12 The Isoprenoid Biosynthetic Pathway and Statins SARAH A. HOLSTEIN Department of Internal Medicine University of Iowa Iowa City, Iowa, USA
I.
Abstract
The isoprenoid biosynthetic pathway (IBP) is the source of a vast array of isoprenoids. This pathway is highly conserved and isoprenoids play key roles throughout all forms of life. The discovery and development of the statins, a class of drugs which inhibit the rate-limiting step in the mevalonate-dependent IBP, has led to improved scientific understanding of the complex regulation of the pathway and has provided therapeutic agents which have had far-reaching effects on human health. Here, we provide an overview of the IBP and discuss the impact of pharmacological manipulation of the pathway by the statins.
II.
The Isoprenoid Biosynthetic Pathway
A. HISTORICAL OVERVIEW The IBP (Figure 12.1) is the source of over 23,000 naturally occurring isoprenoids [1]. The first chemical studies of isoprenoids were begun in the early 1800s (Figure 12.2). Between 1800 and 1884, a large number of compounds with the empirical formula of C5H8 were isolated. Otto Wallach, in a series of papers published between 1884 and 1887, was the first to THE ENZYMES, Vol. XXX # 2011 Elsevier Inc. All rights reserved.
279
ISSN NO: 1874-6047 DOI: 10.1016/B978-0-12-415922-8.00012-4
280
SARAH A. HOLSTEIN
Acetyl-CoA
HMG-CoA HMGR
Statins
Mevalonate Pyruvate + D-glyceraldehyde 3-phosphate DXPS
MK 5-Phosphomevalonate
1-Deoxy-D-xylulose 5-phosphate
IPP
DMAPP DMAPP
FDPS GPP
IPP
FDPS SQS Squalene
FTase
FPP
GGDPS
IPP
GGPP
GGTases
Farnesylated Proteins Geranylgeranylated Proteins
E-IDS Cholesterol
Ubiquinone
FIG. 12.1. The isoprenoid biosynthetic pathway. Intermediates and products are shown in black and enzymes in blue. Components of the mevalonate-independent DOXP pathway are shown in green. The HMGR inhibitors (statins) are shown in red.
propose what is now known as the ‘‘isoprene rule’’ [2]. He suggested that all terpenes could be built from isoprene (C5H8) units. In the 1950s, Leopold Ruzicka proposed the ‘‘biogenetic isoprene rule’’ which stated that all terpenes could be derived through cyclization or other rearrangement from a precursor composed of isoprene units [3]. In 1956, mevalonic acid was discovered and subsequent studies demonstrated that mevalonic acid could be incorporated into cholesterol, monoterpenes, rubber, and other terpenes. By 1957, the pathway for the formation of hydroxymethylglutaryl coenzyme A (HMG-CoA) from acetate was elucidated. Shortly thereafter, the connection between HMG-CoA and mevalonate was made when it was demonstrated that HMG-CoA could be enzymatically reduced to
Mevalonic acid discovered, found to be incorporated into cholesterol
1880s
Wallach’s Isoprene Rule
1956
HMGR activity identified as key enzyme
1959–1960
FPP and GGPP characterized
Brown & Goldstein demonstrate that treatment of cells with compactin results in upregulation of HMGR
1959–1967 1976
Discovery of compactin (mevastatin), the first HMGR inhibitor
1978
HMGR cloned
1980
Discovery of lovastatin
1982
Identification of protein farnesylation & geranylgeranylation in higher eukaryotes
FDA approval of lovastatin
1984
Discovery of posttranslational modification of proteins by a product of mevalonic acid
1985
1987
1989–1990
Brown & Goldstein win the Nobel Prize
FIG. 12.2. Timeline of milestones involving the isoprenoid biosynthetic pathway and statins.
Endo receives the Lasker Award
1998
Identification of the mevalonateindependent DOXP isoprenoid biosynthetic pathway
2008
282
SARAH A. HOLSTEIN
mevalonate. In 1959, Lynen and coworkers reported the characterization of the 5-carbon isopentenyl pyrophosphate (IPP) and the 15-carbon farnesyl pyrophosphate (FPP) [4]. Soon after, IPP was demonstrated to be an intermediate in the synthesis of squalene [5]. In 1960, Goodman and Popja´k identified dimethylallyl pyrophosphate (DMAPP) and geranyl pyrophosphate (GPP) as intermediates in the mevalonate-squalene pathway [6]. A year later, Grob et al. reported the synthesis of the 20-carbon geranylgeranyl pyrophosphate (GGPP) from FPP and IPP [7]. B. PRODUCTS OF THE IBP The IBP and its products are displayed in Figure 12.1. HMG-CoA, ultimately derived from acetyl-CoA is converted to mevalonate via the enzyme HMG-CoA reductase (HMGR) [8]. This reaction is the rate-limiting step in the pathway. Mevalonate is then phosphorylated via mevalonate kinase (MK) to yield 5-phosphomevalonate [9]. IPP is formed following additional phosphorylation and decarboxylation steps [10]. Isomerization of IPP via the enzyme IPP isomerase yields DMAPP [11]. In mammals, the enzyme farnesyl pyrophosphate synthase (FDPS) catalyzes the synthesis of both GPP and FPP [12]. In plants, a separate GPP synthase has been identified [13]. GPP is a key intermediate in plants as it serves as the precursor for all monoterpenes. In animals, however, GPP appears to serve only as an intermediate in the synthesis of FPP. Very low basal levels of GPP have been measured in cell culture, although cellular GPP levels can become markedly increased in the setting of FDPS inhibition [14]. FPP is necessary for the synthesis of both sterols and longer chain nonsterol isoprenoids. The first committed step in sterol synthesis is catalyzed by the enzyme squalene synthesis and involves the head-to-head condensation of two FPP molecules to form squalene [15]. This is followed by cyclization steps, leading to sterol synthesis. The addition of IPP to FPP via the enzyme GGPP synthase yields the 20-carbon GGPP [16]. FPP and GGPP are substrates in the prenylation reactions catalyzed by the enzymes farnesyl transferase (FTase) and geranylgeranyl transferase (GGTase) I and II [17–20]. Longer chain isoprenoids are synthesized via two other isoprenyl diphosphate enzyme systems in mammals [21]. Long E-isoprenyl diphosphate synthase (IDS) produces the side chains of ubiquinone. The length of the side chain varies amongst species, and in humans a C50 synthase has been identified [22]. Dehydrodolichyl diphosphate synthase, the only Z-IDS found in mammals, is responsible for the synthesis of the sugar carriers dolichol and dolichyl phosphate [23]. Plants have additional Z-IDS which can catalyze the production of very long isoprene species, such as natural
12. THE ISOPRENOID BIOSYNTHETIC PATHWAY AND STATINS
283
rubber which is composed of over 1000 isoprene units [24]. Most bacteria have FPP synthase, as well as both E- and Z-long IDS [25–27]. C. THE NONMEVALONATE-DEPENDENT IBP Plants and bacteria also have a nonmevalonate-dependent IBP, referred to as the deoxy-D-xylulose 5-phosphate (DOXP) pathway (Figure 12.1). The initial step in this pathway involves the condensation of pyruvate and D-glyceraldehyde-3-phosphate to form 1-deoxy-D-xylulose 5-phosphate in a reaction catalyzed by deoxyxylulose 5-phosphate synthase (DXPS) [28– 30]. Subsequent reactions lead to the synthesis of IPP [31,32]. There is compartmentalization of isoprenoid biosynthesis in higher plants, such that the mevalonate pathway produces sterols, sesquiterpenes, triterpenes, and polyterpenes in the cytosol while the DOXP pathway synthesizes monoterpenes, diterpenes, carotenoids, plastoquinones, and the prenyl side chain of chlorophyll in the plastid [33]. In bacteria, the DOXP pathway appears to be the most ancient pathway and is more common than the mevalonatedependent pathway [34]. The DOXP pathway has been identified in a variety of bacteria, mycobacteria, and algae [35–39] but not in fungi or yeasts [40].
III.
Statins
A. STATINS AS NATURAL PRODUCTS In 1976, Endo and coworkers isolated the first HMGR inhibitor, mevastatin (compactin) (Figure 12.3), from a culture of Penicillium citrinum [41]. During that same year researchers at Beecham Laboratories isolated mevastatin from Penicillium brevicompactum [42]. Mevastatin was found to potently inhibit HMGR in vitro with a Ki of 1.4 nM [43], to inhibit cholesterol synthesis in tissue culture cells [44,45], and to reduce plasma cholesterol levels in dogs [46], monkeys [47], and humans [48]. In 1980, lovastatin (mevinolin) was isolated from a strain of Aspergillus terreus [49]. HMGR inhibitors have subsequently been isolated from Pleurotus, Monascus, Paecilomyces, Trichoderma, Scopulariopsis, Doratomyces fungal genera as well as several yeast including Candida cariosilignicola and Pichia labacensis [50–53]. Lovastatin and mevastatin are synthesized via polyketide pathways. Polyketides are a large group of structurally diverse secondary metabolites produced by bacteria, fungi, and plants. The factors influencing production of lovastatin or mevastatin have not been fully elucidated. Studies of Aspergillus terreus grown in chemically defined media indicate that
284
SARAH A. HOLSTEIN O
HO O
O H3C
H
HO
H
O
O H3C
CH3
H
H
CH3
H3C
Mevastatin HO
O
O CH3 H
H3C
CO2Na OH
O H
H 3C
CH3
H
CH3
HO
H3C
Lovastatin
Simvastatin
Pravastatin F
CO2Na OH H
F
HO
O
HO
O
OH
O
N
OH
OH
CO2H
N
OH
N
O OH
N N F
Fluvastatin
SO2Me
Atorvastatin
Rosuvastatin F
F
OH
OH
O
OH OH
O
N
OH
O OH
N
N
Cerivastatin
Pitavastatin FIG. 12.3. Structures of statins.
lovastatin synthesis is initiated after glucose exhaustion and after cessation of lactose consumption, suggesting that lovastatin synthesis occurs in the setting of starvation conditions [54]. Two genes have been identified that may play roles in conferring resistance to compactin. One encodes a protein with significant homology to HMGR while the other appears to be an efflux pump [55]. A similar mechanism of self-resistance is found in Aspergillus terreus where the lvrA gene encodes a protein related to HMGR [56]. The nature of advantage provided by the production of HMGR inhibitors by select fungi is not well understood. Potential explanations include use as inhibitors of environmental competitors or enhancers of their own growth. Although mevastatin was initially detected by its antifungal activity [57], the magnitude of this effect was not published. Only 4 out of over 300 strains of yeast were found to be growth inhibited by compactin analogs [58]. As noted above, since bacteria predominantly use the mevalonate-independent pathway, it is less likely that they will be significantly affected by the statins. There has been some evidence to suggest that lovastatin can act as an herbicide, at
12. THE ISOPRENOID BIOSYNTHETIC PATHWAY AND STATINS
285
least with respect to radish seedling growth [59] and cell cultures of Solanum xanthocarpum [60]. Whether this applies more broadly to other plants is not known. It also is not clear that statins provide a direct growth advantage for the fungi from which they are derived. Mevinolin production was noted to reach its peak only after the dry weight of A. terreus had plateaued [53] and is dependent on the composition of the media [60]. Thus the reason for fungal HMGR inhibitor production remains to be determined. B. SYNTHETIC STATINS Following the success of lovastatin, a number of other statins were developed (Figure 12.3). Simvastatin, a semisynthetic derivative of lovastatin, was first approved for marketing in Sweden in 1988 and then later worldwide. Pravastatin, isolated from Nocardia autotropica, was approved in 1991. The purely synthetic statins fluvastatin (1994), atorvastatin (1997), cerivastatin (1998), rosuvastatin (2003), and pitavastatin (2009) soon followed. Cerivastatin was subsequently pulled from the market in 2001 because of postmarketing surveillance reports which revealed 52 deaths that were attributed to rhabdomyolysis and resulting renal failure [61]. C. PHARMACOLOGY OF STATINS All statins share an HMG-like moiety which is linked to rigid hydrophobic groups (Figure 12.3). Lovastatin and simvastatin are lactone prodrugs which are converted to the active open hydroxyl acid form in the liver. Enzyme studies show that the statins are competitive inhibitors of HMGR with respect to HMG-CoA and have Ki values in the 0.1–2.3 nM range [62]. Crystal structure studies have revealed that the statins occupy the active site where HMG-CoA binds but do not affect NADPH binding [63]. While statins do inhibit endogenous cholesterol biosynthesis, their hypocholesterolemic effect is secondary to increased clearance of LDL from the plasma due to upregulation of the hepatic LDL receptor [64,65]. Although the statins have differing potency, the maximal recommended dose of each statin can lead to a similar mean reduction in LDL cholesterol by 35–55%. The majority of the statins are metabolized by the cytochrome P450 system: lovastatin, simvastatin, and atorvastatin are substrates of CYP3A4 while fluvastatin and rosuvastatin are substrates of CYP2C9 [66]. Pravastatin and pitavastatin, however, are minimally metabolized by the cytochrome P450 system, and therefore have the potential for fewer drug–drug interactions [66,67]. There have been several reports suggesting activities of statins unrelated to HMGR inhibition. Rao et al. reported that the prodrug closed-ring form
286
SARAH A. HOLSTEIN
of lovastatin inhibited the proteasome [68]. Wojcik et al. also published studies demonstrating the ability of the closed-ring forms of lovastatin and simvastatin to inhibit proteasome activity, although the authors disagreed with Rao et al. regarding the effects on the chymotrypsin-like protease activity [69]. These two groups also presented conflicting results with regard to the ability of mevalonate to abrogate the effects of the statin prodrugs on proteasomal activity. Kumar et al. suggested that the open and closed-ring forms of mevastatin differed in their activity as neurotoxic or neuroprotective agents in a cell culture system [70]. Other investigators, however, have reported that these agents do not influence proteasomal activity [71–73]. At this time there are no data available to suggest that the putative effect of the closed-ring form of statins on proteasome activity is clinically meaningful. D. CLINICAL USE OF STATINS The statins represent some of the most-widely prescribed drugs in the United States and the world. Mevastatin was the first statin to be tested in humans. In a study involving 11 patients with primary hypercholesterolemia, serum cholesterol levels were reduced by approximately 30% following daily treatment for 4–8 weeks [48]. In 1987, lovastatin was the first statin to be approved for use in humans. Numerous trials have led to various statins being approved for multiple indications, including primary hypercholesterolemia, coronary heart disease, prophylaxis for patients with risk factors for coronary heart disease, prophylaxis for cerebrovascular accident, hypertriglyceridemia, and familial hypercholesterolemia. The majority of the clinical benefit of the statins has been attributed to their ability to lower LDL levels. However, there is increasing evidence that statins have pleiotropic effects in cardiovascular disease, including effects on endothelial function, atherosclerotic plaques, myocardial remodeling and vascular inflammation [74]. There has also been considerable interest in the use of statins in other clinical indications, including cancer [75], neurological disorders [76], osteoporosis [77], atrial fibrillation [78], asthma [79], angiogenesis [80], immunomodulatory effects [81], coagulation and thrombosis [82,83]. Whether these effects can all be attributed to the cholesterol-lowering activity or are a consequence of depletion of other isoprenoid species remains to be determined. E. STATINS, MYOPATHY, AND UBIQUINONE Although statins are generally well tolerated, some patients do develop myopathy. This can range from asymptomatic increases in creatinine kinase (CK) to renal failure from rhabdomyolysis. Risk factors include the dose of
12. THE ISOPRENOID BIOSYNTHETIC PATHWAY AND STATINS
287
statin, concomitant medications, age, and comorbid conditions [84]. The mechanism of action underlying the statins’ effects on muscle remains ill-defined; however, attention has focused on the role of ubiquinone (coenzyme Q). Ubiquinone is derived from GGPP (Figure 12.1) and plays an important role in the electron transport system in mitochondria. Studies in both animals and humans have demonstrated that statins decrease ubiquinone blood levels by as much as 50% [85–88]. It has been hypothesized that statin-induced decrease in ubiquinone results in mitochondrial dysfunction, causing myotoxicity. Supplementation of ubiquinone was able to restore plasma levels in patients taking atorvastatin, however this did not correlate with changes in the CK level [89]. It has also been argued that since ubiquinone is transported by LDL, that the observed decrease in serum/ plasma ubiquinone levels is simply a consequence of the statin-induced decrease in LDL levels and that tissue levels of ubiquinone may not be affected [90]. There have been conflicting reports in the literature as to whether tissue ubiquinone levels decrease following statin treatment in both animal and human studies [87,91–94]. Laaksonen et al. reported an increase in muscle ubiquinone levels after 1 or 6 months of statin treatment while Paiva et al. reported that simvastatin, but not atorvastatin decreased muscle levels [92–94]. Finally, trials evaluating ubiquinone supplementation have yielded equivocal results. Caso et al. reported that supplementation with ubiquinone, but not vitamin E, improved muscle pain symptoms while Young et al., reported that ubiquinone supplementation did not improve symptoms [95,96]. Further basic science and clinical studies are needed to determine both the mechanism of action and management of statin-induced myopathy. F. MK DEFICIENCY Although a number of genetic disorders associated with isoprenoid biosynthesis have been identified, the vast majority involve enzymes necessary for sterol synthesis. HMGR knock-out mice are embryonic lethal [97]. However, deficiency of MK activity results in two disorders: mevalonic aciduria (MA) and hyper-IgD and periodic fever syndrome (HIDS). Genetic analysis has revealed that the two diseases represent the same disorder, albeit with differing degrees of severity. MK enzyme activity is undetectable in the fibroblasts of MA patients, while in HIDS patients, activity of 1–7% of control can be found in fibroblasts and leukocytes [98–100]. Despite the nondetectable MK activity in MA fibroblasts, studies have shown that these cells are capable of synthesizing cholesterol from radiolabeled acetate [101,102]. In addition, near normal plasma levels of cholesterol have been found in MA patients [103]. Under control
288
SARAH A. HOLSTEIN
conditions, levels of prenylated Ras and RhoA proteins in MA and HIDS fibroblasts are similar to control fibroblasts under control conditions [103]. However, the patient cells are more sensitive to simvastatin such that accumulation of cytosolic Ras and RhoA occurs with lower concentrations of the HMGR inhibitor in the patient cells than in the control cells, consistent with the reduced ability of these cells to synthesize FPP and GGPP [103]. As it was hypothesized that excess mevalonate levels were responsible for the clinical features of the disease, two patients with MA were administered lovastatin. However, treatment was discontinued following worsening of their condition [103], suggesting that high mevalonate levels may not be the cause of the clinical phenotype and that high HMGR activity and mevalonate levels are required in order for these cells to maintain nonsterol synthesis.
IV.
Statins and the IBP
A. STATINS AND REGULATION OF THE IBP Statins have proven to be invaluable tools with which to study the regulation of HMGR, the IBP, and sterol homeostasis. In 1978, Brown and Goldstein used compactin to demonstrate that mevalonate depletion results in upregulation of HMGR [104]. The generation of a CHO-derived cell line (UT-1) selected for resistance to compactin and characterized by markedly increased levels of HMGR protein, aided in structural studies of HMGR as well as with the discovery that LDL and 25-hydroxycholesterol affect HMGR synthesis [105–107]. The roles for both sterol- and nonsterolmediated regulation of HMGR protein levels were investigated in studies involving statin-treated cells [108–110]. Studies involving the elucidation of other key regulators of the IBP and sterol homeostasis, including sterol regulatory element-binding proteins (SREBPs), SREBP cleavage-activating protein (SCAP), and Insigs, have also incorporated statins [111–114]. B. STATINS AND PRENYLATION Statins were also instrumental in the discovery of protein prenylation. In 1984, Glomset and coworkers used mevinolin (lovastatin) and radiolabeled mevalonate to demonstrate that a product of mevalonate could become posttranslationally incorporated into select proteins [115]. The use of the statin to deplete cells of endogenous mevalonate and downstream isoprenoids enabled sufficient incorporation of the exogenous radiolabeled mevalonate and subsequent detection of the radiolabeled protein fraction.
12. THE ISOPRENOID BIOSYNTHETIC PATHWAY AND STATINS
289
Further studies in the late 1980s revealed that proteins such as lamin B and Ras are farnesylated [116–118]. In the early 1990s, several groups identified geranylgeranylated proteins [119–121]. Similar labeling studies were performed using radiolabeled FPP or GGPP following statin-induced mevalonate depletion [122]. Innumerable studies have now been published utilizing statins to delineate the role of prenylation in modulating protein function. Statins, by virtue of their ability to deplete cells of all isoprenoid species downstream of mevalonate, including FPP and GGPP, globally diminish protein prenylation. Submicromolar doses of lovastatin can decrease intracellular FPP and GGPP levels in cultured cells [123]; however; under those conditions, disruption of protein prenylation is not detected. Work done in cultured cells has demonstrated that relative levels of FPP and GGPP vary amongst cell and tissue type, that disparate concentrations of statin are needed to lower FPP and GGPP levels by equivalent amounts, and that there is a hierarchy with respect to the conservation of prenylation of different prenylated proteins under conditions of mevalonate depletion [123–125] (R.J. Hohl, personal communication). Further studies are needed to better understand the relationship between isoprenoid flux and protein prenylation. While a multitude of statin effects have been shown to be due to disruption of protein prenylation in vitro, there is less evidence to suggest that clinically relevant doses of statins alter protein prenylation in vivo. The concentrations required to limit prenylation in vitro are significantly higher (low micromolar) than the concentrations that inhibit cholesterol biosynthesis (IC50 10 nM) [126]. Therefore, it is generally believed that under standard hypercholesterolemia dosing regimens which result in serum drug levels of 0.1 mM [127], cholesterol synthesis is inhibited but protein prenylation is conserved. Animal studies utilizing high-dose statins have shown evidence of disruption of protein prenylation [128,129]. Several phase I studies involving oncology patients have demonstrated that administration of high-dose statin can yield serum drug levels in the low micromolar range [130,131]. However, assessment of protein prenylation was not described in these reports. In a small study in which patients with acute myeloid leukemia were given high doses of lovastatin, changes in HMGR activity but not Ras farnesylation were detected [132]. Thus, whether any of the described pleiotropic effects of statins in humans are attributable to disruption of protein prenylation has yet to be established. The observation that statins are cytotoxic to a wide variety of cancer cells in vitro coupled with the identification of Ras as an important oncogene, generated much interest in the potential use of statins as
290
SARAH A. HOLSTEIN
anticancer agents. Phase I and II studies demonstrated that high-doses of statins were generally well tolerated, although minimal anticancer activity was noted [130,131,133–135]. As statins have been shown to increase the cytotoxicity of a wide variety of standard chemotherapeutic agents in vitro, there has also been interest in combination therapy. A number of clinical trials have now been conducted evaluating the combination of statins and chemotherapy [136–142]. As these trials involve different kinds and doses of statins, multiple different chemotherapeutic agents, and multiple types of malignancies, it is difficult to generate a definitive conclusion regarding the efficacy of statins in this setting. In general however, significant clinical benefit has not been demonstrated. This may be a consequence of insufficient disruption of protein prenylation. Not only does statin-induced mevalonate depletion affect the function of prenylated small GTPases, but studies have also revealed an effect on the expression of the GTPases. It was observed that lovastatin, in addition to diminishing Ras farnesylation in cultured cells, also appeared to increase the total amount of Ras protein [143]. Subsequent studies demonstrated that mevalonate depletion results in the upregulation of Ras and Ras-related proteins by discrete mechanisms including modulation of transcriptional, translational, and posttranslational processes [144]. Studies utilizing specific prenyltransferase inhibitors revealed that inhibition of prenylation was not the signal required for the observed upregulation but instead was a consequence of depletion of key regulatory isoprenoid species [145]. The identification of isoprenoids with either functional agonist or antagonist properties with respect to the endogenous isoprenoid pyrophosphates suggested the existence of specific isoprenoid-binding factors which are involved in the regulation of Ras-related protein expression [146].
V.
Future Directions
Since their discovery over 30 years ago, statins have proven to be remarkably useful agents in both the basic science and clinical arenas. Worldwide it is estimated that 25 million people are taking these agents. This number could further increase as we learn more about the potential use of statins in other disorders. Better understanding of the role of isoprenoids and isoprenoid-derivatives in human health and disease will undoubtedly lead to the identification of new therapeutic targets and pharmaceutical agents.
12. THE ISOPRENOID BIOSYNTHETIC PATHWAY AND STATINS
291
REFERENCES 1. Sacchettini, J.C., and Poulter, C.D. (1997). Creating isoprenoid diversity. Science 277:1788–1789. 2. Porter, J.W., and Spurgeon, S.L. (1981). Biosynthesis of Isoprenoid Compounds. Wiley-Interscience Publication, New York. 3. Ruzicka, L. (1953). The isoprene rule and the biogenesis of terpenic compounds. Experientia 9:357–367. 4. Lynen, F., Agranoff, B.W., Eggerer, H., Henning, U., and Moslein, E.M. (1959). Gammagamma-dimethyl-allyl-pyrophosphate and geranyl-pyrophosphate, biological preliminary stages of squalene. 6. Biosynthesis of Terpenes. Angew Chem 71:657. 5. Witting, L.A., and Porter, J.W. (1959). Intermediates in the conversion of mevalonic acid to squalene by the rat liver enzyme system. J Biol Chem 234:2841–2846. 6. Goodman, D.S., and Popjak, G. (1960). Studies on the biosynthesis of cholesterol: XII. Synthesis of allyl pyrophosphates from mevalonate and their conversion into squalene with liver enzymes. J Lipid Res 1:286–300. 7. Grob, E.C., Kirschner, K., and Lynen, F. (1961). Neues uber die biosynthese der carotinoide. Chimia 15:308–310. 8. Ferguson, J.J., Durr, I.F., and Rudney, H. (1959). The Biosynthesis of Mevalonic Acid. Proc Natl Acad Sci USA 45:499–504. 9. Tchen, T.T. (1958). Mevalonic kinase: purification and properties. J Biol Chem 233:1100–1103. 10. Tada, M., and Lynen, F. (1961). On the biosynthesis of terpenes. XIV. On the determination of phosphomevalonic acid kinase and pyrophosphomevalonic acid decarboxylase in cell extracts. J Biochem 49:758–764. 11. Agranoff, B.W., Eggerer, H., Henning, U., and Lynen, F. (1960). Biosynthesis of terpenes. VII. Isopentenyl pyrophosphate isomerase. J Biol Chem 235:326–332. 12. Poulter, C.D., and Rilling, H.C. (1978). Enzymatic and mechanistic studies on the 1’-4 coupling reaction in the terpene biosynthetic pathway. Acc Chem Res 11:307–313. 13. Croteau, R., and Purkett, P.T. (1989). Geranyl pyrophosphate synthase: characterization of the enzyme and evidence that this chain-length specific prenyltransferase is associated with monoterpene biosynthesis in sage (Salvia officinalis). Arch Biochem Biophys 271:524–535. 14. Holstein, S.A., Tong, H., Kuder, C.H., and Hohl, R.J. (2009). Quantitative determination of geranyl diphosphate levels in cultured human cells. Lipids 44:1055–1062. 15. Beytia, E., Qureshi, A.A., and Porter, J.W. (1973). Squalene synthetase. 3. Mechanism of the reaction. J Biol Chem 248:1856–1867. 16. Kandutsch, A.A., Paulus, H., Levin, E., and Bloch, K. (1964). Purification of Geranylgeranyl Pyrophosphate Synthetase from Micrococcus Lysodeikticus. J Biol Chem 239:2507–2515. 17. Moomaw, J.F., and Casey, P.J. (1992). Mammalian protein geranylgeranyltransferase. Subunit composition and metal requirements. J Biol Chem 267:17438–17443. 18. Reiss, Y., Goldstein, J.L., Seabra, M.C., Casey, P.J., and Brown, M.S. (1990). Inhibition of purified p21ras farnesyl: protein transferase by Cys-AAX tetrapeptides. Cell 62:81–88. 19. Yokoyama, K., and Gelb, M.H. (1993). Purification of a mammalian protein geranylgeranyltransferase. Formation and catalytic properties of an enzyme-geranylgeranyl pyrophosphate complex. J Biol Chem 268:4055–4060. 20. Armstrong, S.A., Seabra, M.C., Sudhof, T.C., Goldstein, J.L., and Brown, M.S. (1993). cDNA cloning and expression of the alpha and beta subunits of rat Rab geranylgeranyl transferase. J Biol Chem 268:12221–12229.
292
SARAH A. HOLSTEIN
21. Wang, K.C., and Ohnuma, S. (2000). Isoprenyl diphosphate synthases. Biochim Biophys Acta 1529:33–48. 22. Teclebrhan, H., Olsson, J., Swiezewska, E., and Dallner, G. (1993). Biosynthesis of the side chain of ubiquinone: trans-prenyltransferase in rat liver microsomes. J Biol Chem 268:23081–23086. 23. Matsuoka, S., Sagami, H., Kurisaki, A., and Ogura, K. (1991). Variable product specificity of microsomal dehydrodolichyl diphosphate synthase from rat liver. J Biol Chem 266:3464–3468. 24. Ibata, K., Mizuno, M., Takigawa, T., and Tanaka, Y. (1983). Long-chain betulaprenoltype polyprenols from the leaves of Ginkgo biloba. Biochem J 213:305–311. 25. Fujisaki, S., Hara, H., Nishimura, Y., Horiuchi, K., and Nishino, T. (1990). Cloning and nucleotide sequence of the ispA gene responsible for farnesyl diphosphate synthase activity in Escherichia coli. J Biochem 108:995–1000. 26. Apfel, C.M., Takacs, B., Fountoulakis, M., Stieger, M., and Keck, W. (1999). Use of genomics to identify bacterial undecaprenyl pyrophosphate synthetase: cloning, expression, and characterization of the essential uppS gene. J Bacteriol 181:483–492. 27. Ogura, K., Koyama, T., and Sagami, H. (1997). Polyprenyl diphosphate synthases. Subcell Biochem 28:57–87. 28. Sprenger, G.A., Schorken, U., Wiegert, T., Grolle, S., de Graaf, A.A., Taylor, S.V., Begley, T.P., Bringer-Meyer, S., and Sahm, H. (1997). Identification of a thiamin-dependent synthase in Escherichia coli required for the formation of the 1-deoxy-D-xylulose 5-phosphate precursor to isoprenoids, thiamin, and pyridoxol. Proc Natl Acad Sci USA 94:12857–12862. 29. Lange, B.M., Wildung, M.R., McCaskill, D., and Croteau, R. (1998). A family of transketolases that directs isoprenoid biosynthesis via a mevalonate-independent pathway. Proc Natl Acad Sci USA 95:2100–2104. 30. Eisenreich, W., Schwarz, M., Cartayrade, A., Arigoni, D., Zenk, M.H., and Bacher, A. (1998). The deoxyxylulose phosphate pathway of terpenoid biosynthesis in plants and microorganisms. Chem Biol 5:R221–R233. 31. Flesch, G., and Rohmer, M. (1988). Prokaryotic hopanoids: the biosynthesis of the bacteriohopane skeleton. Formation of isoprenic units from two distinct acetate pools and a novel type of carbon/carbon linkage between a triterpene and D-ribose. Eur J Biochem 175:405–411. 32. Lange, B.M., and Croteau, R. (1999). Isopentenyl diphosphate biosynthesis via a mevalonate-independent pathway: isopentenyl monophosphate kinase catalyzes the terminal enzymatic step. Proc Natl Acad Sci USA 96:13714–13719. 33. Lichtenthaler, H.K., Rohmer, M., and Schwender, J. (1997). Two independent biochemical pathways for isopentenyl diphosphate and isoprenoid biosynthesis in higher plants. Physiol Plant 101:643–652. 34. Boucher, Y., and Doolittle, W.F. (2000). The role of lateral gene transfer in the evolution of isoprenoid biosynthesis pathways. Mol Microbiol 37:703–716. 35. Flesch, G., and Rohmer, M. (1989). Prokaryotic triterpenoids. A novel hopanoid from the ethanol-producing bacterium Zymomonas mobilis. Biochem J 262:673–675. 36. Rohmer, M., Knani, M., Simonin, P., Sutter, B., and Sahm, H. (1993). Isoprenoid biosynthesis in bacteria: a novel pathway for the early steps leading to isopentenyl diphosphate. Biochem J 295(Pt 2):517–524. 37. Putra, S.R., Disch, A., Bravo, J.M., and Rohmer, M. (1998). Distribution of mevalonate and glyceraldehyde 3-phosphate/pyruvate routes for isoprenoid biosynthesis in some gram-negative bacteria and mycobacteria. FEMS Microbiol Lett 164:169–175.
12. THE ISOPRENOID BIOSYNTHETIC PATHWAY AND STATINS
293
38. Knoss, W., Reuter, B., and Zapp, J. (1997). Biosynthesis of the labdane diterpene marrubiin in Marrubium vulgare via a non-mevalonate pathway. Biochem J 326(Pt 2):449–454. 39. Schwender, J., Seemann, M., Lichtenthaler, H.K., and Rohmer, M. (1996). Biosynthesis of isoprenoids (carotenoids, sterols, prenyl side-chains of chlorophylls and plastoquinone) via a novel pyruvate/glyceraldehyde 3-phosphate non-mevalonate pathway in the green alga Scenedesmus obliquus. Biochem J 316(Pt 1):73–80. 40. Disch, A., and Rohmer, M. (1998). On the absence of the glyceraldehyde 3-phosphate/ pyruvate pathway for isoprenoid biosynthesis in fungi and yeasts. FEMS Microbiol Lett 168:201–208. 41. Endo, A., Kuroda, M., and Tsujita, Y. (1976). ML-236A, ML-236B, and ML-236C, new inhibitors of cholesterogenesis produced by Penicillium citrinium. J Antibiot 29:1346–1348. 42. Brown, A.G., Smale, T.C., King, T.J., Hasenkamp, R., and Thompson, R.H. (1976). Crystal and molecular structure of compactin, a new antifungal metabolite from Penicillium brevicompactum. J Chem Soc Perk 1 1:1165–1170. 43. Tanzawa, K., and Endo, A. (1979). Kinetic analysis of the reaction catalyzed by rat-liver 3-hydroxy-3-methylglutaryl-coenzyme-A reductase using two specific inhibitors. Eur J Biochem 98:195–201. 44. Kaneko, I., Hazama-Shimada, Y., and Endo, A. (1978). Inhibitory effects on lipid metabolism in cultured cells of ML-236B, a potent inhibitor of 3-hydroxy-3-methylglutarylcoenzyme-A reductase. Eur J Biochem 87:313–321. 45. Doi, O., and Endo, A. (1978). Specific inhibition of desmosterol synthesis by ML–236B in mouse LM cells grown in suspension in a lipid-free medium. Jpn J Med Sci Biol 31:225–233. 46. Tsujita, Y., Kuroda, M., Tanzawa, K., Kitano, N., and Endo, A. (1979). Hypolipidemic effects in dogs of ML-236B, a competitive inhibitor of 3-hydroxy-3-methylglutaryl coenzyme A reductase. Atherosclerosis 32:307–313. 47. Kuroda, M., Tsujita, Y., Tanzawa, K., and Endo, A. (1979). Hypolipidemic effects in monkeys of ML-236B, a competitive inhibitor of 3-hydroxy-3-methylglutaryl coenzyme A reductase. Lipids 14:585–589. 48. Yamamoto, A., Sudo, H., and Endo, A. (1980). Therapeutic effects of ML-236B in primary hypercholesterolemia. Atherosclerosis 35:259–266. 49. Alberts, A.W., Chen, J., Kuron, G., Hunt, V., Huff, J., Hoffman, C., Rothrock, J., Lopez, M., Joshua, H., Harris, E., Patchett, A., Monaghan, R., et al. (1980). Mevinolin: a highly potent competitive inhibitor of hydroxymethylglutaryl-coenzyme A reductase and a cholesterol-lowering agent. Proc Natl Acad Sci USA 77:3957–3961. 50. Gunde-Cimerman, N., Plemenitas, A., and Cimerman, A. (1993). Pleurotus fungi produce mevinolin, an inhibitor of HMG CoA reductase. FEMS Microbiol Lett 113:333–337. 51. Gunde-Cimerman, N., Plemenitas, A., and Cimerman, A. (1995). A hydroxymethylglutaryl-CoA reductase inhibitor synthesized by yeasts. FEMS Microbiol Lett 132:39–43. 52. Endo, A. (1979). Monacolin K, a new hypocholesterolemic agent produced by a Monascus species. J Antibiot (Tokyo) 32:852–854. 53. Shindia, A.A. (2000). Studies on mevinolin production by some fungi. Microbios 102:53–61. 54. Hajjaj, H., Niederberger, P., and Duboc, P. (2001). Lovastatin biosynthesis by Aspergillus terreus in a chemically defined medium. Appl Environ Microb 67:2596–2602. 55. Abe, Y., Suzuki, T., Ono, C., Iwamoto, K., Hosobuchi, M., and Yoshikawa, H. (2002). Molecular cloning and characterization of an ML-236B (compactin) biosynthetic gene cluster in Penicillium citrinum. Mol Genet Genomics 267:636–646.
294
SARAH A. HOLSTEIN
56. Hutchinson, C.R., Kennedy, J., Park, C., Kendrew, S., Auclair, K., and Vederas, J. (2000). Aspects of the biosynthesis of non-aromatic fungal polyketides by iterative polyketide synthases. Antonie Van Leeuwenhoek 78:287–295. 57. Ikeura, R., Murakawa, S., and Endo, A. (1988). Growth inhibition of yeast by compactin (ML-236B) analogues. J Antibiot (Tokyo) 41:1148–1150. 58. Bach, T.J., and Lichtenthaler, H.K. (1982). Mevinolin: a highly specific inhibitor of microsomal 3-hydroxy-3-methylglutaryl-coenzyme A reductase of radish plants. Z Naturforsch C 37:46–50. 59. Josekutty, P.C. (1998). Inhibition of plant growth by mevinolin and reversal of this inhibition by isoprenoids. S Afr J Bot 64:18–24. 60. Shindia, A.A. (2001). Some nutritional factors influencing mevinolin production by Aspergillus terreus strain. Folia Microbiol 46:413–416. 61. Furberg, C.D., and Pitt, B. (2001). Withdrawal of cerivastatin from the world market. Curr Control Trials Cardiovasc Med 2:205–207. 62. Corsini, A., Maggi, F.M., and Catapano, A.L. (1995). Pharmacology of competitive inhibitors of HMG-CoA reductase. Pharmacol Res 31:9–27. 63. Istvan, E.S., and Deisenhofer, J. (2001). Structural mechanism for statin inhibition of HMG-CoA reductase. Science 292:1160–1164. 64. Chao, Y.S., Kroon, P.A., Yamin, T.T., Thompson, G.M., and Alberts, A.W. (1983). Regulation of hepatic receptor-dependent degradation of LDL by mevinolin in rabbits with hypercholesterolemia induced by a wheat starch-casein diet. Biochem Biophys Acta 754:134–141. 65. Bilheimer, D.W., Grundy, S.M., Brown, M.S., and Goldstein, J.L. (1983). Mevinolin and colestipol stimulate receptor-mediated clearance of low density lipoprotein from plasma in familial hypercholesterolemia heterozygotes. Proc Natl Acad Sci USA 80:4124–4128. 66. Beaird, S.L. (2000). HMG-CoA reductase inhibitors: assessing differences in drug interactions and safety profiles. J Am Pharm Assoc (Wash) 40:637–644. 67. Kajinami, K., Takekoshi, N., and Saito, Y. (2003). Pitavastatin: efficacy and safety profiles of a novel synthetic HMG-CoA reductase inhibitor. Cardiovasc Drug Rev 21:199–215. 68. Rao, S., Porter, D.C., Chen, X., Herliczek, T., Lowe, M., and Keyomarsi, K. (1999). Lovastatin-mediated G1 arrest is through inhibition of the proteasome, independent of hydroxymethyl glutaryl-CoA reductase. Proc Natl Acad Sci USA 96:7797–7802. 69. Wojcik, C., Bury, M., Stoklosa, T., Giermasz, A., Feleszko, W., Mlynarczuk, I., Pleban, E., Basak, G., Omura, S., and Jakobisiak, M. (2000). Lovastatin and simvastatin are modulators of the proteasome. Int J Biochem Cell Biol 32:957–965. 70. Kumar, B., Andreatta, C., Koustas, W.T., Cole, W.C., Edwards-Prasad, J., and Prasad, K.N. (2002). Mevastatin induces degeneration and decreases viability of cAMP-induced differentiated neuroblastoma cells in culture by inhibiting proteasome activity, and mevalonic acid lactone prevents these effects. J Neurosci Res 68:627–635. 71. Ludwig, A., Friedel, B., Metzkow, S., Meiners, S., Stangl, V., Baumann, G., and Stangl, K. (2005). Effect of statins on the proteasomal activity in mammalian endothelial and vascular smooth muscle cells. Biochem Pharmacol 70:520–526. 72. Murray, S.S., Tu, K.N., Young, K.L., and Murray, E.J. (2002). The effects of lovastatin on proteasome activities in highly purified rabbit 20 S proteasome preparations and mouse MC3T3-E1 osteoblastic cells. Metabolism 51:1153–1160. 73. Garrett, I.R., Chen, D., Gutierrez, G., Zhao, M., Escobedo, A., Rossini, G., Harris, S.E., Gallwitz, W., Kim, K.B., Hu, S., Crews, C.M., and Mundy, G.R. (2003). Selective inhibitors of the osteoblast proteasome stimulate bone formation in vivo and in vitro. J Clin Invest 111:1771–1782.
12. THE ISOPRENOID BIOSYNTHETIC PATHWAY AND STATINS
295
74. Zhou, Q., and Liao, J.K. (2010). Pleiotropic effects of statins—basic research and clinical perspectives. Circ J 74:818–826. 75. Hindler, K., Cleeland, C.S., Rivera, E., and Collard, C.D. (2006). The role of statins in cancer therapy. Oncologist 11:306–315. 76. Willey, J.Z., and Elkind, M.S. (2010). 3-Hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors in the treatment of central nervous system diseases. Arch Neurol 67:1062–1067. 77. Tang, Q.O., Tran, G.T., Gamie, Z., Graham, S., Tsialogiannis, E., Tsiridis, E., and Linder, T. (2008). Statins: under investigation for increasing bone mineral density and augmenting fracture healing. Expert Opin Investig Drugs 17:1435–1463. 78. Lee, Y.L., Blaha, M.J., and Jones, S.R. (2011). Statin therapy in the prevention and treatment of atrial fibrillation. J Clin Lipidol 5:18–29. 79. Zeki, A.A., Kenyon, N.J., and Goldkorn, T. (2011). Statin drugs, metabolic pathways, and asthma: a therapeutic opportunity needing further research. Drug Metab Lett 5:40–44. 80. Elewa, H.F., El-Remessy, A.B., Somanath, P.R., and Fagan, S.C. (2010). Diverse effects of statins on angiogenesis: new therapeutic avenues. Pharmacotherapy 30:169–176. 81. Smaldone, C., Brugaletta, S., Pazzano, V., and Liuzzo, G. (2009). Immunomodulator activity of 3-hydroxy-3-methilglutaryl-CoA inhibitors. Cardiovasc Hematol Agents Med Chem 7:279–294. 82. Krysiak, R., Okopien, B., and Herman, Z. (2003). Effects of HMG-CoA reductase inhibitors on coagulation and fibrinolysis processes. Drugs 63:1821–1854. 83. Mitsios, J.V., Papathanasiou, A.I., Goudevenos, J.A., and Tselepis, A.D. (2010). The antiplatelet and antithrombotic actions of statins. Curr Pharm Des 16:3808–3814. 84. Mas, E., and Mori, T.A. (2010). Coenzyme Q(10) and statin myalgia: what is the evidence? Curr Atheroscler Rep 12:407–413. 85. Ghirlanda, G., Oradei, A., Manto, A., Lippa, S., Uccioli, L., Caputo, S., Greco, A.V., and Littarru, G.P. (1993). Evidence of plasma CoQ10-lowering effect by HMG-CoA reductase inhibitors: a double-blind, placebo-controlled study. J Clin Pharmacol 33:226–229. 86. Miyake, Y., Shouzu, A., Nishikawa, M., Yonemoto, T., Shimizu, H., Omoto, S., Hayakawa, T., and Inada, M. (1999). Effect of treatment with 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors on serum coenzyme Q10 in diabetic patients. Arzneimittelforschung 49:324–329. 87. Willis, R.A., Folkers, K., Tucker, J.L., Ye, C.Q., Xia, L.J., and Tamagawa, H. (1990). Lovastatin decreases coenzyme Q levels in rats. Proc Natl Acad Sci USA 87:8928–8930. 88. Folkers, K., Langsjoen, P., Willis, R., Richardson, P., Xia, L.J., Ye, C.Q., and Tamagawa, H. (1990). Lovastatin decreases coenzyme Q levels in humans. Proc Natl Acad Sci USA 87:8931–8934. 89. Mabuchi, H., Nohara, A., Kobayashi, J., Kawashiri, M.A., Katsuda, S., Inazu, A., and Koizumi, J. (2007). Effects of CoQ10 supplementation on plasma lipoprotein lipid, CoQ10 and liver and muscle enzyme levels in hypercholesterolemic patients treated with atorvastatin: a randomized double-blind study. Atherosclerosis 195:e182–e189. 90. Marcoff, L., and Thompson, P.D. (2007). The role of coenzyme Q10 in statin-associated myopathy: a systematic review. J Am Coll Cardiol 49:2231–2237. 91. Schaefer, W.H., Lawrence, J.W., Loughlin, A.F., Stoffregen, D.A., Mixson, L.A., Dean, D.C., Raab, C.E., Yu, N.X., Lankas, G.R., and Frederick, C.B. (2004). Evaluation of ubiquinone concentration and mitochondrial function relative to cerivastatin-induced skeletal myopathy in rats. Toxicol Appl Pharmacol 194:10–23. 92. Laaksonen, R., Jokelainen, K., Sahi, T., Tikkanen, M.J., and Himberg, J.J. (1995). Decreases in serum ubiquinone concentrations do not result in reduced levels in muscle tissue during short-term simvastatin treatment in humans. Clin Pharmacol Ther 57:62–66.
296
SARAH A. HOLSTEIN
93. Laaksonen, R., Jokelainen, K., Laakso, J., Sahi, T., Harkonen, M., Tikkanen, M.J., and Himberg, J.J. (1996). The effect of simvastatin treatment on natural antioxidants in low-density lipoproteins and high-energy phosphates and ubiquinone in skeletal muscle. Am J Cardiol 77:851–854. 94. Paiva, H., Thelen, K.M., Van Coster, R., Smet, J., De Paepe, B., Mattila, K.M., Laakso, J., Lehtimaki, T., von Bergmann, K., Lutjohann, D., and Laaksonen, R. (2005). High-dose statins and skeletal muscle metabolism in humans: a randomized, controlled trial. Clin Pharmacol Ther 78:60–68. 95. Caso, G., Kelly, P., McNurlan, M.A., and Lawson, W.E. (2007). Effect of coenzyme q10 on myopathic symptoms in patients treated with statins. Am J Cardiol 99:1409–1412. 96. Young, J.M., Florkowski, C.M., Molyneux, S.L., McEwan, R.G., Frampton, C.M., George, P.M., and Scott, R.S. (2007). Effect of coenzyme Q(10) supplementation on simvastatin-induced myalgia. Am J Cardiol 100:1400–1403. 97. Ohashi, K., Osuga, J., Tozawa, R., Kitamine, T., Yagyu, H., Sekiya, M., Tomita, S., Okazaki, H., Tamura, Y., Yahagi, N., Iizuka, Y., Harada, K., et al. (2003). Early embryonic lethality caused by targeted disruption of the 3-hydroxy-3-methylglutaryl-CoA reductase gene. J Biol Chem 278:42936–42941. 98. Hoffmann, G.F., Charpentier, C., Mayatepek, E., Mancini, J., Leichsenring, M., Gibson, K.M., Divry, P., Hrebicek, M., Lehnert, W., Sartor, K., et al. (1993). Clinical and biochemical phenotype in 11 patients with mevalonic aciduria. Pediatrics 91:915–921. 99. Houten, S.M., Koster, J., Romeijn, G.J., Frenkel, J., Di Rocco, M., Caruso, U., Landrieu, P., Kelley, R.I., Kuis, W., Poll-The, B.T., Gibson, K.M., Wanders, R.J., et al. (2001). Organization of the mevalonate kinase (MVK) gene and identification of novel mutations causing mevalonic aciduria and hyperimmunoglobulinaemia D and periodic fever syndrome. Eur J Hum Genet 9:253–259. 100. Houten, S.M., Kuis, W., Duran, M., de Koning, T.J., van Royen-Kerkhof, A., Romeijn, G.J., Frenkel, J., Dorland, L., de Barse, M.M., Huijbers, W.A., Rijkers, G.T., Waterham, H.R., et al. (1999). Mutations in MVK, encoding mevalonate kinase, cause hyperimmunoglobulinaemia D and periodic fever syndrome. Nat Genet 22:175–177. 101. Hoffmann, G., Gibson, K.M., Nyhan, W.L., and Sweetman, L. (1988). Mevalonic aciduria: pathobiochemical effects of mevalonate kinase deficiency on cholesterol metabolism in intact fibroblasts. J Inherit Metab Dis 11(Suppl 2):229–232. 102. Gibson, K.M., Hoffmann, G., Schwall, A., Broock, R.L., Aramaki, S., Sweetman, L., Nyhan, W.L., Brandt, I.K., Wappner, R.S., Lehnert, W., et al. (1990). 3-Hydroxy-3methylglutaryl coenzyme A reductase activity in cultured fibroblasts from patients with mevalonate kinase deficiency: differential response to lipid supplied by fetal bovine serum in tissue culture medium. J Lipid Res 31:515–521. 103. Houten, S.M., Schneiders, M.S., Wanders, R.J., and Waterham, H.R. (2003). Regulation of isoprenoid/cholesterol biosynthesis in cells from mevalonate kinase-deficient patients. J Biol Chem 278:5736–5743. 104. Brown, M.S., Faust, J.R., Goldstein, J.L., Kaneko, I., and Endo, A. (1978). Induction of 3-hydroxy-3-methylglutaryl coenzyme A reductase activity in human fibroblasts incubated with compactin (ML-236B), a competitive inhibitor of the reductase. J Biol Chem 253:1121–1128. 105. Chin, D.J., Luskey, K.L., Anderson, R.G., Faust, J.R., Goldstein, J.L., and Brown, M.S. (1982). Appearance of crystalloid endoplasmic reticulum in compactin-resistant Chinese hamster cells with a 500-fold increase in 3-hydroxy-3-methylglutaryl-coenzyme A reductase. Proc Natl Acad Sci USA 79:1185–1189. 106. Faust, J.R., Luskey, K.L., Chin, D.J., Goldstein, J.L., and Brown, M.S. (1982). Regulation of synthesis and degradation of 3-hydroxy-3-methylglutaryl-coenzyme A reductase by
12. THE ISOPRENOID BIOSYNTHETIC PATHWAY AND STATINS
107.
108.
109.
110.
111.
112.
113.
114.
115.
116. 117. 118. 119. 120.
121.
122.
123.
297
low density lipoprotein and 25-hydroxycholesterol in UT-1 cells. Proc Natl Acad Sci USA 79:5205–5209. Liscum, L., Finer-Moore, J., Stroud, R.M., Luskey, K.L., Brown, M.S., and Goldstein, J.L. (1985). Domain structure of 3-hydroxy-3-methylglutaryl coenzyme A reductase, a glycoprotein of the endoplasmic reticulum. J Biol Chem 260:522–530. Nakanishi, M., Goldstein, J.L., and Brown, M.S. (1988). Multivalent control of 3-hydroxy3-methylglutaryl coenzyme A reductase. Mevalonate-derived product inhibits translation of mRNA and accelerates degradation of enzyme. J Biol Chem 263:8929–8937. Roitelman, J., and Simoni, R.D. (1992). Distinct sterol and nonsterol signals for the regulated degradation of 3-hydroxy-3-methylglutaryl-CoA reductase. J Biol Chem 267:25264–25273. Dawson, P.A., Metherall, J.E., Ridgway, N.D., Brown, M.S., and Goldstein, J.L. (1991). Genetic distinction between sterol-mediated transcriptional and posttranscriptional control of 3-hydroxy-3-methylglutaryl-coenzyme A reductase. J Biol Chem 266:9128–9134. Sheng, Z., Otani, H., Brown, M.S., and Goldstein, J.L. (1995). Independent regulation of sterol regulatory element-binding proteins 1 and 2 in hamster liver. Proc Natl Acad Sci USA 92:935–938. Mascaro, C., Ortiz, J.A., Ramos, M.M., Haro, D., and Hegardt, F.G. (2000). Sterol regulatory element binding protein-mediated effect of fluvastatin on cytosolic 3hydroxy-3-methylglutaryl-coenzyme A synthase transcription. Arch Biochem Biophys 374:286–292. Sever, N., Yang, T., Brown, M.S., Goldstein, J.L., and DeBose-Boyd, R.A. (2003). Accelerated degradation of HMG CoA reductase mediated by binding of insig-1 to its sterol-sensing domain. Mol Cell 11:25–33. Xu, L., and Simoni, R.D. (2003). The inhibition of degradation of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase by sterol regulatory element binding protein cleavage-activating protein requires four phenylalanine residues in span 6 of HMG-CoA reductase transmembrane domain. Arch Biochem Biophys 414:232–243. Schmidt, R.A., Schneider, C.J., and Glomset, J.A. (1984). Evidence for post-translational incorporation of a product of mevalonic acid into Swiss 3T3 cell proteins. J Biol Chem 259:10175–10180. Farnsworth, C.C., Wolda, S.L., Gelb, M.H., and Glomset, J.A. (1989). Human lamin B contains a farnesylated cysteine residue. J Biol Chem 264:20422–20429. Hancock, J.F., Magee, A.I., Childs, J.E., and Marshall, C.J. (1989). All ras proteins are polyisoprenylated but only some are palmitoylated. Cell 57:1167–1177. Casey, P.J., Solski, P.A., Der, C.J., and Buss, J.E. (1989). p21ras is modified by a farnesyl isoprenoid. Proc Natl Acad Sci USA 86:8323–8327. Rilling, H.C., Breunger, E., Epstein, W.W., and Crain, P.F. (1990). Prenylated proteins: the structure of the isoprenoid group. Science 247:318–320. Mumby, S.M., Casey, P.J., Gilman, A.G., Gutowski, S., and Sternweis, P.C. (1990). G protein gamma subunits contain a 20-carbon isoprenoid. Proc Natl Acad Sci USA 87:5873–5877. Kinsella, B.T., and Maltese, W.A. (1992). rab GTP-binding proteins with three different carboxyl-terminal cysteine motifs are modified in vivo by 20-carbon isoprenoids. J Biol Chem 267:3940–3945. Danesi, R., McLellan, C.A., and Myers, C.E. (1995). Specific labeling of isoprenylated proteins: application to study inhibitors of the post-translational farnesylation and geranylgeranylation. Biochem Biophys Res Commun 206:637–643. Tong, H., Holstein, S.A., and Hohl, R.J. (2005). Simultaneous determination of farnesyl and geranylgeranyl pyrophosphate levels in cultured cells. Anal Biochem 336:51–59.
298
SARAH A. HOLSTEIN
124. Holstein, S.A., Tong, H., and Hohl, R.J. (2010). Differential activities of thalidomide and isoprenoid biosynthetic pathway inhibitors in multiple myeloma cells. Leuk Res 34:344–351. 125. Tong, H., Wiemer, A.J., Neighbors, J.D., and Hohl, R.J. (2008). Quantitative determination of farnesyl and geranylgeranyl diphosphate levels in mammalian tissue. Anal Biochem 378:138–143. 126. Sinensky, M., Beck, L.A., Leonard, S., and Evans, R. (1990). Differential inhibitory effects of lovastatin on protein isoprenylation and sterol synthesis. J Biol Chem 265:19937–19941. 127. Pan, H.Y., DeVault, A.R., Wang-Iverson, D., Ivashkiv, E., Swanson, B.N., and Sugerman, A.A. (1990). Comparative pharmacokinetics and pharmacodynamics of pravastatin and lovastatin. J Clin Pharmacol 30:1128–1135. 128. Sebti, S.M., Tkalcevic, G.T., and Jani, J.P. (1991). Lovastatin, a cholesterol biosynthesis inhibitor, inhibits the growth of human H-ras oncogene transformed cells in nude mice. Cancer Commun 3:141–147. 129. Staal, A., Frith, J.C., French, M.H., Swartz, J., Gungor, T., Harrity, T.W., Tamasi, J., Rogers, M.J., and Feyen, J.H. (2003). The ability of statins to inhibit bone resorption is directly related to their inhibitory effect on HMG-CoA reductase activity. J Bone Miner Res 18:88–96. 130. Thibault, A., Samid, D., Tompkins, A.C., Figg, W.D., Cooper, M.R., Hohl, R.J., Trepel, J., Liang, B., Patronas, N., Venzon, D.J., Reed, E., and Myers, C.E. (1996). Phase I study of lovastatin, an inhibitor of the mevalonate pathway, in patients with cancer. Clin Cancer Res 2:483–491. 131. Holstein, S.A., Knapp, H.R., Clamon, G.H., Murry, D.J., and Hohl, R.J. (2006). Pharmacodynamic effects of high dose lovastatin in subjects with advanced malignancies. Cancer Chemother Pharmacol 57:155–164. 132. Lewis, K.A., Holstein, S.A., and Hohl, R.J. (2005). Lovastatin alters the isoprenoid biosynthetic pathway in acute myelogenous leukemia cells in vivo. Leuk Res 29:527–533. 133. Larner, J., Jane, J., Laws, E., Packer, R., Myers, C., and Shaffrey, M. (1998). A phase I-II trial of lovastatin for anaplastic astrocytoma and glioblastoma multiforme. Am J Clin Oncol 21:579–583. 134. Knox, J.J., Siu, L.L., Chen, E., Dimitroulakos, J., Kamel-Reid, S., Moore, M.J., Chin, S., Irish, J., LaFramboise, S., and Oza, A.M. (2005). A Phase I trial of prolonged administration of lovastatin in patients with recurrent or metastatic squamous cell carcinoma of the head and neck or of the cervix. Eur J Cancer 41:523–530. 135. Kim, W.S., Kim, M.M., Choi, H.J., Yoon, S.S., Lee, M.H., Park, K., Park, C.H., and Kang, W.K. (2001). Phase II study of high-dose lovastatin in patients with advanced gastric adenocarcinoma. Invest New Drugs 19:81–83. 136. van der Spek, E., Bloem, A.C., van de Donk, N.W., Bogers, L.H., van der Griend, R., Kramer, M.H., de Weerdt, O., Wittebol, S., and Lokhorst, H.M. (2006). Dose-finding study of high-dose simvastatin combined with standard chemotherapy in patients with relapsed or refractory myeloma or lymphoma. Haematologica 91:542–545. 137. van der Spek, E., Bloem, A.C., Sinnige, H.A., and Lokhorst, H.M. (2007). High dose simvastatin does not reverse resistance to vincristine, adriamycin, and dexamethasone (VAD) in myeloma. Haematologica 92:e130–e131. 138. Lopez-Aguilar, E., Sepulveda-Vildosola, A.C., Betanzos-Cabrera, Y., Rocha-Moreno, Y.G., Gascon-Lastiri, G., Rivera-Marquez, H., Wanzke-del-Angel, V., Cerecedo-Diaz, F., and de la Cruz-Yanez, H. (2008). Phase II study of metronomic chemotherapy with thalidomide, carboplatin-vincristine-fluvastatin in the treatment of brain stem tumors in children. Arch Med Res 39:655–662.
12. THE ISOPRENOID BIOSYNTHETIC PATHWAY AND STATINS
299
139. Lee, J., Jung, K.H., Park, Y.S., Ahn, J.B., Shin, S.J., Im, S.A., Oh do, Y., Shin, D.B., Kim, T.W., Lee, N., Byun, J.H., Hong, Y.S., et al. (2009). Simvastatin plus irinotecan, 5-fluorouracil, and leucovorin (FOLFIRI) as first-line chemotherapy in metastatic colorectal patients: a multicenter phase II study. Cancer Chemother Pharmacol 64:657–663. 140. Konings, I.R., van der Gaast, A., van der Wijk, L.J., de Jongh, F.E., Eskens, F.A., and Sleijfer, S. (2010). The addition of pravastatin to chemotherapy in advanced gastric carcinoma: a randomised phase II trial. Eur J Cancer 46:3200–3204. 141. Han, J.Y., Lim, K.Y., Yu, S.Y., Yun, T., Kim, H.T., and Lee, J.S. (2011). A phase 2 study of irinotecan, cisplatin, and simvastatin for untreated extensive-disease small cell lung cancer. Cancer 117:2178–2185. 142. Han, J.Y., Lee, S.H., Yoo, N.J., Hyung, L.S., Moon, Y.J., Yun, T., Kim, H.T., and Lee, J.S. (2011). A randomized phase II study of gefitinib plus simvastatin versus gefitinib alone in previously treated patients with advanced non-small cell lung cancer. Clin Cancer Res 17:1553–1560. 143. Hohl, R.J., and Lewis, K. (1995). Differential effects of monoterpenes and lovastatin on RAS processing. J Biol Chem 270:17508–17512. 144. Holstein, S.A., Wohlford-Lenane, C.L., and Hohl, R.J. (2002). Consequences of mevalonate depletion. Differential transcriptional, translational, and post-translational up-regulation of Ras, Rap1a, RhoA, AND RhoB. J Biol Chem 277:10678–10682. 145. Holstein, S.A., Wohlford-Lenane, C.L., and Hohl, R.J. (2002). Isoprenoids influence expression of Ras and Ras-related proteins. Biochemistry 41:13698–13704. 146. Holstein, S.A., Wohlford-Lenane, C.L., Wiemer, D.F., and Hohl, R.J. (2003). Isoprenoid pyrophosphate analogues regulate expression of Ras-related proteins. Biochemistry 42:4384–4391.