• No results found

University of Groningen Development of PET tracers for investigation of arginase-related pathways dos Santos Clemente, Gonçalo

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen Development of PET tracers for investigation of arginase-related pathways dos Santos Clemente, Gonçalo"

Copied!
13
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

University of Groningen

Development of PET tracers for investigation of arginase-related pathways

dos Santos Clemente, Gonçalo

DOI:

10.33612/diss.143845684

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date: 2020

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

dos Santos Clemente, G. (2020). Development of PET tracers for investigation of arginase-related pathways. University of Groningen. https://doi.org/10.33612/diss.143845684

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)

Chapter 5

Abstract

Several research groups have suggested that part of the therapeutic success of statins, and also some of the pleiotropic effects associated with this class of drugs, are due to their impact on the arginase/nitric oxide synthase (NOS) signaling pathways. Although the proposed mechanisms of action are not always universally accepted, the undeniable capacity of statins to increase NO• levels has attracted

much research interest to expand their therapeutic spectrum. Therefore, the potential effects induced by statins in ʟ-arginine metabolic pathways are briefly reviewed.

Introduction

As reviewed in chapter 2, the balance between arginase isozymes (Arg1 and Arg2) and the endothelial, neuronal, and inducible nitric oxide synthase (e/n/iNOS) has shown to be highly linked to several pathophysiological processes. Besides a range of immune-mediated pathologies, arginase is particularly overexpressed in cardiovascular diseases [1]. The upregulation of the arginase levels leads to arterial stiffening, to the depletion of ʟ-arginine (the common substrate with NOS), and consequently to the reduction of NO•-mediated cardioprotective effects [2]. For

example, a high fat/cholesterol diet may cause liver damage, which contributes to the systemic release of the hepatic cytosolic arginase and worsens further hypercholesterolemia-induced vascular dysfunction [3]. Therefore, several reports have suggested that part of the therapeutic success of statins, the most prescribed cholesterol-lowering drugs for the prevention of atherosclerotic diseases, may be related to off-target effects influencing arginase/NOS signaling pathways, in parallel to the primary HMG-CoA (3-hydroxy-3-methyl-glutaryl-coenzyme A) reductase inhibition [4].

(3)

Statins as HMG-CoA reductase inhibitors

HMG-CoA reductase is an intermediate enzyme of the mevalonate pathway, involved in the biosynthesis of cholesterol. Although this enzyme is widely expressed throughout most tissues [5], the liver is central to the balancing of cholesterol levels in the body by regulating its synthesis, transport, and elimination through bile. Thus, the inhibition of HMG-CoA reductase became an essential clinical strategy for the treatment of hypercholesterolemia, which is currently one of the lead risk factors for heart disease and stroke [6].

From the early 1970s on, when mevastatin was first isolated from Penicillium citrinum and was screened as being a potent HMG-CoA reductase inhibitor, numerous other statins have been isolated from fungi or chemically synthesized [7]. Some of these statins revealed high affinity and specificity to inhibit HMG-CoA reductase and reached clinical trials, being later added, and occasionally even withdrawn (due to adverse effects), from medical prescription (Figure 1) [8].

Figure 1. Chemical structure of mevastatin and natural, semi-synthetic, and synthetic statins currently approved for clinical use.

Statins efficiently reduce the synthesis of endogenous cholesterol by mimicking and outcompeting HMG-CoA, the HMG-CoA reductase natural substrate (Figure 2) [9]. This selective inhibition of HMG-CoA reductase triggers the activity of hepatic LDL (low-density lipoprotein) receptors, lowering plasma LDL cholesterol concentration, which is causally linked to the risk of cardiovascular events, while modestly increasing levels of the beneficial HDL (high-density lipoprotein)

(4)

cholesterol [10]. The high therapeutic effect of statins, and their relevance for the primary and secondary prevention of atherosclerotic cardiovascular diseases, made this class of drugs the most successful of all-time, with a global market value of tens of billions of dollars per year [11].

Figure 2. Cholesterol biosynthetic pathway and inhibition of HMG-CoA reductase by statins that leads to decreased levels of cholesterol and small guanosine triphosphatases (Ras, Rho, Rac1).

Pleiotropic effects of statins

In addition to the unambiguous cholesterol-lowering action, the success of statins became increasingly connected with broader pleiotropic effects [12]. More and more often, statins are being associated with potential protective effects on pathologies beyond cardiovascular diseases (e.g., respiratory [13], carcinogenic [14], viral [15], and neurodegenerative [16]), which may expand their therapeutic spectrum.

As depicted in Figure 2, the mevalonate pathway leads to pyrophosphate intermediates that are necessary for the biosynthesis of cholesterol and small guanosine triphosphatases (Ras, Rho, and Rac1) important in intracellular signal transduction. Multiple processes, such as cell migration, proliferation, or apoptosis, depend on the downstream products of mevalonate [17]. The decrease in the systemic levels of those pyrophosphate intermediates due to HMG-CoA reductase

(5)

inhibition is expected to reduce the signals from Ras, Rho, and Rac1, which may explain many of statins pleiotropic effects (e.g., antioxidant, anti-inflammatory, anti-proliferative, and immunomodulatory effects [18]), including a potential reduction of arginase activity and increase of NO• levels [19].

Arginase/NOS regulation by statins

Several in vitro and in vivo studies have confirmed the capacity of statins to suppress arginase activity and/or increase the NO• levels (Table 1).

Table 1. Statin-induced effects on the arginase/NOS signaling pathways and suggested mechanisms.

Research model Statin Output Ref

Bovine aortic endothelial cells Atorvastatin

 caveolin-1 expression  eNOS expression  NO•

[4c]

Bovine aortic endothelial cells Simvastatin

 eNOS mRNA expression  eNOS expression  NO•

[4k]

Bovine aortic endothelial cells Simvastatin Rosuvastatin

↑ eNOS mRNA stability ↑ eNOS expression  NO•

[20]

Human aortic endothelial cells Lovastatin

Simvastatin  Rho

 arginase activity [19]

Human umbilical vein endothelial cells Cerivastatin

 GTPCH mRNA  BH4 levels

 eNOS activity  NO•

[21]

Human umbilical vein endothelial cells Pitavastatin

 Akt phosphorylation  eNOS activity  NO•

[4e]

Human umbilical vein endothelial cells Pitavastatin Cerivastatin

 Akt phosphorylation  eNOS activity  NO•

[4g]

Human umbilical vein endothelial cells Fluvastatin Cerivastatin ↑ eNOS mRNA ↑ BH4 levels ↑ GTPCH mRNA  Akt phosphorylation  NO• [22]

Human umbilical vein endothelial cells Healthy mice

Atorvastatin Lovastatin

↑ AMPK

↑ eNOS [23]

Human umbilical vein and mouse cardiac

microvascular endothelial cells Atorvastatin

 caveolin-1 expression  eNOS expression  NO•

(6)

Research model Statin Output Ref

MCF-7 breast cancer cells Fluvastatin

Simvastatin  iNOS mRNA  iNOS expression  NO•  cell proliferation [25]

MCF-7 breast cancer cells Atorvastatin  PI3K signaling through Akt  cell growth suppression [26]

Healthy mice Rosuvastatin

 caveolin-1 expression  eNOS expression  NO•

[27]

Normocholesterolemic mice Atorvastatin

 Rho

↑ eNOS expression  NO•

 neuroprotection effect

[28]

Mouse model of allergic asthma Simvastatin  goblet cell hyperplasia  arginase expression [29]

Erchlich acid (breast cancer) xenograft mice Rosuvastatin

 arginase activity  ornithine levels  polyamines levels  NO•

[30]

Diabetic mice Pitavastatin

 eNOS uncoupling  GTPCH mRNA expression  eNOS uncoupling  NO•

[31]

Diabetic rats Simvastatin

 Rho

 eNOS expression  NO•

 endothelial function

[32]

Diabetic rats Simvastatin  Rho

 arginase activity [33]

Cirrhotic rats Atorvastatin

 Rho

↑ eNOS expression  NO•

[34]

Cirrhotic rats Simvastatin

 Akt phosphorylation  eNOS activity  NO•

[35]

Rats with hypoxia-induced pulmonary

hypertension Fluvastatin

 eNOS mRNA expression  endothelin synthesis/secretion  NO•

 lung function

[36]

Hypercholesterolemic rabbits Simvastatin

 eNOS mRNA expression  eNOS expression  O2

•- NO•

[4b]

Clinical trials in hypercholesterolemic

humans Atorvastatin  arginase activity  BH4 levels  eNOS uncoupling  NO• [4i, 4j, 37]

(7)

Within the scope of this chapter, assays were performed to evaluate the capacity of statins to bind and inhibit arginase. Using a label-free surface plasmon resonance ResidenceTimer™ assay developed by NTRC (Oss, The Netherlands) in a BiaCore T200 (GE Healthcare), with Arg1 (diluted in 50 mM Na2HPO4, pH 7.4, 150 mM KCl,

and 0.01% Tween-20, in a concentration of 60 μg/mL) immobilized on a sensor chip and five concentrations (between 0.1-10 µM) of a reference arginase inhibitor (2-(S)-amino-6-boronohexanoic acid) and atorvastatin, it was confirmed that atorvastatin does not inhibit or bind to arginase [unpublished data]. Therefore, HMG-CoA reductase inhibition (on-target) downstream routes and other apparently off-target mechanisms have been proposed in the literature to explain the statin-induced increase of NO• levels (Figure 3).

Figure 3. Schematic summary of the main pathways involved in the capacity of statins to suppress arginase activity and/or increase the NO• levels.

As mentioned above, statins inhibit HMG-CoA reductase by suppressing the downstream products of mevalonate, including the Rho small guanosine triphosphatase. By decreasing the availability of Rho effectors (Figure 3, pathway 1), which negatively influence eNOS mRNA stability and activity by inducing the cleavage of the poly(A) signal sequence, statins showed to increase the subsequent translation to eNOS [18c, 20]. Rho effectors also can inhibit protein kinase B (Akt), an enzyme that, together with the phosphatidylinositol-3 kinase

(8)

(PI3K), is essential for the phosphorylation and activation of eNOS [4e, 4g]. Thus, by suppressing the Rho-induced inhibition of PI3K/Akt, statins are contributing to the increase of eNOS activity. Furthermore, a specific Rho effector (mDial1) seems to be responsible for the liberation of mitochondrial Arg2, leading to arginase upregulation and depletion of ʟ-arginine. The inhibition of mDial1 by statins is also thought to be one of the mechanisms that lead to NO• increase [19].

Another way how statins may affect the phosphorylation of eNOS seems to be via activation of AMPK (adenosine monophosphate-activated protein kinase, Figure 3, pathway 2). This activation of AMPK has been shown to increase NO• production

either in vitro or in vivo, but there are still uncertainties whether or not this mechanism is directly dependent on the inhibition of the mevalonate pathway [23]. The pretreatment of endothelial cells with statins showed to decrease caveolin-1 levels (Figure 3, pathway 3) [4c, 24, 38]. Caveolin-1 is a component of plasma membranes that is usually co-localized with eNOS and can inactivate this enzyme by blocking its essential interaction with the Ca2+/calmodulin cofactor [39].

Statins seem to exert some epigenetic modifications (Figure 3, pathway 4 and 5), but the exact mechanisms of action and target regions remain unclear [4m]. Measurement of the guanosine triphosphate cyclohydrolase I (GTPCH) mRNA levels by real-time polymerase chain reaction (PCR) in vascular endothelial cells pretreated with statins showed an enhancement of this mRNA [21, 31]. GTPCH is a rate-limiting enzyme for the synthesis of tetrahydrobiopterin (BH4), which is an

essential cofactor for eNOS coupling and function. It is noteworthy that uncoupled eNOS leads to the production of O2•− parallelly to NO•, forming cytotoxic

peroxynitrite species (ONOO−). Additionally, in vitro studies with statins showed a

reduction in the expression of micro RNAs that negatively impact eNOS expression, which led to an increase of NO• release [40].

Even though some proposed mechanisms are still controversial, it is undeniable that statins increase eNOS activity and NO• levels. The production of NO involves

the synthesis of an intermediate molecule by NOS, Nω-hydroxy-ʟ-arginine (NOHA),

that is known to exert an inhibitory effect on arginase [41], which may also be an indirect way of how statins can inhibit arginase [4i, 29].

Discussion

Although the reported experimental data on the pleiotropic effects of statins are strongly suggestive and triggered a growing interest in the use of statins beyond

(9)

cardiovascular diseases [12b], there has been limited evidence of the potential benefits in humans. The long-term knowledge of some adverse effects of statins (e.g., muscle pain, digestive problems, mental fuzziness, or hepatotoxicity), and the reports of some intolerant or resistant patients, might have delayed the stimulation of further clinical trials. However, the confirmation that statins could influence the arginase–NO• network (Table 1), and the recent emergence of this enzymatic

system as a potential therapeutic target [1], led to the resurgence of the interest to use statins as an innovative or complementary therapy, especially for carcinogenic, neurodegenerative, and airway diseases [42].

Despite the fact that randomized controlled trials showed no benefits on the use of statins in patients with advanced cancer and, therefore, poor prognosis [43], epidemiological studies have associated the use of statins with lower breast and colorectal cancer risk in humans [44]. Patients with neurodegenerative and cognitive disorders treated with statins have also shown inconsistent results, as both an aggravation and an improvement in symptoms have been reported [45]. More consistent results have been achieved regarding the benefits of using statins in chronic obstructive pulmonary diseases and asthma [46], two airway inflammation disorders that recently showed to potentially benefit from the treatment with arginase inhibitors due to the increase of NO• levels and

concomitant decrease of polyamines [47]. Highly lipophilic statins, such as atorvastatin, apparently have a greater propensity to cause pleiotropic effects [26, 48] as they easily diffuse across cell plasma membranes achieving a broader biodistribution and, therefore, are more likely to reach off-target regions [49]. Nevertheless, this lipophilicity may also be the reason why off-target effects are less visible in vivo than in vitro [50], as statins are subject to an extensive liver (primary target) first-pass retention [51]. Thus, to bypass hepatic retention and increase the bioavailability to other regions, the development of injectable and inhaled formulations of statins have sparked clinical interest [52]. Recently, a first step towards the evaluation of inhaled statins has been taken with the safety testing in non-human primates, which revealed good tolerance to the nebulization in the airways without evidence of adverse injuries [53].

Many in vitro and in vivo studies have shown promising benefits for using statins beyond cardiovascular diseases, but the still scarce translations to human trials have mostly failed to replicate the same results consistently. Furthermore, additional studies are needed to confirm the relevance of the pathways involved in the capacity of statins to suppress arginase activity and/or increase the NO• levels

(10)

bioavailability, type of statin, treatment duration, or on the sensitivity of the model used.

Conclusion

The pleiotropic effects of statins have been the subject of scientific and clinical interest for several years. These effects are still controversial as their outcomes are not always observed or significant when translated to human clinical trials. One of the most regarded effects of statins, in addition to the primary function of lowering cholesterol for which they are clinically prescribed, is the impact on several small guanosine triphosphatases leading to downstream signals that may upregulate NO•

levels and inhibit arginase. However, a better understanding of all the molecular determinants involved in the statin-induced arginase/NOS signaling pathways that lead to potentially protective effects, such as a reduction of cell proliferation, cytotoxicity, or inflammation, is still needed. Therefore, the use of high resolution and sensitive molecular imaging techniques relying on radiolabeled molecules may have the potential to unveil some of the statin-related mechanisms of action by supporting the development of in vitro quantitative techniques to evaluate cellular and subcellular localization and by mapping the biodistribution profile in complex living organisms.

Although the most commonly used statins in the clinic have comparable LDL-lowering efficacy, highly lipophilic statins, such as atorvastatin, seem to have greater pleiotropic benefits [26, 48]. Additionally, atorvastatin has also been regarded as an effective inhibitor of the permeability glycoprotein [54]. This glycoprotein is highly expressed in many tumor cells, conferring resistance to chemotherapy [55]. Thus, the development of 18F-labeled atorvastatin may add

some relevant value, compared to the recently synthesized [18F]pitavastatin,

[11C]rosuvastatin, or other radiolabeled derivatives of statins not corresponding to

the drug actually used in the clinic [56], for the investigation of the potential capacity of statins to suppress arginase activity and/or increase the NO• levels.

References

[1] R. W. Caldwell, P. C. Rodriguez, H. A. Toque, et al., Physiol Rev, 2018, 98, 641. [2] A. Bhatta, L. Yao, H. A. Toque, et al., PLoS One, 2015, 10, e0127110.

[3] A. Erdely, D. Kepka-Lenhart, R. Salmen-Muniz, et al., PLoS One, 2010, 5, e15253.

[4] a) U. Laufs, V. La Fata, J. Plutzky, et al., Circulation, 1998, 97, 1129; b) N. K. Thakur, T. Hayashi, D. Sumi, et al., Am J Physiol Heart Circ Physiol, 2001, 281, H75; c) O. Feron, C. Dessy, J. P. Desager,

(11)

et al., Circulation, 2001, 103, 113; d) L. Kalinowski, L. W. Dobrucki, V. Brovkovych, et al., Circulation, 2002, 105, 933; e) J. Wang, T. Tokoro, K. Matsui, et al., Life Sci, 2005, 76, 2257; f) C.

Rasmusen, L. Cynober, R. Couderc, Ann Biol Clin, 2005, 63, 443; g) J. Wang, Z. Xu, I. Kitajima, et

al., Int J Cardiol, 2008, 127, 33; h) Y. G. Kaminskii, A. V. Suslikov, L. A. Tikhonova, et al., Biol Bull,

2011, 38, 446; i) L. A. Holowatz, L. Santhanam, A. Webb, et al., J Physiol, 2011, 589, 2093; j) L. A. Holowatz, W. L. Kenney, Am J Physiol Regul Integr Comp Physiol, 2011, 301, R763; k) M. C. Berthe, M. Bernard, C. Rasmusen, et al., Eur J Pharmacol, 2011, 670, 566; l) E. Kosenko, L. Tikhonova, A. Suslikov, et al., J Clin Pharmacol, 2012, 52, 102; m) A. M. Gorabi, N. Kiaie, S. Hajighasemi, et al., J Clin Med, 2019, 8, 2051.

[5] Human Protein Atlas available from http://www.proteinatlas.org.

[6] WHO, available from https://www.who.int/gho/ncd/risk_factors/cholesterol_text/en. [7] a) A. Endo, Nat Med, 2008, 14, 1050; b) A. Endo, Proc Jpn Acad Ser B Phys Biol Sci, 2010, 86, 484. [8] R. Hajar, Heart Views, 2011, 12, 121.

[9] T. Carbonell, E. Freire, Biochemistry, 2005, 44, 11741.

[10] a) C. Stancu, A. Sima, J Cell Mol Med, 2001, 5, 378; b) N. W. Tsai, L. H. Lee, C. R. Huang, et al.,

Crit Care, 2014, 18, R16; c) B. C. Storey, N. Staplin, R. Haynes, et al., Kidney Int, 2018, 93, 1000.

[11] a) S. Jung, t. Kim, H. Kwon, et al., Value Health, 2017, 20, A628; b) J. F. Slejko, A. Basu, S. D. Sullivan, Health Econ, 2018, 27, 282.

[12] a) J. K. Liao, U. Laufs, Annu Rev Pharmacol Toxicol, 2005, 45, 89; b) Q. Zhou, J. K. Liao, Circ J, 2010, 74, 818; c) A. Oesterle, U. Laufs, J. K. Liao, Circ Res, 2017, 120, 229.

[13] a) S. Yuan, mBio, 2015, 6, e01120; b) B. Xiong, C. Wang, J. Tan, et al., Respirology, 2016, 21, 1026; c) N. C. Thomson, Curr Mol Pharmacol, 2017, 10, 60; d) J. Y. So, S. Dhungana, J. J. Beros, et al.,

Curr Opin Pharmacol, 2018, 40, 26; e) A. C. Melo, I. Cattani-Cavalieri, M. V. Barroso, et al., Biomed Pharmacother, 2018, 102, 160.

[14] a) T. P. Ahern, T. L. Lash, P. Damkier, et al., Lancet Oncol, 2014, 15, e461; b) S. Pisanti, P. Picardi, E. Ciaglia, et al., Pharmacol Res, 2014, 88, 84; c) N. S. Nevadunsky, A. Van Arsdale, H. D. Strickler,

et al., Obstet Gynecol, 2015, 126, 144; d) L. Matusewicz, J. Meissner, M. Toporkiewicz, et al., Tumour Biol, 2015, 36, 4889; e) S. Borgquist, O. Bjarnadottir, S. Kimbung, et al., J Intern Med,

2018, 284, 346; f) M. Telfah, T. Iwakuma, A. Bur, et al., J Clin Oncol, 2019, 37, TPS3165. [15] a) B. Verpaalen, J. Neyts, L. Delang, Antiviral Res, 2014, 105, 92; b) O. L. Bryan-Marrugo, D.

Arellanos-Soto, A. Rojas-Martinez, et al., Mol Med Rep, 2016, 14, 2155; c) H. Drechsler, C. Ayers, J. Cutrell, et al., PloS One, 2017, 12, e0172175; d) E. S. Marakasova, B. Eisenhaber, S. Maurer-Stroh, et al., Bioessays, 2017, 39, 1700014; e) P. Shrivastava-Ranjan, M. Flint, É. Bergeron, et al.,

mBio, 2018, 9, e00660; f) S. P. Parihar, R. Guler, F. Brombacher, Nat Rev Immunol, 2019, 19, 104.

[16] a) R. R. Biswas, D. M C, S. Rao A S R, et al., J Clin Diagn Res, 2014, 8, HF01; b) F. C. Lin, Y. S. Chuang, H. M. Hsieh, et al., Medicine, 2015, 94, e2143; c) S. S. Saeedi Saravi, S. S. Saeedi Saravi, A. Arefidoust, et al., Metab Brain Dis, 2017, 32, 949; d) A. J. McFarland, A. K. Davey, C. M. McDermott, et al., Toxicol Appl Pharmacol, 2018, 344, 56; e) H. H. Li, C. L. Lin, C. N. Huang, Neural

Regen Res, 2018, 13, 198.

[17] A. A. Zeki, N. J. Kenyon, T. Goldkorn, Drug Metab Lett, 2011, 5, 40.

[18] a) M. Takemoto, J. K. Liao, Arterioscler Thromb Vasc Biol, 2001, 21, 1712; b) M. Ii, D. W. Losordo,

Vascul Pharmacol, 2007, 46, 1; c) C. Rasmusen, C. Moinard, C. Martin, et al., Br J Nutr, 2007, 97,

1083; d) S. O. Almeida, M. Budoff, Trends Cardiovasc Med, 2019, 29, 451. [19] S. Ryoo, A. Bhunia, F. Chang, et al., Atherosclerosis, 2011, 214, 279.

[20] I. Kosmidou, J. P. Moore, M. Weber, et al., Arterioscler Thromb Vasc Biol, 2007, 27, 2642. [21] Y. Hattori, N. Nakanishi, K. Akimoto, et al., Arterioscler Thromb Vasc Biol, 2003, 23, 176. [22] C. Aoki, A. Nakano, S. Tanaka, et al., Int J Cardiol Heart Vasc, 2012, 156, 55.

[23] W. Sun, T.-S. Lee, M. Zhu, et al., Circulation, 2006, 114, 2655. [24] A. Brouet, P. Sonveaux, C. Dessy, et al., Circ Res, 2001, 89, 866.

[25] C. A. Sánchez, E. Rodríguez, E. Varela, et al., Cancer Invest, 2008, 26, 698. [26] C. H. Beckwitt, K. Shiraha, A. Wells, PLoS One, 2018, 13, e0197422.

(12)

[27] M. Pelat, C. Dessy, P. Massion, et al., Circulation, 2003, 107, 2480. [28] U. Laufs, K. Gertz, P. Huang, et al., Stroke, 2000, 31, 2442.

[29] A. A. Zeki, J. M. Bratt, M. Rabowsky, et al., Transl Res, 2010, 156, 335. [30] H. Erbaş, O. Bal, E. Çakır, Balkan Med J, 2015, 32, 89.

[31] M. Matsumoto, M. Tanimoto, T. Gohda, et al., Metabolism, 2008, 57, 691.

[32] H. E. Tawfik, A. B. El-Remessy, S. Matragoon, et al., J Pharmacol Exp Ther, 2006, 319, 386. [33] M. J. Romero, D. H. Platt, H. E. Tawfik, et al., Circ Res, 2008, 102, 95.

[34] J. Trebicka, M. Hennenberg, W. Laleman, et al., Hepatology, 2007, 46, 242.

[35] J. G. Abraldes, A. Rodríguez-Vilarrupla, M. Graupera, et al., J Hepatol, 2007, 46, 1040. [36] T. Murata, K. Kinoshita, M. Hori, et al., Arterioscler Thromb Vasc Biol, 2005, 25, 2335. [37] L. A. Holowatz, W. L. Kenney, J Physiol, 2011, 589, 4787.

[38] G. A. Plenz, O. Hofnagel, H. Robenek, Circulation, 2004, 109, e7.

[39] a) H. Ju, R. Zou, V. J. Venema, et al., J Biol Chem, 1997, 272, 18522; b) G. García-Cardeña, P. Martasek, B. S. Masters, et al., J Biol Chem, 1997, 272, 25437; c) H. Yokomori, M. Oda, M. Ogi,

et al., Liver, 2001, 21, 198; d) C. Mineo, P. W. Shaul, in Caveolins and Caveolae: Roles in Signaling and Disease Mechanisms, Springer US, New York, 2012, Regulation of eNOS in Caveolae, pp. 51.

[40] A. Cerda, C. M. Fajardo, R. G. Basso, et al., Arq Bras Cardiol, 2015, 104, 195.

[41] a) F. Daghigh, J. M. Fukuto, D. E. Ash, Biochem Biophys Res Commun, 1994, 202, 174; b) J. L. Boucher, J. Custot, S. Vadon, et al., Biochem Biophys Res Commun, 1994, 203, 1614.

[42] J. T. Davies, S. F. Delfino, C. E. Feinberg, et al., Lipid insights, 2016, 9, 13.

[43] M. A. M. Farooqi, N. Malhotra, S. D. Mukherjee, et al., PLoS One, 2018, 13, e0209486. [44] A. Fatehi Hassanabad, Transl Lung Cancer Res, 2019, 8, 692.

[45] a) X. Zhang, J. Wen, Z. Zhang, Medicine, 2018, 97, e11304; b) B. G. Schultz, D. K. Patten, D. J. Berlau, Transl Neurodegener, 2018, 7, 5; c) J. Yan, L. Qiao, J. Tian, et al., Medicine, 2019, 98, e14852.

[46] a) C. Yuan, L. Zhou, J. Cheng, et al., Respir Res, 2012, 13, 108; b) A. A. Zeki, J. Oldham, M. Wilson,

et al., BMJ Open, 2013, 3, e003314; c) A. Walsh, L. Perrem, A. S. Khashan, et al., Cochrane Database Syst Rev, 2019Statins versus placebo for people with chronic obstructive pulmonary

disease, CD011959; d) Y. Lu, R. Chang, J. Yao, et al., Respir Res, 2019, 20, 17; e) N. A. Al-Sawalha, B. J. Knoll, Pharmacology, 2016, 98, 279.

[47] a) M. P. van den Berg, H. Meurs, R. Gosens, Curr Opin Pharmacol, 2018, 40, 126; b) H. Meurs, J. Zaagsma, H. Maarsingh, et al., Allergy, 2019, 74, 1206; c) M. P. M. van den Berg, S. H. Kurhade, H. Maarsingh, et al., J Pharmacol Exp Ther, 2020, 374, 62.

[48] S. Kato, S. Smalley, A. Sadarangani, et al., J Cell Mol Med, 2010, 14, 1180. [49] A. A. Zeki, M. Elbadawi-Sidhu, Expert Rev Respir Med, 2018, 12, 461. [50] A. Moini, G. Azimi, A. Farivar, Allergy Asthma Immunol Res, 2012, 4, 290.

[51] A. G. Brotis, P. P. Tsitsopoulos, T. M. Paraskevi, et al., Interdiscip Neurosurg, 2014, 1, 11. [52] a) P. Bradbury, D. Traini, A. J. Ammit, et al., Adv Drug Deliv Rev, 2018, 133, 93; b) L. M. Tatham,

N. J. Liptrott, S. P. Rannard, et al., Molecules, 2019, 24, 2685.

[53] M. Elbadawi-Sidhu, S. Ott, O. Fiehn, Am J Respir Crit Care Med, 2017, 195, A6459.

[54] a) E. Wang, C. N. Casciano, R. P. Clement, W. W. Johnson, Pharm Res, 2001, 18, 800; b) Management of Statin Treatment in Adult Solid Organ Transplant Recipients, available from https://www.lipid.org/node/2225.

[55] J. H. Lin, M. Yamazaki, Drug Metab Rev, 2003, 35, 417.

[56] a) R. Ijuin, T. Takashima, Y. Watanabe, et al., Bioorg Med Chem, 2012, 20, 3703; b) J. He, Y. Yu, B. Prasad, et al., Mol Pharm, 2014, 11, 2745; c) Y. Yagi, H. Kimura, K. Arimitsu, et al., Org Biomol Chem, 2015, 13, 1113.

(13)

Referenties

GERELATEERDE DOCUMENTEN

Thus, in this chapter is described the synthesis of several structurally different MCR scaffolds, containing both biologically relevant groups (e.g., simple arenes,

The reaction mixture was followed by TLC-SG (10% MeOH in DCM, ninhydrin), washed with bicarbonate, and purified by flash chromatography to yield product 6 (70%)..

The late-stage Cu-mediated radiofluorination strategy has already shown to be very dependent on the type and complexity of the labeling precursor used, and very sensitive to

Blocking assays using rat liver sections with and without standard atorvastatin (12) pretreatment were performed to evaluate [ 18 F]atorvastatin ([ 18 F]12) binding selectivity

However, as the preliminary tests in arginase-overexpressing tumor xenografts and in ex vivo asthmatic lung sections were promising [Chapter 4], and the use of arginase

Aangezien arginase een potentiële biomarker van ziekte is en een nieuw therapeutisch doelwit, werd de hypothese gesteld dat arginase-remmers zouden kunnen worden gebruikt

5 A specific “toolbox” of molecular imaging probes can be used to map arginase expression and assess statin-related mechanisms of action and provide the fundamental basis to

PhD training was facilitated by Research Institute School of Health Research (SHARE), part of the Graduate School of Medical Science Groningen, and by the Graduate Program in