its condensation with CTP, a subsequent condensation with an ATP molecule, and a reduction. After these steps, hydroxymethyl-butenyl pyrophosphate (HMBPP) is produced. Later, this metabolite is converted into IPP/DMAPP by the enzyme hydroxymethyl-butenyl diphosphate reductase (LytB).
Regardless of the biosynthetic pathway, IPP and DMAPP can be interconverted by the IPP isomerases. In plants, IPP can be directly used for the production of phytohormones [10]. However, most isoprenoid-dependent pathways require isoprenic chains with lengths greater than one isoprenic unit. The first elongation step is the condensation of IPP and DMAPP to form geranyl pyrophosphate (GPP; 10 carbon), this step is catalyzed by the enzyme GPP synthase (GPPs). GPP can be condensed again with an IPP molecule to form farnesyl pyrophosphate (FPP; 15 carbon), with the help of the FPP synthase (FPPs), the enzyme target of bisphosphonates (e.g., alendronate, risedronate), which is widely used as a drug in the treatment of osteopenia and osteoporosis. Similarly, the geranylgeranyl pyrophosphate synthase (GGPPs) catalyzes the production of geranylgeranyl pyrophosphate (GGPP; 20 carbon) by condensing FPP with one more IPP molecule. After the formation of GGPP, several enzymes continue to extend the isoprenic chain to form longer structures: for example, octaprenyl pyrophosphate (8 isoprenic units); nonaprenyl pyrophosphate (9 isoprenic units; also known as solanesyl pyrophosphate); polyprenols of 13–21 isoprenic units; or even longer ones, such as natural rubber (>300 isoprenic units) [11,12]. Isoprenic moieties can suffer various chemical modifications. For example, in most photosynthetic organisms, hydrogenation of GGPP to phytyl pyrophosphate (phytyl-PP) is known to occur. Later, this phytyl-PP serves as a precursor for chlorophyll, tocopherols (vitamin E), and phylloquinone biosynthesis (vitamin K1) [13]. Another example is the reduction polyprenols, of 13–21 isoprenic units, to dolichols in eukaryotic organisms. Dolichols are phosphorylated to dolichyl-P by a dolichol kinase and then mainly employed as lipid anchors for sugar transport in the eukaryotic protein glycosylation and GPI biosynthesis pathways [14,15].
In any case, all polyprenyl pyrophosphates cited previously are employed as substrates for diverse anabolic pathways. For example, FPP and GGPP can be employed for an important post-translational modification of proteins. Typically, protein prenylation occurs when farnesyl or geranylgeranyl moieties are attached to soluble proteins, resulting in their anchoring to membranes. The most studied prenylated proteins are Ras, Rho, and Rap, which are small GTPases involved in cellular signaling and intracellular trafficking [16,17]. This post-translational modification is catalyzed by specific transferases that attach the FPP or GGPP moieties to the C-terminal cysteine residues of proteins containing the conserved motif for prenylation CAAX (C = cysteine, A = aliphatic amino acid, X = diverse terminal residue) [16,17]. Interestingly, alternative isoprenoid moieties were identified when linked to proteins. For example, in plants, a phytylation of proteins also occurs, in this case using phytyl-PP instead of FPP or GGPP [18], and protein dolichylation occurs in malaria parasites and human colon carcinoma cells [19,20]. Besides protein prenylation, polyprenyl pyrophosphates also play a role as constituents of membranes in plants [21,22,23], are required for the formation of menaquinone in bacteria (vitamin K2; requires GGPP) [24], and in the biosynthesis of sterols, like cholesterol in animals (requires FPP) [25]. Finally, polyprenyl-PPs also plays an important part in respiratory processes and lipoperoxidation defense. These molecules are necessary for the biosynthesis of ubiquinones (coenzyme Q), a metabolite that forms part of the electron transport chain at the mitochondrial membrane and an antioxidant cofactor elsewhere. The biosynthesis of ubiquinones involves the condensing of a benzoquinone ring with a polyprenyl-PP moiety of 6–10 isoprenic units; the actual length of the isoprene depends on the organism [26,27].
Isoprenoid biosynthesis is the target of some of the most prescribed drugs worldwide, as already cited. For example, statins are employed to treat hypercholesterolemia, as they inhibit mammalian biosynthesis of cholesterol, while bisphosphonates are used in the treatment of osteoporosis due to their ability to accumulate in bone mineral, inhibiting osteoclast activity [7,28,29]. Finally, fosmidomycin has been purposed as an antibiotic for the treatment of MEP-pathway-dependent protozoa and bacteria [8,30]. Moreover, isoprenoid biosynthesis inhibitors are under study for the treatment of many other diseases, which will be detailed below. An overview of the different pathways in isoprenoid metabolism and drugs discussed in this review is shown in Figure 1.
As explained above, the natural substrates for the biosynthesis of several cellular metabolites are the polyprenyl-PPs. However, their unphosphorylated counterparts, polyprenols (also referred to here as isoprenoid alcohols, despite their length), are also widespread, from bacteria to human cells [31,32,33,34]. Plants are probably the group of organisms where these metabolites have been most extensively studied [34]. Plants such as the acacia tree, Acacia caven, contain great amounts of farnesol (FOH) [35]; geranylgeraniol (GGOH) is particularly abundant in annatto (Bixa orellana) [36], as is solanesol in Nicotiana tabacum [37], and polyprenols of 13 isoprenic units in conifer extracts [38]. Medium-length isoprenoid alcohols of 3–4 isoprenic units in length serve as components of plant membranes [23,39]. However, just a few biosynthetic pathways seem to employ directly unphosphorylated polyprenols. For example, FOH is used to synthesize the hormone farnesoic acid by Diploptera punctata (Pacific beetle cockroach) [40], and, in most mammals, dietary phytol is degraded to phytanic acid for its excretion [41]. Similarly, both FOH and GGOH can be catabolized by mammals to farnesal and geranylgeranial, and subsequently to farnesoic acid and geranylgeranoic acid and some prenyl dicarboxylic acids. The enzymes for this pathway had already been identified as an excretion mechanism of polyprenols [32,42]. However, the most studied function of both FOH and GGOH in mammals is their role as metabolic regulators. Thus, FOH and GGOH possess a great ability to promote the degradation of the enzyme HMG-CoA reductase, resulting in a reduction in the MVA pathway, with concomitant reductions in the production of cholesterol or steroidogenic hormones [43,44].

2. The Origin of Geranylgeraniol and Farnesol

FOH and GGOH are also produced from GGPP and FPP by cells through the action of (already identified) specific phosphatases in mammals [45,46,47,48], plants [49], and insect tissues [50]. In mammalian-cultured cells, it has been observed that the overexpression of these phosphatases substantially decreases the prenylation of proteins and, as a result, induce cell growth defects, dysregulate the MVA pathway via degradation of the enzyme HMG CoA reductase, affect the organization of the cytoskeleton, and deregulate Rho proteins [45,46]. Therefore, it has been suggested that polyprenyl-PP phosphatases contribute to a feedback mechanism involved in maintaining optimal isoprenoid intracellular levels. In addition to endogenous FOH and GGOH, their dietary sourcing in mammals has been recently demonstrated [51,52]. Several foods, especially those of plant origin (e.g., sunflower oil, coconut oil, olive oil, tomato, or lettuce), display a high level of FOH and GGOH, which could be incorporated into animal cells after ingestion [52]. In addition to these, GGPP and FPP are constituents of all living organisms; thus, they are expected to be present in most foods. However, the pyrophosphate moiety of these compounds is unstable under acidic conditions and, therefore, are likely to be absorbed in digestion as isoprenoid alcohols [53]. Despite their importance, the reality of their dietary uptake, and their modulatory effects on biosynthesis, there is still no precise quantification of FOH and GGOH in human tissues (based on data from the Human Metabolome Database site, http://www.hmdb.ca/. Last accessed on 11 August 2022). For these reasons, and others that will be detailed in the next pages, they will be considered as nonessential nutrients of rising biomedical interest.

3. Geranylgeraniol and Farnesol Are Incorporated into Cellular Components

Both FOH and GGOH show several bioactive and clinical properties, among them, antibacterial [54], anti-tumorigenic [55], anti-inflammatory [56], and neuroprotective action [57,58], in addition to the regulation of hormone production [59]. However, the greatest biological activity of FOH and GGOH is related to their pyrophosphate derivatives, not to the alcoholic forms, i.e., their ability to provide a bypass to the isoprenoid biosynthetic pathway. In most cellular models, FOH and GGOH have a great ability to protect the cells from pharmacological or genetic-induced defects in isoprenoid metabolism. However, not all defects have been observed to be rescued. For example, there is no evidence that defects in cholesterol biosynthesis can be rescued by supplementation with GGOH. As was to be expected, it seems that the FOH- and GGOH-rescue effects are restricted to defects in enzymes of isoprenoid metabolism that are prior to the formation of the respective pyrophosphate forms of FPP and GGPP in biosynthesis. Considering this, GGOH has been showed to partially restore statin-induced contraction fatigue in mammalian muscle cells [60], prevent cytotoxicity in bisphosphonate-treated oral mucosa cells and fibroblasts [61,62], or ameliorate the phenotypes associated with a deficiency in MVA kinase in murine models and human cultured cells [63,64]. Furthermore, GGOH is required to maintain endotoxin tolerance in macrophages [65] and reverses the toxic effects of mevastatin in monocytic human THP-1 cells, without affecting its ability to reduce cholesterol synthesis [66]. Stemming from this, several authors have employed radio-labelled polyprenols to assess their possible incorporation into proteins. For example, the incubation of apicomplexan parasites, mammalian, or plant cells with either radio-labelled FOH or GGOH, has presented the incorporation of radioactivity into proteins [67,68,69,70]. Besides protein prenylation, evidences exist for the incorporation of radio-labelled FOH or GGOH into ubiquinone [71,72,73], dolichols [74], and steroids such as cholesterol [71,72,75], in different organisms. Interestingly, it has been observed that protein farnesylation and/or geranylgeranylation are the major isoprenoid-dependent processes for short-term viability in some organisms. Specifically, the biological importance of protein prenylation has been studied through metabolomics, drug-rescue assays and microscopic techniques in yeasts [76], in malaria parasites treated with isoprenoid biosynthesis inhibitors [77], as well as in the Chinese hamster ovary cell line UT-2, that has a defect in HGM-CoA reductase and is auxotrophic for MVA [78]. In all these cases, cell viability was not readily affected by deficiencies in ubiquinone, cholesterol, or dolichol biosynthesis, but solely by the protein prenylation defects.

4. The Geranylgeraniol and Farnesol Salvage Pathway

Despite the FOH and GGOH metabolization by several organisms, the natural substrates of all characterized polyprenyl transferases and synthases are polyprenyl pyrophosphates. Further, no evidence exists for this kind of enzyme being able to use polyprenols or any other polyprenyl derivative. This is likely due to the different orientation of these two types of lipids in the membrane. While polyprenols could be oriented parallel to the plane of the membrane, polyprenyl pyrophosphates strongly favor a perpendicular orientation, with their pyrophosphate moieties fiscally available for interaction with polyprenyl transferases and synthases [1,79]. Therefore, the incorporation of FOH and GGOH into the major isoprenoid pathways has been proposed to occur by the existence of a polyprenol salvage pathway acting via phosphorylation [76]. To the best of our knowledge, the first evidence for the existence of this pathway was presented in 1994 by Inoue et al. The authors observed CTP-mediated monophosphorylation of radio-labelled FOH in the 100,000 g membrane pellet from the microalga Botryococcus braunii [80]. The same activity was also observed in membrane extracts of the archeobacterium Sulfolobolus acidocaldarius two years later [81]. In this case, monophosphorylalation of radio-labelled GGOH was detected using a mixture of CTP, ATP, GTP, and UTP. Furthermore, a second phosphorylation, producing GGPP from GGP, was also detected using cytosolic fractions from S. acidocaldarius. The first evidence of this pathway in mammals was reported in 1998 by Bentinger et al. [82]. These authors observed farnesyl monophosphate (FP) being enzymatically synthesized from FOH in the 10,000 g supernatant of rat liver homogenates. Although this activity was observed with several nucleotides, the greatest activity was observed using ATP. This FOH kinase activity was located in rough and smooth microsomes, and associated with the inner, luminal surface of the vesicles. Further analyses in this system identified an activity able to phosphorylate FP to FPP. In this case, the catalytic activity was specific to CTP and probably localized on the cytoplasmic outer surface of microsomal vesicles. Alterations in FOH kinase and FPP phosphatase activities were observed in whole liver homogenates from rats receiving cholesterol- or cholestyramine-rich diets, compounds known to downregulate or up regulate the MVA pathway, respectively [83]. Remarkably, only FOH kinase was decreased in whole liver homogenates from rats on a cholesterol-rich diet. This suggests that FOH salvage can be inhibited if cholesterol requirements are satisfied.
In plants, phosphorylation of FOH and GGOH to their respective mono- and diphosphates was first observed in Nicotiana tabacum cell cultures [70]. In this case, the authors indicated a mechanism involving two successive monophosphorylation reactions, since radio-labelled CTP was formed when microsomes were put in contact with [3H] CDP and FPP or GGPP, but not with FP. Similar to this, a phytol salvage pathway was described in Arabidopsis thaliana [84]. The authors assessed if phytol could be phosphorylated to phytyl phosphate (phytyl-P) and phytyl-PP by two kinases located at the envelope membranes of plastids. The first phosphorylation was found to be CTP dependent, while the second one could be mediated by either CTP, GTP, UTP, or ATP. Our group also observed similar polyprenol phosphorylation reactions in malaria parasite extracts (Plasmodium falciparum); in this system, most probably, those reactions could serve as a mechanism to recycle polyprenols for the biosynthesis of parasitic cofactors. Furthermore, we observed parasites that could use these phosphorylated isoprenoid alcohols, including in Ras and Rap protein prenylation, in addition to condensing them into longer polyprenols and dolichols [85].

5. The Few Polyprenol/Polyprenyl-P Kinases Discovered

The consensus, thus far, is that the polyprenol salvage pathway is composed by two independent enzymes: a polyprenol kinase and a polyprenyl phosphate kinase [72]. However, to date, only a handful of genes have been unequivocally identified to encode this kind of enzyme; in Solanum lycopersicum plants, the FOLK and VTE5 genes encode FOH kinase and phytol kinase, respectively [86]. The Synechocystis sp. gene ScVTE5 encodes a phytol kinase [87]. Finally, in A. thaliana, the AtFOLK gene encodes a FOH kinase [88], namely, the AtVTE5 gene, by encoding a plastidic phytol kinase [87,89], and the recently described gene AtVTE6, by encoding a plastidic phytyl-P kinase [90]. In all these cases, the enzymes require divalent cations and, although they can use different nucleotide trisphosphates, they showed a strong preference for CTP [87,88,89,90]. Concomitantly, some evidences on the great biological importance of the polyprenol salvage pathway in plants emerged. First, AtFOLK gene expression was shown to be negatively regulated by the phytohormone abscisic acid. As a consequence, loss-of-function-FOLK mutants displayed an abscisic acid-hypersensitive phenotype and anomalous flower growth under abiotic stress, with the development of supernumerary carpels [88]. On the other hand, defects in the VTE5 and VTE6 genes in plants produced accumulations of free phytol and deficiencies in phytyl-PP-dependent pathways, such as tocopherol and vitamin K1 biosynthesis, as well as photosystem I instability and the reduced tolerance to abiotic stress [87,89,90]. All of these results suggest that de novo phytyl-PP biosynthesis may not be the major source of phytol for vitamin E and K1 biosynthesis in plants, but the remobilization of phytol from chlorophyl degradation.
In terms of structure, these genes and their encoded proteins show similarities and differences which must be commented on. The first of the genes reported, VTE5, was identified in mutagenesis studies of plants defective for vitamin E and K1 biosynthesis. FOLK was identified to encode a FOH kinase by similarity with VTE5 [88]. On the other hand, both VTE5 and FOLK gene products seem to be related enzymes, despite their different functions. Moreover, both show a modest similarity to dolichol kinase [90]. Remarkably, the putative function of VTE6 as a phytyl-P kinase was first suggested when an analysis of similarities with bacterial genomes was carried out [91]. This gene showed similarities to those genes after putatively encoding for missing steps in isoprenoid metabolism, particularly with COG1836 sequences from photosynthetic prokaryotes. These genomic comparison tools could help in future identifications of polyprenol kinase genes in other organisms.
The biochemical characterization of these proteins was not straightforward. The identified polyprenol kinases needed to be recombinantly expressed in S. cerevisiae and E. coli, organisms where polyprenol and polyprenyl-P kinase activities are not naturally found [87,88,90]. Assays were performed in membrane extracts and required the use of radioactive-labelled substrates, followed by chromatography. Exceptionally, the recombinant expression of phytol kinase and phytyl-P kinase in E. coli allowed for phytol-feeding experiments, followed by the analysis of the product by mass spectrometry [87,90]. Advances in techniques using stable isotope labelling would be most beneficial in this field. Unfortunately, to date, no such assays have been reported for these kind of enzymatic activities.

6. Biomedical Importance of the Geranylgeraniol and Farnesol Salvage Pathway

As mentioned above, the polyprenol salvage pathway was first observed more than 25 years ago, but the enzymes responsible in animals or any pathogen are still unidentified [72,80]. Despite this, the interest in this pathway for the treatment of several human pathologies, ranging from osteonecrosis to infectious diseases, is increasing [92,93]. As a consequence, it is impractical to mention here everything published in this field, and we needed to make a selection of those reports that best illustrated the topics discussed, with a focus on biomedical problems.

6.1. Oncology

Oncology is probably the main area where polyprenols have been found to exert a dramatic role. This is somewhat unsurprising since isoprenoids are required for several cancer-related processes such as the Hedgehog pathway, the nuclear localization of the Yes-associated protein, steroid hormone formation, and transcriptional co-activation with the PDZ-binding motif. Furthermore, protein prenylation appears to be strongly related to oncogenesis and post-translational modification of growth-related oncoproteins such as Ras, Rac, and the Rho GTPase family [94]. Among all these proteins, alterations in Ras genes (including the oncogene KRAS) are very common in cancer, where they are present in approximately 22% of cases of these pathologies [95,96]. The search for treatments that effectively regulate this type of altered proteins is still on-going and thus, the issue is known as “the Ras problem”, and represents the focus of the NIH Ras Initiative (https://www.cancer.gov/news-events/cancer-currents-blog/2015/turning-off-broken-switch, last accessed on 7 July 2022). Among the strategies devised, inhibitors of the MVA pathway for the treatment and prevention of different types of cancers have been explored for decades. However, the use of statins mostly shows an unsatisfying ability to treat and prevent cancer [51,97,98,99]. Remarkably, it has been shown that GGOH can limit the antineoplastic activity of statins against some types of cancer, such as ovarian carcinoma and neuroblastoma [51,98]. Furthermore, de Wolf et al. observed extracts from foods rich in GGOH, such as vegetable oils, being able to block pitavastatin-induced apoptosis in ovarian cancer cells, suggesting that diet may influence the outcome of clinical trials. In agreement with this, oral administration of pitavastatin has caused regression of ovarian tumour xenografts in mice maintained on a GGOH-free diet. However, when their diet was supplemented with GGOH, pitavastatin failed in reducing tumor growth. This is a strong indication that GGOH may be naturally, exogenously incorporated and bioactive in animals. Recently in 2022, GGOH-deficient diets have been designed to make statins viable for the treatment of some types of human cancers [52]. Interestingly, polyprenols alone have long been known to affect tumor cell proliferation [55,100], but the interest in these compounds has not dwindled. Recently, the application of low doses of farnesol was observed to reduce proliferation of lung cancer A549 and colon adenocarcinoma Caco-2 cell lines, compared with a healthy human lung epithelial BEAS-2B cell line, but, surprisingly, at high concentrations the situation reversed [101]. The antioxidant property of farnesol may be one of the reasons for this antitumoral activity, as it has been shown to protect rats against diethyl nitrosamine-induced hepatocellular carcinoma [102].

6.2. Infectious Diseases

The use of MVA pathway inhibitors as antibiotics and antiviral agents was also explored in several reports. Among the organisms targeted, there were viruses (e.g., respiratory syncytial virus, murine cytomegalovirus, gamma herpesvirus and influenza virus), protozoa (e.g., Leishmania donovani, Leishmania major and Toxoplasma gondii), fungi (e.g., Candida spp. and Cryptococcus neoformans), and bacteria (e.g., Listeria monocytogenes, Coxiella burnetiid, Rickettsia conorii and Staphylococcus aureus) [103,104,105]. In general, statins compromised the pathogen’s ability of immune evasion and survival, associated with a deficit of host-derived MVA pathway intermediates [103,104,105]. Similarly, the MEP-pathway has also been found to be an interesting drug target against Plasmodium falciparum [30,106,107,108], Toxoplasma gondii [109], Babesia microti [110], or Mycobacterium tuberculosis [111]. Furthermore, fosmidomycin has already been evaluated in several clinical trials as a promising treatment for malaria, albeit with limited success [112]. Importantly, most MEP-dependent pathogens can also be rescued from fosmidomycin by FOH and GGOH [77,109,110,111,112,113,114]. Therefore, the incorporation of exogenous polyprenols seems to be a possible mechanism limiting the efficacy of treatments against many pathogens and cancer. It could be envisaged, then, that pharmacological inhibition of FOH/GGOH phosphorylation could be of interest to potentiate the effects of isoprenoid biosynthesis inhibitors in a therapeutical setting.

6.3. Isoprenoid Deficiencies

The polyprenol salvage pathway plays a role in several conditions etiologically related to isoprenoid deficiencies. These deficiencies may occur as a side effect of pharmacological treatment, during aging, due to a genetic disorder or idiopathic reasons. For exemple, osteoporosis is a bone calcification deficiency that correlates with aging and is usually treated with biphosphponates [115]. Osteonecrosis of the jaw is the most severe adverse effect in patients on bisphosphonates [116,117]. At therapeutical doses, bisphosphonates decrease the levels of prenyl pyrophosphates available for the prenylation of GTPases in osteoclasts, inhibiting bone resorption [28,29]. However, if in excess, bisphosphonates can also uncouple bone remodeling and healing, leading to osteonecrosis. In this context, isoprenoids, with GGOH among them, have been shown in in vitro and in vivo models to be able to restore GTPase prenylation and, thus, to be potentially useful in preventing and treating osteonecrosis of the jaw [116,117].
Statins, used to manage dyslipidemias, also possess several side effects, including headache, dizziness, discomfort or weakness in skeletal muscles, constipation, diarrhea, and hepatotoxicity [118]. Specifically, hepatotoxicity and muscle pain are the most severe side effects, and are associated with deficiencies in protein prenylation [94,119,120]. Furthermore, long term treatment with statins has been shown to decrease dolichol and ubiquinone levels and, as a consequence, makes organisms more susceptible to oxidative damage, and to be inefficient in protein post-translational modifications and in obtaining energy via oxidative phosphorylation [94,119,120]. Despite statin side effects being rarely severe, at times it is necessary to stop the therapy, exposing the patient to an increased cardiac risk [118]. With this in mind, the administration of ubiquinone complements was first proposed as a strategy to ameliorate statin side effects [121,122]. However, these complements were not demonstrated to satisfactorily reduce those major side effects, and ubiquinone was poorly orally absorbed [121,122]. Recently, commercial GGOH rich extracts and preparations were launched that have shown promising effects of restoring protein prenylation in this context [94,119,120]. In particular, GGOH supplements supported a beneficial effect against statin-induced myopathy and in the prevention of skeletal muscle fatigue in preliminary studies [119,120]. It must be noted that polyprenols could also be effective against statin-induced insulin resistance in muscle tissue [123] and against the dysregulation of hepatic glucose metabolism [124]. Furthermore, polyprenol-rich extracts of >13 isoprenic units was also commercialized. Once incorporated, these polyprenols were probably converted into dolichols, and then phosphorylated to dolichyl-P by dolichol kinase. In this case, however, dolichols were seen to have no protective effect against pitavastatin [49], which is in agreement with them being uninvolved in protein prenylation. Conceivably, another possible strategy to increase physiological levels of FPP and GGPP in statin-treated patients could be the inhibition of polyprenyl-PP phosphatase. However, this possibility remains unexplored experimentally.

6.4. Dyslipidemias

It could be envisaged that FOH may promote dyslipidemias, as some studies demonstrated that this compound is converted into cholesterol in human cells due to the polyprenols salvage pathway (a metabolic route presumably not regulable with statins [72,75]). However, the contribution of FOH to high cardiovascular risk has not been proven, while the opposite effect has been reported. Abukhalil et al., showed that administration of FOH attenuated oxidative stress and liver injury, at the same time that it modulated fatty acid synthase and acetyl-CoA carboxylase in high cholesterol-fed rats [125]. Similarly, long chain polyprenols and phytol have been shown to have a direct effect as lipid-lowering compounds [126]. Mechanistically, FOH would act similarly as a statin, downregulating MVA pathway, at the same time as the fatty acid synthase and acetyl-CoA carboxylase, while preserving protein farnesylation. Therefore, FOH itself may be an interesting novel strategy for the treatment of dyslipidemias.

6.5. Rare Diseases

Mutations in the human mevalonate kinase gene are the cause of a short spectrum of related rare diseases. Mevalonate kinase deficiency (ORPHA code 309025; see www.orpha.net, last access: 21 October 2022) is caused by hypomorphic mutations, and the patients typically display fever episodes and hyperimmunoglobulinemia [127], while complete loss of function of the MVK gene brings about mevalonic acidura (ORPHA code 29; see www.orpha.net, last access: 21 October 2022), a severe form of the condition associated with short life expectancy and dysmorphic features. Since the major effects of these diseases are also related to defects from isoprenoids biosynthesis and protein prenylation, the dietary supplementation with polyprenols has been tested as a treatment for mevalonate kinase deficiency. Again, the administration of geraniol, FOH, and GGOH improved symptoms and inflammation markers both in in vitro and in vivo models [63,128,129]. Interestingly, polyprenols could also act as anti-inflammatory agents per se [56], as well as being effective compounds to promote neuroprotection [57,58] or regulate hormone production [59]. What is especially interesting is the GGOH ability to attenuate lipopolysaccharide-induced inflammation in rats and mouse-derived microglial cells, a phenomenon strongly associated with several neurodegenerative diseases [130,131]. While some of these effects of polyprenols may one day be ascribed to its salvage, many may be related to other characteristics, such as their intrinsic ability to reduce oxidative stress [132]. Thus, this falls outside the scope of this review.

7. Conclusions and Perspectives

In animals, both FOH and GGOH can be enzymatically synthetized or incorporated from the diet, producing diverse biological effects. Most effects are the consequence of the existence of a pathway for polyprenol salvage, composed by a polyprenol kinase and a polyprenyl-phosphate kinase. Once FOH and GGOH are pyrophosphorylated, they are incorporated into the protein prenylation process, as well as in other isoprenoid-dependent anabolic pathways, such as the synthesis of ubiquinone, cholesterol, and dolichol. This FOH/GGOH salvage pathway seems to be active in bacteria, animals, and plants as a mechanism to regulate or by-pass the isoprenoid biosynthetic pathway. However, to date, just a few enzymes have been identified, only in plants and cyanobacteria. Most likely, the study of this pathway has suffered from technical problems: first, these enzymes seem to present little sequence similarity to others with like functions, and no easy complementation assays are available. Furthermore, polyprenyl kinases are presumably membrane proteins and thus, are difficult to express heterologously. Enzyme activity and cellular polyprenol-distribution assays are cumbersome, since they usually require radio-labelling techniques, which are nowadays much in disuse. As a consequence, the enzymes responsible for this pathway in animals or any pathogen are still unidentified. However, considering the evidence, it seems patent that the identification of these enzymes could shed light on several infectious diseases, cancer, dyslipidemias, nutritional deficiencies, and the secondary effects of several drugs. Throughout this text, we aimed to present, in a simplified but comprehensive way, the main data on the FOH and GGOH salvage pathway, emphasizing its biomedical importance. Therefore, the main goal of this review was to raise awareness among the scientific community of the need for a better study of the polyprenol salvage pathway, and to discover the corresponding kinases in animals and pathogens.

Author Contributions

I.B.V., M.C. and A.H. contributed in conceptualization, formal analysis, investigation, methodology, and writing. A.M.K. and A.H. contributed in conceptualization, data curation, formal analysis, funding acquisition, investigation, project administration, supervision, and writing—review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP) process numbers: 2017/22452-1 and 2014/10443-0, awarded to A.M.K. and A.H.L., respectively, Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES) and Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq). I.B.V. and M.C are fellows from FAPESP process numbers 2019/13419-6 and 2018/02924-9 respectively.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

I.B.V., M.C., A.H. and A.M.K. would like to thank Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP), Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES) and Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq) for the financial support.

Conflicts of Interest

The authors declare that they have no conflicts of interest.

Sample Availability

Samples of the compounds are not available from the authors.

Abbreviations

Acetyl-CoAAcetyl-coenzyme A
ATPAdenosine triphosphate
CoACoenzyme A
CDPCytidine diphosphate
CTPCytidine triphosphate
DMAPPDimethylallyl diphosphate
DOXP1-deoxy-D-xylulose 5-phosphate
DXR1-Deoxy-d-xylulose 5-phosphate reductase
DXS1-Deoxy-d-xylulose 5-phosphate synthase
FOHFarnesol
FPFarnesyl monophosphate
FPPFarnesyl pyrophosphate
FPPsfarnesyl pyrophosphate synthase
GGPPGeranylgeranyl pyrophosphate
GGPPsGeranylgeranyl pyrophosphate synthase
GGOHGeranylgeraniol
GPPGeranyl pyrophosphate
GPPsGeranyl pyrophosphate synthase
HMBPPHydroxymethylbutenyl diphosphate
HMG3-Hydroxy-3-methyl-glutaryl
HMG-CoA3-Hydroxy-3-methyl-glutaryl- coenzyme A
HMGR3-Hydroxy-3-methyl-glutaryl- coenzyme A reductase
IPPIsopentenyl diphosphate
LytBHydroxymethylbutenyl diphosphate reductase
MEPMethyl erythritol phosphate
MVAMevalonate
UTPUridine triphosphate

References

  1. Nakatani, Y.; Ribeiro, N.; Streiff, S.; Gotoh, M.; Pozzi, G.; Désaubry, L.; Milon, A. Search for the most ‘primitive’ membranes and their reinforcers: A review of the polyprenyl phosphates theory. Orig. Life Evol. Biosph. 2014, 44, 197–208. [Google Scholar] [CrossRef] [Green Version]
  2. Lichtenthaler, H.K. Non-mevalonate isoprenoid biosynthesis: Enzymes, genes and inhibitors. Biochem. Soc. Trans. 2000, 28, 785–789. [Google Scholar] [CrossRef]
  3. Miziorko, H.M. Enzymes of the mevalonate pathway of isoprenoid biosynthesis. Arch. Biochem. Biophys. 2011, 505, 131–143. [Google Scholar] [CrossRef] [Green Version]
  4. Suga, T.; Hirata, T.; Shishibori, T.; Tange, K. The first proof of the biosynthesis of isoprenoid from amino acid in higher plant. The incorporation of L-Leucine into linalool. Chem. Lett. 1974, 3, 189–192. [Google Scholar] [CrossRef]
  5. Ginger, M.L.; Chance, M.L.; Sadler, I.H.; Goad, L.J. The biosynthetic incorporation of the intact leucine skeleton into sterol by the trypanosomatid Leishmania mexicana. J. Biol. Chem. 2001, 276, 11674–11682. [Google Scholar] [CrossRef] [Green Version]
  6. Verdaguer, I.B.; Zafra, C.A.; Crispim, M.; Sussmann, R.A.; Kimura, E.A.; Katzin, A.M. Prenylquinones in human parasitic protozoa: Biosynthesis, physiological functions, and potential as chemotherapeutic targets. Molecules 2019, 24, 3721. [Google Scholar] [CrossRef] [Green Version]
  7. Stancu, C.; Sima, A. Statins: Mechanism of action and effects. J. Cell. Mol. Med. 2001, 5, 378–387. [Google Scholar] [CrossRef]
  8. Jackson, E.R.; Dowd, C.S. Inhibition of 1-deoxy-D-xylulose-5-phosphate reductoisomerase (Dxr): A review of the synthesis and biological evaluation of recent inhibitors. Curr. Top. Med. Chem. 2012, 12, 706–728. [Google Scholar] [CrossRef]
  9. Lell, B.; Ruangweerayut, R.; Wiesner, J.; Missinou, M.A.; Schindler, A.; Baranek, T.; Kremsner, P.G. Fosmidomycin, a novel chemotherapeutic agent for malaria. Antimicrob. Agents Chemother. 2003, 47, 735–738. [Google Scholar] [CrossRef] [Green Version]
  10. Kakimoto, T. Biosynthesis of cytokinins. J. Plant Res. 2003, 116, 233–239. [Google Scholar] [CrossRef]
  11. Kellogg, B.A.; Poulter, C.D. Chain elongation in the isoprenoid biosynthetic pathway. Curr. Opin. Chem. Biol. 1997, 1, 570–578. [Google Scholar] [CrossRef]
  12. Yamashita, S.; Takahashi, S. Molecular mechanisms of natural rubber biosynthesis. Annu. Rev. Biochem. 2020, 89, 821–851. [Google Scholar] [CrossRef] [PubMed]
  13. Gutbrod, K.; Romer, J.; Dörmann, P. Phytol metabolism in plants. Prog. Lipid Res. 2019, 74, 1–17. [Google Scholar] [CrossRef] [PubMed]
  14. Skorupinska-Tudek, K.; Wojcik, J.; Swiezewska, E. Polyisoprenoid alcohols—Recent results of structural studies. Chem. Rec. 2008, 8, 33–45. [Google Scholar] [CrossRef]
  15. Burda, P.; Aebi, M. The dolichol pathway of N-linked glycosylation. Biochim. Et Biophys. Acta (BBA)-Gen. Subj. 1999, 1426, 239–257. [Google Scholar] [CrossRef]
  16. Glomset, J.A.; Gelb, M.H.; Farnsworth, C.C. Prenyl proteins in eukaryotic cells: A new type of membrane anchor. Trends Biochem. Sci. 1990, 15, 139–142. [Google Scholar] [CrossRef]
  17. McTaggart, S.J. Isoprenylated proteins. Cell. Mol. Life Sci. CMLS 2006, 63, 255–267. [Google Scholar] [CrossRef]
  18. Gutkowska, M.; Bieńkowski, T.; Hung, V.S.; Wanke, M.; Hertel, J.; Danikiewicz, W.; Swiezewska, E. Proteins are polyisoprenylated in Arabidopsis thaliana. Biochem. Biophys. Res. Commun. 2004, 322, 998–1004. [Google Scholar] [CrossRef]
  19. D’Alexandri, F.L.; Kimura, E.A.; Peres, V.J.; Katzin, A.M. Protein dolichylation in Plasmodium falciparum. FEBS Lett. 2006, 580, 6343–6348. [Google Scholar] [CrossRef] [Green Version]
  20. Hjertman, M.; Wejde, J.; Dricu, A.; Carlberg, M.; Griffiths, W.J.; Sjövall, J.; Larsson, O. Evidence for protein dolichylation. FEBS Lett. 1997, 416, 235–238. [Google Scholar] [CrossRef]
  21. Hartley, M.D.; Imperiali, B. At the membrane frontier: A prospectus on the remarkable evolutionary conservation of polyprenols and polyprenyl-phosphates. Arch. Biochem. Biophys. 2012, 517, 83–97. [Google Scholar] [CrossRef] [Green Version]
  22. Van Gelder, K.; Rea, K.A.; Virta, L.; Whitnell, K.L.; Osborn, M.; Vatta, M.; Khozin, A.; Skorupinska-Tudek, K.; Surmacz, L.; Akhtar, T.A. Medium-Chain Polyprenols Influence Chloroplast Membrane Dynamics in Solanum lycopersicum. Plant Cell Physiol. 2018, 59, 2350–2365. [Google Scholar] [CrossRef]
  23. Swiezewska, E.; Danikiewicz, W. Polyisoprenoids: Structure, biosynthesis and function. Prog. Lipid Res. 2005, 44, 235–258. [Google Scholar] [CrossRef]
  24. Bentley, R.; Meganathan, R. Biosynthesis of vitamin K (menaquinone) in bacteria. Microbiol. Rev. 1982, 46, 241–280. [Google Scholar] [CrossRef]
  25. Russell, D.W. Cholesterol biosynthesis and metabolism. Cardiovasc. Drugs Ther. 1992, 6, 103–110. [Google Scholar] [CrossRef]
  26. Kawamukai, M. Biosynthesis, bioproduction and novel roles of ubiquinone. J. Biosci. Bioeng. 2002, 94, 511–517. [Google Scholar] [CrossRef]
  27. Nowicka, B.; Kruk, J. Occurrence, biosynthesis and function of isoprenoid quinones. Biochim. Et Biophys. Acta (BBA)-Bioenerg. 2010, 1797, 1587–1605. [Google Scholar] [CrossRef] [Green Version]
  28. Fleisch, H.; Reszka, A.; Rodan, G.; Rogers, M. Bisphosphonates: Mechanisms of action. Princ. Bone Biol. 2002, 1, 1361-XLIII. [Google Scholar]
  29. Rogers, M.J.; Watts, D.J.; Russell RG, G. Overview of bisphosphonates. Cancer: Interdiscip. Int. J. Am. Cancer Soc. 1997, 80, 1652–1660. [Google Scholar] [CrossRef]
  30. Wiesner, J.; Jomaa, H. Isoprenoid biosynthesis of the apicoplast as drug target. Curr. Drug Targets 2007, 8, 3–13. [Google Scholar] [CrossRef]
  31. Schroepfer, G.J., Jr.; Gore, I.Y. Chromatographic separation of allylic alcohols on silicic acid columns: Analysis of the nonsaponifiable lipids of an ascites tumor derived from a benzpyrene-induced sarcoma. J. Lipid Res. 1963, 4, 266–269. [Google Scholar] [CrossRef]
  32. Endo, S.; Matsunaga, T.; Ohta, C.; Soda, M.; Kanamori, A.; Kitade, Y.; Hara, A. Roles of rat and human aldo–keto reductases in metabolism of farnesol and geranylgeraniol. Chem. Biol. Interact. 2011, 191, 261–268. [Google Scholar] [CrossRef] [Green Version]
  33. Caple, M.B.; Chow, H.C.; Strouse, C.E. Photosynthetic pigments of green sulfur bacteria. The esterifying alcohols of bacteriochlorophylls c from Chlorobium limicola. J. Biol. Chem. 1978, 253, 6730–6737. [Google Scholar] [CrossRef]
  34. Huchelmann, A.; Brahim, M.S.; Gerber, E.; Tritsch, D.; Bach, T.J.; Hemmerlin, A. Farnesol-mediated shift in the metabolic origin of prenyl groups used for protein prenylation in plants. Biochimie 2016, 127, 95–102. [Google Scholar] [CrossRef]
  35. Malizia, R.A.; Cardell, D.A.; Molli, J.S.; Grau, R.J. Volatile constituents of Acacia caven (Mol.) Mol. flower concrete from species growing in Argentina. J. Essent. Oil Res. 2002, 14, 132–134. [Google Scholar] [CrossRef]
  36. Silva, E.K.; Zabot, G.L.; Meireles, M.A.A. Ultrasound-assisted encapsulation of annatto seed oil: Retention and release of a bioactive compound with functional activities. Food Res. Int. 2015, 78, 159–168. [Google Scholar] [CrossRef]
  37. Chaberlain, W.J.; Severson, R.F.; Chortyk, O.T.; Sisson, V.E. Determination of solanesol in tobacco by capillary gas chromatography. J. Chromatogr. A 1990, 513, 55–60. [Google Scholar] [CrossRef]
  38. Vanaga, I.; Gubernator, J.; Nakurte, I.; Kletnieks, U.; Muceniece, R.; Jansone, B. Identification of Abies sibirica L. polyprenols and characterisation of polyprenol-containing liposomes. Molecules 2020, 25, 1801. [Google Scholar] [CrossRef] [Green Version]
  39. Surmacz, L.; Swiezewska, E. Polyisoprenoids–secondary metabolites or physiologically important superlipids? Biochem. Biophys. Res. Commun. 2011, 407, 627–632. [Google Scholar] [CrossRef]
  40. Yagi, K.J.; Konz, K.G.; Stay, B.; Tobe, S.S. Production and utilization of farnesoic acid in the juvenile hormone biosynthetic pathway by corpora allata of larval Diploptera punctata. Gen. Comp. Endocrinol. 1991, 81, 284–294. [Google Scholar] [CrossRef]
  41. Wierzbicki, A.S.; Mayne, P.D.; Lloyd, M.D.; Burston, D.; Mei, G.; Sidey, M.C.; Gibberd, F.B. Metabolism of phytanic acid and 3-methyl-adipic acid excretion in patients with adult Refsum disease. J. Lipid Res. 2003, 44, 1481–1488. [Google Scholar] [CrossRef]
  42. Endo, S.; Matsunaga, T.; Mamiya, H.; Ohta, C.; Soda, M.; Kitade, Y.; Tajima, K.; Zhao, H.T.; El-Kabbani, O.; Hara, A. Kinetic studies of AKR1B10, human aldose reductase-like protein: Endogenous substrates and inhibition by steroids. Arch. Biochem. 2009, 487, 1–9. [Google Scholar] [CrossRef]
  43. Meigs, T.E.; Roseman, D.S.; Simoni, R.D. Regulation of 3-hydroxy-3-methylglutaryl-coenzyme A reductase degradation by the nonsterol mevalonate metabolite farnesol in vivo. J. Biol. Chem. 1996, 271, 7916–7922. [Google Scholar] [CrossRef] [Green Version]
  44. Fernandes, N.V.; Yeganehjoo, H.; Katuru, R.; DeBose-Boyd, R.A.; Morris, L.L.; Michon, R.; Mo, H. Geranylgeraniol suppresses the viability of human DU145 prostate carcinoma cells and the level of HMG CoA reductase. Exp. Biol. Med. 2013, 238, 1265–1274. [Google Scholar] [CrossRef]
  45. Miriyala, S.; Subramanian, T.; Panchatcharam, M.; Ren, H.; McDermott, M.I.; Sunkara, M.; Morris, A.J. Functional characterization of the atypical integral membrane lipid phosphatase PDP1/PPAPDC2 identifies a pathway for interconversion of isoprenols and isoprenoid phosphates in mammalian cells. J. Biol. Chem. 2010, 285, 13918–13929. [Google Scholar] [CrossRef] [Green Version]
  46. Elsabrouty, R.; Jo, Y.; Hwang, S.; Jun, D.-J.; DeBose-Boyd, R.A. Type 1 polyisoprenoid diphosphate phosphatase modulates geranylgeranyl-mediated control of HMG CoA reductase and UBIAD1. Elife 2021, 10, e64688. [Google Scholar] [CrossRef]
  47. Bansal, V.S.; Vaidya, S. Characterization of 2 Distinct Allyl Pyrophosphatase Activities from Rat-Liver Microsomes. Arch. Biochem. Biophys. 1994, 315, 393–3994. [Google Scholar] [CrossRef]
  48. Goodman, D.S.; Popjak, G. Studies on the biosynthesis of cholesterol: XII. synthesis of allyl pyrophosphates from mevalonate and their conversion into squalene with liver enzymes. J. Lipid Res. 1960, 1, 286–300. [Google Scholar] [CrossRef]
  49. Nualkaew, N.; De-Eknamkul, W.; Kutchan, T.M.; Zenk, M.H. Membrane-bound geranylgeranyl diphosphate phosphatases: Purification and characterization from Croton stellatopilosus leaves. Phytochemistry 2006, 67, 1613–1620. [Google Scholar] [CrossRef]
  50. Nyati, P.; Nouzova, M.; Rivera-Perez, C.; Clifton, M.E.; Mayoral, J.G.; Noriega, F.G. Farnesyl phosphatase, a Corpora allata enzyme involved in juvenile hormone biosynthesis in Aedes aegypti. PLoS ONE 2013, 8, e71967. [Google Scholar] [CrossRef] [Green Version]
  51. de Wolf, E.; Abdullah, M.I.; Jones, S.M.; Menezes, K.; Moss, D.M.; Drijfhout, F.P.; Richardson, A. Dietary geranylgeraniol can limit the activity of pitavastatin as a potential treatment for drug-resistant ovarian cancer. Sci. Rep. 2017, 7, 1–10. [Google Scholar]
  52. Jawad, M.J.; Ibrahim, S.; Kumar, M.; Burgert, C.; Li, W.W.; Richardson, A. Identification of foods that affect the anti-cancer activity of pitavastatin in cells. Oncol. Lett. 2022, 23, 1–8. [Google Scholar] [CrossRef]
  53. Popjak, G.; Edmond, J.; Clifford, K.; Williams, V. Biosynthesis and structure of a new intermediate between farnesyl pyrophosphate and squalene. J. Biol. Chem. 1969, 244, 1897–1918. [Google Scholar] [CrossRef]
  54. Togashi, N.; Inoue, Y.; Hamashima, H.; Takano, A. Effects of two terpene alcohols on the antibacterial activity and the mode of action of farnesol against Staphylococcus aureus. Molecules 2008, 13, 3069–3076. [Google Scholar] [CrossRef] [Green Version]
  55. Burke, Y.D.; Stark, M.J.; Roach, S.L.; Sen, S.E.; Crowell, P.L. Inhibition of pancreatic cancer growth by the dietary isoprenoids farnesol and geraniol. Lipids 1997, 32, 151. [Google Scholar] [CrossRef]
  56. Jung, Y.Y.; Hwang, S.T.; Sethi, G.; Fan, L.; Arfuso, F.; Ahn, K.S. Potential anti-inflammatory and anti-cancer properties of farnesol. Molecules 2018, 23, 2827. [Google Scholar] [CrossRef] [Green Version]
  57. Kotti, T.J.; Ramirez, D.M.; Pfeiffer, B.E.; Huber, K.M.; Russell, D.W. Brain cholesterol turnover required for geranylgeraniol production and learning in mice. Proc. Natl. Acad. Sci. USA 2006, 103, 3869–3874. [Google Scholar] [CrossRef] [Green Version]
  58. Kotti, T.; Head, D.D.; McKenna, C.E.; Russell, D.W. Biphasic requirement for geranylgeraniol in hippocampal long-term potentiation. Proc. Natl. Acad. Sci. USA 2008, 105, 11394–11399. [Google Scholar] [CrossRef] [Green Version]
  59. Ho, H.J.; Shirakawa, H.; Giriwono, P.E.; Ito, A.; Komai, M. A novel function of geranylgeraniol in regulating testosterone production. Biosci. Biotechnol. Biochem. 2018, 82, 956–962. [Google Scholar] [CrossRef] [Green Version]
  60. Irwin, J.C.; Fenning, A.S.; Vella, R.K. Geranylgeraniol prevents statin-induced skeletal muscle fatigue without causing adverse effects in cardiac or vascular smooth muscle performance. Transl. Res. 2020, 215, 17–30. [Google Scholar] [CrossRef] [Green Version]
  61. Rattanawonsakul, K.; Bullock, G.; Bolt, R.; Claeyssens, F.; Atkins, S.; Hearnden, V. In vitro Effect of Geranylgeraniol (GGOH) on Bisphosphonate-Induced Cytotoxicity of Oral Mucosa Cells. Front. Oral Health 2022, 3, 892615. [Google Scholar] [CrossRef] [PubMed]
  62. Zafar, S.; Coates, D.E.; Cullinan, M.P.; Drummond, B.K.; Milne, T.; Seymour, G.J. Zoledronic acid and geranylgeraniol regulate cellular behaviour and angiogenic gene expression in human gingival fibroblasts. J. Oral Pathol. Med. 2014, 43, 711–721. [Google Scholar] [CrossRef] [PubMed]
  63. Marcuzzi, A.; Pontillo, A.; Leo, L.D.; Tommasini, A.; Decorti, G.; Not, T.; Ventura, A. Natural isoprenoids are able to reduce inflammation in a mouse model of mevalonate kinase deficiency. Pediatr. Res. 2008, 64, 177–182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Marcuzzi, A.; Piscianz, E.; Zweyer, M.; Bortul, R.; Loganes, C.; Girardelli, M.; Celeghini, C. Geranylgeraniol and neurological impairment: Involvement of apoptosis and mitochondrial morphology. Int. J. Mol. Sci. 2016, 17, 365. [Google Scholar] [CrossRef] [Green Version]
  65. Kim, J.; Lee, J.N.; Ye, J.; Hao, R.; DeBose-Boyd, R.; Ye, J. Sufficient production of geranylgeraniol is required to maintain endotoxin tolerance in macrophages. J. Lipid Res. 2013, 54, 3430–3437. [Google Scholar] [CrossRef] [Green Version]
  66. Campia, I.; Lussiana, C.; Pescarmona, G.; Ghigo, D.; Bosia, A.; Riganti, C. Geranylgeraniol prevents the cytotoxic effects of mevastatin in THP-1 cells, without decreasing the beneficial effects on cholesterol synthesis. Br. J. Pharmacol. 2009, 158, 1777–1786. [Google Scholar] [CrossRef] [Green Version]
  67. Crick, D.C.; Waechter, C.J.; Andres, D.A. Utilization of geranylgeraniol for protein isoprenylation in C6 glial cells. Biochem. Biophys. Res. Commun. 1994, 205, 955–961. [Google Scholar] [CrossRef]
  68. Andres, D.A.; Crick, D.C.; Finlin, B.S.; Waechter, C.J. Rapid Identification of Cysteine-Linked Isoprenyl Groups by Metabolic Labeling with [3H] Farnesol and [3H] Geranylgeraniol. Protein Lipidation Protoc. 1998, 116, 107–123. [Google Scholar]
  69. Moura, I.C.; Wunderlich, G.; Uhrig, M.L.; Couto, A.S.; Peres, V.J.; Katzin, A.M.; Kimura, E.A. Limonene arrests parasite development and inhibits isoprenylation of proteins in Plasmodium falciparum. Antimicrob. Agents Chemother. 2001, 45, 2553–2558. [Google Scholar] [CrossRef] [Green Version]
  70. Thai, L.; Rush, J.S.; Maul, J.E.; Devarenne, T.; Rodgers, D.L.; Chappell, J.; Waechter, C.J. Farnesol is utilized for isoprenoid biosynthesis in plant cells via farnesyl pyrophosphate formed by successive monophosphorylation reactions. Proc. Natl. Acad. Sci. USA 1999, 96, 13080–13085. [Google Scholar] [CrossRef] [Green Version]
  71. Crick, D.C.; Andres, D.A.; Waechter, C.J. Farnesol is utilized for protein isoprenylation and the biosynthesis of cholesterol in mammalian cells. Biochem. Biophys. Res. Commun. 1995, 211, 590–599. [Google Scholar] [CrossRef] [PubMed]
  72. Crick, D.C.; Andres, D.A.; Waechter, C.J. Novel salvage pathway utilizing farnesol and geranylgeraniol for protein isoprenylation. Biochem. Biophys. Res. Commun. 1997, 237, 483–487. [Google Scholar] [CrossRef] [PubMed]
  73. Hartmann, M.A.; Bach, T.J. Incorporation of all-trans-farnesol into sterols and ubiquinone in Nicotiana tabacum L. cv Bright Yellow-2 cell cultures. Tetrahedron Lett. 2001, 42, 655–657. [Google Scholar] [CrossRef]
  74. Rodrigues Goulart, H.; Kimura, E.A.; Peres, V.J.; Couto, A.S.; Aquino Duarte, F.A.; Katzin, A.M. Terpenes arrest parasite development and inhibit biosynthesis of isoprenoids in Plasmodium falciparum. Antimicrob. Agents Chemother. 2004, 48, 2502–2509. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Fliesler, S.J.; Keller, R.K. Metabolism of [3H] farnesol to cholesterol and cholesterogenic intermediates in the living rat eye. Biochem. Biophys. Res. Commun. 1995, 210, 695–702. [Google Scholar] [CrossRef]
  76. Callegari, S.; McKinnon, R.A.; Andrews, S.; de Barros Lopes, M.A. Atorvastatin-induced cell toxicity in yeast is linked to disruption of protein isoprenylation. FEMS Yeast Res. 2010, 10, 188–198. [Google Scholar] [CrossRef] [PubMed]
  77. Kennedy, K.; Cobbold, S.A.; Hanssen, E.; Birnbaum, J.; Spillman, N.J.; McHugh, E.; Ralph, S.A. Delayed death in the malaria parasite Plasmodium falciparum is caused by disruption of prenylation-dependent intracellular trafficking. PLoS Biol. 2019, 17, e3000376. [Google Scholar] [CrossRef] [Green Version]
  78. Crick, D.C.; Andres, D.A.; Waechter, C.J. Geranylgeraniol promotes entry of UT-2 cells into the cell cycle in the absence of mevalonate. Exp. Cell Res. 1997, 231, 302–307. [Google Scholar] [CrossRef]
  79. Hartley, M.D.; Schneggenburger, P.E.; Imperiali, B. Lipid bilayer nanodisc platform for investigating polyprenol-dependent enzyme interactions and activities. Proc. Natl. Acad. Sci. USA 2013, 110, 20863–20870. [Google Scholar] [CrossRef] [Green Version]
  80. Inoue, H.; Korenaga, T.; Sagami, H.; Koyama, T.; Ogura, K. Phosphorylation of farnesol by a cell-free system from Botryococcus braunii. Biochem. Biophys. Res. Commun. 1994, 200, 1036–1041. [Google Scholar] [CrossRef]
  81. Ohnuma, S.I.; Watanabe, M.; Nishino, T. Identification and characterization of geranylgeraniol kinase and geranylgeranyl phosphate kinase from the Archaebacterium Sulfolobus acidocaldariu. J. Biochem. 1996, 119, 541–547. [Google Scholar] [CrossRef] [PubMed]
  82. Bentinger, M.; Grünler, J.; Peterson, E.; Swiezewska, E.; Dallner, G. Phosphorylation of farnesol in rat liver microsomes: Properties of farnesol kinase and farnesyl phosphate kinase. Arch. Biochem. Biophys. 1998, 353, 191–198. [Google Scholar] [CrossRef] [PubMed]
  83. Westfall, D.; Aboushadi, N.; Shackelford, J.E.; Krisans, S.K. Metabolism of farnesol: Phosphorylation of farnesol by rat liver microsomal and peroxisomal fractions. Biochem. Biophys. Res. Commun. 1997, 230, 562–568. [Google Scholar] [CrossRef]
  84. Ischebeck, T.; Zbierzak, A.M.; Kanwischer, M.; Dörmann, P. A salvage pathway for phytol metabolism in Arabidopsis. J. Biol. Chem. 2006, 281, 2470–2477. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Sussmann, R.A.; Gabriel, H.B.; Ríos, A.G.; Menchaca Vega, D.S.; Yamaguchi, L.F.; Doménech-Carbó, A.; Katzin, A.M. Presence of Phylloquinone in the Intraerythrocytic Stages of Plasmodium falciparum. Front. Cell. Infect. Microbiol. 2022, 12, 434. [Google Scholar] [CrossRef] [PubMed]
  86. Almeida, J.; Azevedo, M.; Spicher, L.; Glauser, G.; vom Dorp, K.; Guyer, L.; del Valle Carranza, A.; Asis, R.; de Souza, A.P.; Buckeridge, M.; et al. Down-regulation of tomato PHYTOL KINASE strongly impairs tocopherol biosynthesis and affects prenyllipid metabolism in an organ-specific manner. J. Exp. Bot. 2016, 67, 919–934. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Valentin, H.E.; Lincoln, K.; Moshiri, F.; Jensen, P.K.; Qi, Q.; Venkatesh, T.V.; Last, R.L. The Arabidopsis vitamin E pathway gene5-1 mutant reveals a critical role for phytol kinase in seed tocopherol biosynthesis. Plant Cell 2006, 18, 212–224. [Google Scholar] [CrossRef] [Green Version]
  88. Fitzpatrick, A.H.; Bhandari, J.; Crowell, D.N. Farnesol kinase is involved in farnesol metabolism, ABA signaling and flower development in Arabidopsis. Plant J. 2011, 66, 1078–1088. [Google Scholar] [CrossRef]
  89. Spicher, L.; Almeida, J.; Gutbrod, K.; Pipitone, R.; Dörmann, P.; Glauser, G.; Kessler, F. Essential role for phytol kinase and tocopherol in tolerance to combined light and temperature stress in tomato. J. Exp. Bot. 2017, 68, 5845–5856. [Google Scholar] [CrossRef] [Green Version]
  90. Vom Dorp, K.; Hölzl, G.; Plohmann, C.; Eisenhut, M.; Abraham, M.; Weber, A.P.; Dörmann, P. Remobilization of phytol from chlorophyll degradation is essential for tocopherol synthesis and growth of Arabidopsis. Plant Cell 2015, 27, 2846–2859. [Google Scholar] [CrossRef] [Green Version]
  91. Seaver, S.M.; Gerdes, S.; Frelin, O.; Lerma-Ortiz, C.; Bradbury, L.M.; Zallot, R.; Hasnain, G.; Niehaus, T.D.; El Yacoubi, B.; Pasternak, S.; et al. High-throughput comparison, functional annotation, and metabolic modeling of plant genomes using the PlantSEED resource. Proc. Natl. Acad. Sci. USA 2014, 111, 9645–9650. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Chin, K.Y.; Ekeuku, S.O.; Trias, A. The Role of Geranylgeraniol in Managing Bisphosphonate-Related Osteonecrosis of the Jaw. Front. Pharmacol. 2022, 13, 878556. [Google Scholar] [CrossRef] [PubMed]
  93. Ivanova, A.; Ivanova, K.; Fiandra, L.; Mantecca, P.; Catelani, T.; Natan, M.; Banin, E.; Jacobi, G.; Tzanov, T. Antibacterial, Antibiofilm, and Antiviral Farnesol-Containing Nanoparticles Prevent Staphylococcus aureus from Drug Resistance Development. Int. J. Mol. Sci. 2022, 23, 7527. [Google Scholar] [CrossRef] [PubMed]
  94. Mo, H.; Jeter, R.; Bachmann, A.; Yount, S.T.; Shen, C.L.; Yeganehjoo, H. The Potential of Isoprenoids in Adjuvant Cancer Therapy to Reduce Adverse Effects of Statins. Front. Pharmacol. 2019, 9, 1515. [Google Scholar] [CrossRef] [Green Version]
  95. Prior, I.A.; Lewis, P.D.; Mattos, C. A comprehensive survey of Ras mutations in cancer. Cancer Res. 2012, 72, 2457–2467. [Google Scholar] [CrossRef] [Green Version]
  96. Healy, F.M.; Prior, I.A.; MacEwan, D.J. The importance of Ras in drug resistance in cancer. Br. J. Pharmacol. 2022, 179, 2844–2867. [Google Scholar] [CrossRef]
  97. Berndt, N.; Hamilton, A.D.; Sebti, S.M. Targeting protein prenylation for cancer therapy. Nat. Rev. Cancer 2011, 11, 775. [Google Scholar] [CrossRef] [Green Version]
  98. Marcuzzi, A.; Zanin, V.; Piscianz, E.; Tricarico, P.M.; Vuch, J.; Girardelli, M.; Crovella, S. Lovastatin-induced apoptosis is modulated by geranylgeraniol in a neuroblastoma cell line. Int. J. Dev. Neurosci. 2012, 30, 451–456. [Google Scholar] [CrossRef]
  99. Matusewicz, L.; Meissner, J.; Toporkiewicz, M.; Sikorski, A.F. The effect of statins on cancer cells. Tumor Biol. 2015, 36, 4889–4904. [Google Scholar] [CrossRef]
  100. Elson, C.E.; Yu, S.G. The chemoprevention of cancer by mevalonate-derived constituents of fruits and vegetables. J Nutr. 1994, 124, 607–614. [Google Scholar] [CrossRef]
  101. Yilmaz Öztürk, B.; Feyzullazade, N.; Dağ, İ.; Şengel, T. The investigation of in vitro effects of farnesol at different cancer cell lines. Microsc. Res. Tech. 2022, 85, 2760–2775. [Google Scholar] [CrossRef] [PubMed]
  102. Balaraman, G.; Sundaram, J.; Mari, A.; Krishnan, P.; Salam, S.; Subramaniam, N.; Thiruvengadam, D. Farnesol alleviates diethyl nitrosamine induced inflammation and protects experimental rat hepatocellular carcinoma. Environ. Toxicol. 2021, 36, 2467–2474. [Google Scholar] [CrossRef] [PubMed]
  103. Dinesh, N.; Pallerla, D.S.; Kaur, P.K.; Kishore Babu, N.; Singh, S. Leishmania donovani 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGR) as a potential drug target by biochemical, biophysical and inhibition studies. Microb. Pathog. 2014, 66, 14–23. [Google Scholar] [CrossRef] [PubMed]
  104. Parihar, S.P.; Guler, R.; Brombacher, F. Statins: A viable candidate for host-directed therapy against infectious diseases. Nat. Rev. Immunol. 2019, 19, 104–117. [Google Scholar] [CrossRef] [PubMed]
  105. Balanco, J.M.F.; Sussmann, R.A.; Verdaguer, I.B.; Gabriel, H.B.; Kimura, E.A.; Katzin, A.M. Tocopherol biosynthesis in Leishmania (L.) amazonensis promastigotes. FEBS Open Bio. 2019, 9, 743–754. [Google Scholar] [CrossRef] [Green Version]
  106. Gabriel, H.; Sussmann, R.; Kimura, E.; Marin Rodriguez, A.; Bofill Verdaguer, I.; Leite, G.; Katzin, A. Terpenes as Potential Antimalarial Drugs; Intechopen: London, UK, 2018. [Google Scholar]
  107. Porta, E.O.; Verdaguer, I.B.; Perez, C.; Banchio, C.; de Azevedo, M.F.; Katzin, A.M.; Labadie, G.R. Repositioning Salirasib as a new antimalarial agent. MedChemComm 2019, 10, 1599–1605. [Google Scholar] [CrossRef]
  108. Verdaguer, I.B.; Crispim, M.; Zafra, C.A.; Sussmann RA, C.; Buriticá, N.L.; Melo, H.R.; Katzin, A.M. Exploring ubiquinone biosynthesis inhibition as a strategy for improving atovaquone efficacy in malaria. Antimicrob. Agents Chemother. 2021, 65, e01516–e01520. [Google Scholar] [CrossRef]
  109. Li, Z.H.; Ramakrishnan, S.; Striepen, B.; Moreno, S.N. Toxoplasma gondii relies on both host and parasite isoprenoids and can be rendered sensitive to atorvastatin. PLoS Pathog. 2013, 9, e1003665. [Google Scholar] [CrossRef] [Green Version]
  110. Wang, S.; Li, M.; Luo, X.; Yu, L.; Nie, Z.; Liu, Q.; He, L. Inhibitory effects of fosmidomycin against Babesia microti in vitro. Front. Cell Dev. Biol. 2020, 8, 247. [Google Scholar] [CrossRef]
  111. Brown, A.C.; Parish, T. Dxr is essential in Mycobacterium tuberculosis and fosmidomycin resistance is due to a lack of uptake. BMC Microbiol. 2008, 8, 1–9. [Google Scholar] [CrossRef] [Green Version]
  112. Fernandes, J.F.; Lell, B.; Agnandji, S.T.; Obiang, R.M.; Bassat, Q.; Kremsner, P.G.; Grobusch, M.P. Fosmidomycin as an antimalarial drug: A meta-analysis of clinical trials. Future Microbiol. 2015, 10, 1375–1390. [Google Scholar] [CrossRef] [PubMed]
  113. Zhang, B.; Watts, K.M.; Hodge, D.; Kemp, L.M.; Hunstad, D.A.; Hicks, L.M.; Odom, A.R. A second target of the antimalarial and antibacterial agent fosmidomycin revealed by cellular metabolic profiling. Biochemistry 2011, 50, 3570–3577. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. He, L.; He, P.; Luo, X.; Li, M.; Yu, L.; Guo, J.; Zhao, J. The MEP pathway in Babesia orientalis apicoplast, a potential target for anti-babesiosis drug development. Parasites Vectors 2018, 11, 1–8. [Google Scholar] [CrossRef] [PubMed]
  115. McConnell, M.; Shieh, A. Polypharmacy in Osteoporosis Treatment. Clin. Geriatr. Med. 2022, 38, 715–726. [Google Scholar] [CrossRef] [PubMed]
  116. Ziebart, T.; Koch, F.; Klein, M.O.; Guth, J.; Adler, J.; Pabst, A.; Walter, C. Geranylgeraniol—A new potential therapeutic approach to bisphosphonate associated osteonecrosis of the jaw. Oral Oncol. 2011, 47, 195–201. [Google Scholar] [CrossRef]
  117. Koneski, F.; Popovic-Monevska, D.; Gjorgoski, I.; Krajoska, J.; Popovska, M.; Muratovska, I.; Popovski, V. In vivo effects of geranylgeraniol on the development of bisphosphonate-related osteonecrosis of the jaws. J. Cranio-Maxillofac. Surg. 2018, 46, 230–236. [Google Scholar] [CrossRef]
  118. Golomb, B.A.; Evans, M.A. Statin adverse effects. Am. J. Cardiovasc. Drugs 2008, 8, 373–418. [Google Scholar] [CrossRef]
  119. Batista, M.A.; de Lima Teixeira dos Santos, A.V.T.; do Nascimento, A.L.; Moreira, L.F.; Souza IR, S.; da Silva, H.R.; Carvalho, J.C.T. Potential of the Compounds from Bixa orellana Purified Annatto Oil and Its Granules (Chronic®) against Dyslipidemia and Inflammatory Diseases: In Silico Studies with Geranylgeraniol and Tocotrienols. Molecules 2022, 27, 1584. [Google Scholar] [CrossRef]
  120. Jaśkiewicz, A.; Pająk, B.; Litwiniuk, A.; Urbańska, K.; Orzechowski, A. Geranylgeraniol prevents statin-dependent myotoxicity in C2C12 muscle cells through RAP1 GTPase prenylation and cytoprotective autophagy. Oxidative Med. Cell. Longev. 2018, 2018, 6463807. [Google Scholar] [CrossRef] [Green Version]
  121. Zhang, Y.; Åberg, F.; Appelkvist, E.L.; Dallner, G.; Ernster, L. Uptake of dietary coenzyme Q supplement is limited in rats. J. Nutr. 1995, 125, 446–453. [Google Scholar]
  122. Young, J.M.; Florkowski, C.M.; Molyneux, S.L.; McEwan, R.G.; Frampton, C.M.; George, P.M.; Scott, R.S. Effect of coenzyme Q10 supplementation on simvastatin-induced myalgia. Am. J. Cardiol. 2007, 100, 1400–1403. [Google Scholar] [CrossRef] [PubMed]
  123. Wang, L.; Zheng, Z.; Zhu, L.; Meng, L.; Liu, H.; Wang, K.; Chen, J.; Li, P.; Yang, H. Geranylgeranyl pyrophosphate depletion by statins compromises skeletal muscle insulin sensitivity. J. Cachexia Sarcopenia Muscle 2022. advance online publication. [Google Scholar] [CrossRef] [PubMed]
  124. Wang, L.; Zhu, L.; Zheng, Z.; Meng, L.; Liu, H.; Wang, K.; Chen, J.; Li, P.; Yang, H. Mevalonate pathway orchestrates insulin signaling via RAB14 geranylgeranylation-mediated phosphorylation of AKT to regulate hepatic glucose metabolism. Metab. Clin. Exp. 2022, 128, 155120. [Google Scholar] [CrossRef] [PubMed]
  125. Abukhalil, M.H.; Hussein, O.E.; Bin-Jumah, M.; Saghir, S.A.; Germoush, M.O.; Elgebaly, H.A.; Mahmoud, A.M. Farnesol attenuates oxidative stress and liver injury and modulates fatty acid synthase and acetyl-CoA carboxylase in high cholesterol-fed rats. Environ. Sci. Pollut. Res. 2020, 27, 30118–30132. [Google Scholar] [CrossRef] [PubMed]
  126. Singh, S.P.; Sashidhara, K.V. Lipid lowering agents of natural origin: An account of some promising chemotypes. Eur. J. Med. Chem. 2017, 140, 331–348. [Google Scholar] [CrossRef]
  127. Favier, L.A.; Schulert, G.S. Mevalonate kinase deficiency: Current perspectives. Appl. Clin. Genet. 2016, 9, 101. [Google Scholar]
  128. Frenkel, J.; Rijkers, G.T.; Mandey, S.H.; Buurman, S.W.; Houten, S.M.; Wanders, R.J.; Waterham, H.R.; Kuis, W. Lack of isoprenoid products raises ex vivo interleukin-1beta secretion in hyperimmunoglobulinemia D and periodic fever syndrome. Arthritis Rheum. 2002, 46, 2794–2803. [Google Scholar] [CrossRef]
  129. Jurczyluk, J.; Munoz, M.A.; Skinner, O.P.; Chai, R.C.; Ali, N.; Palendira, U.; Rogers, M.J. Mevalonate kinase deficiency leads to decreased prenylation of Rab GTPases. Immunol. Cell Biol. 2016, 94, 994–999. [Google Scholar] [CrossRef] [Green Version]
  130. Saputra, W.D.; Shono, H.; Ohsaki, Y.; Sultana, H.; Komai, M.; Shirakawa, H. Geranylgeraniol Inhibits Lipopolysaccharide-Induced Inflammation in Mouse-Derived MG6 Microglial Cells via NF-κB Signaling Modulation. Int. J. Mol. Sci. 2021, 22, 10543. [Google Scholar] [CrossRef]
  131. Giriwono, P.E.; Shirakawa, H.; Ohsaki, Y.; Hata, S.; Kuriyama, H.; Sato, S.; Komai, M. Dietary supplementation with geranylgeraniol suppresses lipopolysaccharide-induced inflammation via inhibition of nuclear factor-κB activation in rats. Eur. J. Nutr. 2013, 52, 1191–1199. [Google Scholar] [CrossRef]
  132. Souza, D.S.; Barreto, T.O.; Menezes-Filho, J.; Heimfarth, L.; Rhana, P.; Rabelo, T.K.; Santana, M.; Durço, A.O.; Conceição, M.; Quintans-Júnior, L.J.; et al. Myocardial hypertrophy is prevented by farnesol through oxidative stress and ERK1/2 signaling pathways. Eur. J. Pharmacol. 2020, 887, 173583. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Isoprenoid biosynthesis in cells. A simplified view of isoprenoid biosynthesis pathways and the targets of different drugs. Although shown as a general situation, some parts may not be present in all organisms. For example, the MVA pathway (light blue lines) is present in mammals and the cytosol of plants; the MEP pathway (green lines) is found in the plastid of plants and apicomplexan parasites, but not in mammals; the geranylgeraniol and farnesol salvage pathway (brown lines) is not present in Saccharomyces cerevisiae or Escherichia coli. Continuous arrows indicate a single enzymatic step, discontinuous arrows indicate multiple steps and blue arrows/text refers to processes of biomedical importance or diseases. Red arrows indicate a drug inhibition or metabolic downregulation. The chemical structures of FOH and GGOH are also shown. Abbreviations are as in the text.
Figure 1. Isoprenoid biosynthesis in cells. A simplified view of isoprenoid biosynthesis pathways and the targets of different drugs. Although shown as a general situation, some parts may not be present in all organisms. For example, the MVA pathway (light blue lines) is present in mammals and the cytosol of plants; the MEP pathway (green lines) is found in the plastid of plants and apicomplexan parasites, but not in mammals; the geranylgeraniol and farnesol salvage pathway (brown lines) is not present in Saccharomyces cerevisiae or Escherichia coli. Continuous arrows indicate a single enzymatic step, discontinuous arrows indicate multiple steps and blue arrows/text refers to processes of biomedical importance or diseases. Red arrows indicate a drug inhibition or metabolic downregulation. The chemical structures of FOH and GGOH are also shown. Abbreviations are as in the text.
Molecules 27 08691 g001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Verdaguer, I.B.; Crispim, M.; Hernández, A.; Katzin, A.M. The Biomedical Importance of the Missing Pathway for Farnesol and Geranylgeraniol Salvage. Molecules 2022, 27, 8691. https://doi.org/10.3390/molecules27248691

AMA Style

Verdaguer IB, Crispim M, Hernández A, Katzin AM. The Biomedical Importance of the Missing Pathway for Farnesol and Geranylgeraniol Salvage. Molecules. 2022; 27(24):8691. https://doi.org/10.3390/molecules27248691

Chicago/Turabian Style

Verdaguer, Ignasi Bofill, Marcell Crispim, Agustín Hernández, and Alejandro Miguel Katzin. 2022. "The Biomedical Importance of the Missing Pathway for Farnesol and Geranylgeraniol Salvage" Molecules 27, no. 24: 8691. https://doi.org/10.3390/molecules27248691

Article Metrics

Back to TopTop