• No results found

Sphingolipids in essential hypertension and endothelial dysfunction - Chapter 1: General introduction

N/A
N/A
Protected

Academic year: 2021

Share "Sphingolipids in essential hypertension and endothelial dysfunction - Chapter 1: General introduction"

Copied!
32
0
0

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

Hele tekst

(1)

UvA-DARE is a service provided by the library of the University of Amsterdam (https://dare.uva.nl)

Sphingolipids in essential hypertension and endothelial dysfunction

Spijkers, L.J.A.

Publication date

2013

Link to publication

Citation for published version (APA):

Spijkers, L. J. A. (2013). Sphingolipids in essential hypertension and endothelial dysfunction.

General rights

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), other than for strictly personal, individual use, unless the work is under an open

content license (like Creative Commons).

Disclaimer/Complaints regulations

If you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please

let the Library know, stating your reasons. In case of a legitimate complaint, the Library will make the material

inaccessible and/or remove it from the website. Please Ask the Library: https://uba.uva.nl/en/contact, or a letter

to: Library of the University of Amsterdam, Secretariat, Singel 425, 1012 WP Amsterdam, The Netherlands. You

will be contacted as soon as possible.

(2)

General introduction

hypertension | endothelial dysfunction | sphingolipids

(3)

10

Hypertension as a risk factor

The high spatial volume of the human body exceeds the limited passive oxygen diffusion toward organs and tissue and therefore demands the existence of a cardiovascular system. The vasculature comprises an interconnected network of blood vessels with decreasing diameter to ensure the supply of oxygen to peripheral tissues. To ascertain adequate perfusion, a pressure gradient is needed. Although many feedback systems exist within the cardiovascular system to maintain blood pressure (BP) within appropriate limits (Table 1), clearly these are insufficient in the persistent pathological condition of high BP (hypertension).

Hypertension complications are related to poor BP control, and can ultimately lead to end-stage organ damage, cerebrovascular accidents (e.g. stroke) and other malignancies as summarized in Table 2. Hypertension is recognized as the quantitatively largest risk factor for cardiovascular diseases. Approximately half of all strokes and one third of ischemic heart diseases is attributable to hypertension 1. Furthermore, a log linear correlation exists between elevated

arterial pressure and increased mortality 2. Worldwide population hypertension prevalence is

increasing, and estimated around 30% (2003) 3, in the USA specifically around 28.9%

(2004) 4,5 and in the Netherlands around 13.5% (2009) 6. Next to low treatment efficacy,

hypertension is additionally increasing as a result of higher adiposity and western diet, sedentary life style and aging population (Figure 1).

Blood pressure ESH-ESC / WHO-ISH

SBP DBP

Optimal <120 <80

Normal (SBP and DBP) 120-129 80-84

High normal/prehypertension (or) 130-139 85-89

Stage/grade 1 hypertension (or) 140-159 90-99

Stage/grade 2 hypertension (or) 160-179 100-109

Stage/grade 3 hypertension ≥180 ≥110

Systolic blood pressure (SBP), diastolic blood pressure (DBP).

Table 1. Classification of blood pressure according to the European Society of

Hypertension-European Society of Cardiology (ESH-ESC) and the World Health Organization-International Society of Hypertension (WHO-ISH).

(4)

11

Hypertension can be separated into two major categories. In approximately 95% of all cases of hypertension, the exact mechanism involved in the pathogenesis of elevated BP is poorly understood. This represents the essential hypertension group, characterized by a largely unidentified polygenic trait 7 influenced by probably hundreds of different genes as well as

environmental factors 8. The other 5% of hypertension cases is denoted as secondary

hypertension, in which elevated BP is secondary to known initiators like: renal disease 9,

endocrine disorders 10, drug-usage (e.g. contraceptives 11, antidepressants and

others 12. Affected systems that are associated with essential hypertension include elevated

sympathetic neural activity 13,14 and tissue angiotensin II activity 15. Several hypertensinogenic

factors have been distilled from epidemiological and large-scale clinical studies implicated in the onset of essential hypertension. These comprise in part; high alcohol 16 or salt intake 17, vitamin

D deficiency 18, obesity and dyslipidemia 19,20, low physical activity 21,22 and genetic background

(ethnicity 23 and parental hypertension 24). About 1% of these patients develop malignant

hypertension.

Structure Pathology

Heart heart failure, myocardial ischemia/infarction

Aorta aortic aneurysm/dissection, stroke

Cerebral/carotid stroke

Kidney nephrosclerosis/renal failure

Eye retinopathy

Figure 1. Percentage of hypertension prevalence in the Netherlands in 2001 and 2009.

Expressed as percentage (%) of total Dutch population, subsequently subdivided by age subsets. (Adapted from CBS; 2011; NL)

(5)

12

Regulation of blood pressure

Blood pressure is the primary outcome of both cardiac output and total peripheral resistance. Many downstream systems influence these two parameters 7,25. Total peripheral resistance has

a direct effect on blood flow and is principally determined by the diameter of small arteries and arterioles. These vessels display profound structural and functional changes in hypertension 26,

as characterized by increased contractility and hypertrophy 27. Clearly, the balance between

blood vessel contraction and relaxation determines the net resistance, or luminal diameter. As indicated by Poiseuille’s equation of flow (F)= (ΔP•r4)/(Ƞ•L) wherein the blood vessel radius (r) is

to the fourth power, any decrease in vessel diameter has a potent inhibitory effect on blood flow, thus making peripheral resistance a powerful BP regulator. Vasomotor tone, in conjunction with structural vascular remodelling, determines the vessel diameter which is orchestrated by both extrinsic and intrinsic regulation. Extrinsic regulators encompass: vasomotor nerves (sympathetic, parasympathetic, C-type sensory nerves) and systemic hormones (e.g. angiotensin II) 28. Intrinsic regulators comprise: the Bayliss myogenic response,

vasoactive metabolites (from active tissue, e.g. lactate, CO2, K+), temperature, autacoids (e.g.

prostaglandins) and release of other endothelium-derived factors 29. As indicated by the latter

mentioned factor, indeed, the endothelium plays an important role in vasomotor tone regulation 30. The endothelium forms the inner monolayer (part of the tunica intima) of all

blood vessels. It is the active regulator of vascular homeostasis involving: vasomotor tone, coagulation, pro/anti-inflammation and remodelling 31. Vasomotor tone regulation by the

endothelium is exerted by the secretion of endothelium-derived relaxing (EDRF) and endothelium-derived contractile factors (EDCF) 32. The main EDRFs involved are: nitric oxide

(NO) 33,34, prostacyclin (PGI

2) 35,36 and endothelium-derived hyperpolarizing factor (EDHF) 37,38.

Under physiological conditions, intra-endothelial calcium elevation leads to activation of endothelial nitric oxide synthase (eNOS) to generate NO, phospholipase A2 (PLA2) to generate

arachidonic acid (the precursor for e.g. PGI2) and increased calcium-activated potassium

channel currents to contribute to the classical 39 EDHF response. The main EDCF involve

endothelin-1 (ET-1) 40,41 and thromboxane A

2 (TXA2) 42. Both are agonists for different

(contractile) G-protein coupled receptors (see Box 1) on VSMCs, resulting in this case in smooth muscle calcium increase, myosin light chain kinase (MLCP) activation and concomitant contraction.

(6)

13

Hypertension and endothelial dysfunction

Hypertension is associated with endothelial dysfunction 43,45. Although the ‘high normal’ BP

classification (Table 1) implies a trivial condition within a physiological normal range, also prehypertension is an independent risk factor for endothelium-dysfunction and cardiovascular events as found in hypertensive patients 46-49. In endothelial dysfunction, the vascular balance is

shifted towards; impaired relaxation, augmented contraction, vascular remodelling and inflammation 27,50. Importantly, investigations have indicated a larger incidence of

cardiovascular events in hypertensive patients with more severe endothelial dysfunction compared to hypertensive patients with less severe endothelial dysfunction, and therefore it is suggested as a marker for future cardiovascular events in hypertensive patients 51.

Many findings have fuelled the discussion of whether endothelial dysfunction precedes hypertension or vice versa, however until now it remains an etiologically-undefined association with hypertension. An important finding is that the normotensive offspring of hypertensive

Figure 2. Mechanisms implicated in essential hypertension-associated endothelial dysfunction. Endothelial cell (EC), von Willebrand Factor (vWF), Weibel-Pallade body (WPB),

endothelin-1 (ET-1), endothelin-converting enzyme (ECE), nitric oxide (NO), prostaglandin (PG), thromboxane (TX), cyclooxygenase-1 (COX-1), prostaglandin synthases (PGS), endothelial nitric oxide synthase (eNOS), reactive oxygen species (ROS), endothelin A receptor (ETA), thromboxane/prostanoids receptor (TP), vascular smooth muscle cell (VSMC).

(7)

14

parents also display impaired endothelium-dependent relaxation, possibly implying endothelial dysfunction as a condition that could prelude hypertension 52,53, although also evidence for a

hypertension-dependency has been presented as well 54,55. Consent has been gained on which

different effectors are pivotal in essential hypertension-associated endothelial dysfunction. These mainly include decreased NO bioavailability 56,57, augmented prostanoid-production and

subsequent activation of the G-protein coupled thromboxane/prostanoid (TP) receptor (see Box 1) 58,59, elevated contractile ET-1 signalling 60, elevated reactive oxygen species (ROS)

production 61, high endothelial expression/excretion of vWF and other adhesion molecules 62, as

schematically depicted in Figure 2. These factors have been implicated in elevated vascular contractility and hypertrophy, decreased barrier function and a inflammatory and pro-thrombotic phenotype. Indeed, endothelial dysfunction is currently recognised as a priming condition for future arteriosclerosis and thrombosis formation 63-65. Next to hypertension, also

other conditions including diabetes mellitus 66, post-menopause 67, smoking 68,

hypercholesterolemia 69,70 and aging itself are associated with endothelial dysfunction 71,

although varying causal effectors are implicated in the endothelial dysfunction between different conditions.

Pharmacological intervention and endothelial dysfunction

The concept of treating hypertension as a single disease state with a single type of drug has been proven obsolete, mainly due to the plethora of causal factors contributing to the elevated BP, and which differ strongly between patients 72. Thus, a vast array of antihypertensive drugs

(73-78)

Box 1 | G-protein coupled receptors (GPCR)

These 7-transmembrane receptors form the largest cell surface receptor super family, and are coupled with their cytosol-directed C-terminal tail to G-proteins. G-proteins consist of a GDP-GTP binding α-subunit and βγ-α-subunit complex. Receptor activation induces a conformational change within the receptor structure, and dissociation of α and βγ-subunits as part of the signal transduction. Depending on which Gα-subunit endogenously couples to the GPCR, signaling events can be initiated by: Gαi/o (adenylyl cyclase inhibition, Ca2+ channel inhibition, K+ channel activation, phosphodiesterase

stimulation), Gαs (adenylyl cyclase stimulation), Gα12/13 (small G-protein stimulation, like RhoGEF) and Gαq/11 (phospholipase C activation). Via βγ-subunit dissociation, several signaling pathways can be initiated possibly involving activation of MAPK, PI3K, PLCβ, calcium flux and adenylyl cyclase.

Importantly, βγ-subunit dissociation leads to receptor phosphorylation by GRK and subsequent recruitment of β-arrestin, leading to diverse signaling cascades, receptor internalization and eventually receptor degradation or recycling. The efficacy and outcome of GPCR-mediated signaling is depended, but not restricted to; the specific subunit coupling, the receptor-activating ligand (ligand-directed signaling), presence of allosteric / orthosteric modulators and the cellular G-protein profile 73-78

.

(8)

15

have been developed, e.g. the angiotensin signalling inhibitors (angiotensin-converting enzyme inhibitors and angiotensin II receptor antagonists), the beta-adrenergic receptor antagonists (beta-blockers), calcium-channel inhibitors and diuretics. The broad diversity of proteins targeted by these drugs point towards the complex heterogeneous nature of hypertension. According to the Guideline Development Group (2007), mono-therapy can be initiated in an attempt to achieve BP lowering in uncomplicated hypertension 79. However, multi-drug therapy

can be initiated if BP remains uncontrolled. Next to pharmacological intervention and clinical intervention (e.g the novel renal nerve ablation); life style modifications are also of significant value to cumulatively achieve optimal BP control 80. In this respect, of interest is a current

dispute regarding ‘optimal’ BP (Table 1). In the hypothesis “the lower the better”, a possible J-curved relationship between BP and cardiovascular outcome indicates otherwise, as the relative risk for several cardiovascular morbidities is proposed to increase below a certain (treatment-induced) BP threshold, and several contributing factors have been postulated for this phenomenon 81-84.

As indicated before, next to BP control, restoration of endothelial dysfunction remains crucial in preventing hypertension-associated end organ damage. The causality of endothelial function is arbitrary, as exemplified by several studies indicated unaffected endothelial dysfunction in hypertensive patients ‘successfully’ treated with several classes of BP lowering drugs and with BP within normal ranges 85. BP lowering drugs, including angiotensin-converting enzyme (ACE)

inhibitors and AT1 blockers, are able to improve endothelium-dependent relaxation in several

vascular beds in rat 86-88 and human 89 essential hypertension. This drug-class is able to reduce

ROS and ET-1 production and decrease bradykinin degradation, therefore augment NO bioavailability and vascular relaxation. These effects all contribute to improved endothelium-derived relaxation in hypertension. Still, other studies indicated no significant effect of the latter drug treatment on EC function 90,91. Clearly, the persistent attrition of drug-efficacy in

preventing hypertension and hypertension-associated end-organ damage in many patients indicates the necessity to explore novel drug targets. Although new compounds targeting relevant ion channels, receptors and enzymes involved in the pathology of hypertension can be expected, this thesis outlook proposes the bioactive lipid class of ‘sphingolipids’ as a novel target in hypertension and hypertension-associated endothelium dysfunction.

(9)

16

Sphingolipids

Structure and generation

Lipids form a broad family of hydrophobic or aliphatic molecules that make up for about 50% of the total mass of most animal cell membranes and are crucially involved in the formation of biological structures and maintenance of homeostasis 92. Overall in biological systems,

endogenous lipid species can be divided into eight categories: fatty acids (e.g. arachidonic acid), glycerolipids (e.g. triglycerides), phospholipids (e.g. phosphatidylcholine), saccharolipids (e.g. lipo-polysaccharide), polyketides (e.g. tetracycline), sterol lipids (e.g. cholesterol), prenol lipids (e.g. vitamin E) and sphingolipids (e.g. sphingomyelin) 93. Mammalian cells contain between

1000-2000 different lipid species 94, predominantly including sterols, glycerolipids and

sphingolipids 95. Since the discovery of sphingomyelin being a major constituent of brain tissue

by Thudichum in 1884, many sphingomyelin metabolites have been described, like ceramide,

Figure 3. The sphingomyelin cycle, depicting the ceramide-S1P rheostat. Proteins involved in lipid

metabolism: sphingomyelinase (SMase), sphingomyelin synthase (SMS), ceramidase (CDase), ceramide synthase (CerS), sphingosine kinase (SK), sphingosine-1-phosphate phosphatase (SPPase). Sphingoid base indicated by the dotted line box 96.

(10)

17

Box 2 | Fatty acids

The fatty acid (carboxylic acid) tail of a large subset of sphingolipids, have a high variety in length and double carbon bond position. In general, fatty acids form a much wider family than those found to be structurally linked to sphingolipids. Fatty acids can differ in length (generally even-numbered), double bond number (monoenoic or polyenoic) and position and linkage to other chemically-distinct substituents. The longer the chain length, the more pronounced lipophylic behaviour it displays, although coupling to phosphate groups or other hydrophylic groups alter the lipophylicity as well. Fatty acids are considered short-chained up to C6, long-chained from C12 and very-long chained from C22. They can be subdivided in straight chain fatty acids (either saturated; e.g. eicosanoic fatty acids), monounsaturated (MUFA; e.g. nervonic acid), polyunsaturated (PUFA; e.g. omega-3 fatty acid) and acetylenic), more chemically-diverse straight chain fatty acids (e.g. 2-hydroxy fatty acids), branched chain fatty acids (e.g. tuberculostearic acid), cyclic fatty acids and fatty aldehydes/alcohols. Still, only a small subset of fatty acids have been described to naturally occur in the mammalian system 102.

sphingosine and sphingosine-1-phospate, forming the major lipid class of sphingolipids (Figure 3). Sphingolipids are scaffolded by a sphingoid base, often linked via amide bonds to fatty acid chains of varying length. This fatty acid chain consists of up to C26 of saturated or monoenoic (unsaturated) components, however chain lengths up to C36 have been identified in specific cell

types (see Box 2). Although early Identified as structural membrane constituents, the recognition of sphingolipids being involved in numerous signalling events has been established only recently.

Sphingomyelin, firstly discovered to reside in high abundance in the myelin sheet surrounding neuronal cells, is a ubiquitous membrane lipid of virtually all cells. It is composed of a sphingoid base, linked to a fatty acid chain and phosphocholine head group (Figure 3). Many enzyme subclasses have been recognized to modify sphingomyelin towards several sphingomyelin metabolites. These sphingomyelin metabolites make up for the sphingomyelin cycle, in which the first step is comprised of sphingomyelin hydrolysis by sphingomyelinases (SMase) to yield ceramide and phosphorylcholine 97. Ceramide consists of sphingosine coupled to a fatty acid

chain. Three major subtypes of sphingomyelinases have been characterized, all discriminated by their pH sensitivity and situated within varying cellular compartments, namely; acid (endosomes/lysosomes), neutral (ER/golgi), and alkaline sphingomyelinases 98. Especially neutral

and acid SMases have been implicated in cellular regulation, whereas alkaline SMase is more, but not exclusively, restricted to intestinal digestion of dietary sphingomyelin 99-100. Activation of

SMases is often involved in stress-induced signalling cascades, and initiators of SMase activation include: γ-irradiation, hypoxia, ROS, oxidized low density lipoproteins (oxLDL), tumour-necrosis factor (TNF) α, and shear stress (for review see 101). Subsequent ceramide formation has been

associated with a myriad of signalling events, of which apoptosis induction is primarily recognised and implicating ceramide as a non-trivial second messenger. 2

(11)

18

Several synthesis and degradation pathways have been described for ceramide to tightly regulate its cellular abundance. For instance, ceramide can be phosphorylated by ceramide kinase to ceramide-1-phosphate (C1P) 103. Following de novo synthesis of ceramide in the

endoplasmic reticulum (ER) 104-107, ceramide can be transported towards the Golgi apparatus via

probably the ceramide transfer protein CERT for further processing 108. In the early Golgi

system, ceramide can be subsequently glycosylated by glucosyltransferases to yield glucosylceramide (GlcCer; a glucocerebroside) or by galactosyltranferases to yield galactosylceramide (GalCer; a galactocerebroside) 109. Non-vesicular transport of GlcCer

towards late Golgi, probably by the transport protein FAPP2, enables the generation of more complex glycosphingolipids including lactosylceramide and higher glycosphingolipids 96-110. In

contrast to the generation of these complex glycosphingolipid species, ceramide can also be deacylated towards sphingosine by ceramidases 111-112. As for sphingomyelinases; ceramidases

are discriminated by their pH dependency including: acid (endosomes/lysosomes), neutral (mitochondria), and alkaline (ER) ceramidases. Ceramidase activation has been shown to be induced by several growth factors including platelet-derived growth factor, insulin-like growth factor and basic fibroblast growth factor. Sphingosine is a substrate for sphingosine kinase (SK) 1 and 2, and upon enzyme activation readily phosphorylated to sphingosine-1-phosphate (S1P) 113. Although both sphingosine kinases yield S1P, the net outcome on cell fate differs

between the two enzymes. SK2 activation contributes to a pro-apoptotic environment, most likely due to a alternative cellular localisation compared to SK1 114. Both phosphorylation and

translocation are major regulators of SK actions. Pitson et al. showed that SK1 phosphorylation at Ser225 by ERK1/2 115 resulted in increased catalytic activity and translocation of SK1 from the

cytosol to the plasma membrane to exert functional activity 116. SK1 translocation to various

subcellular locations largely depends on the specific receptor/agonist complex that activates SK 117. This implies an importance of not only the total presence of specific sphingolipids, but

also the specific subcellular location of synthesis/transport, as described by Pyne et al. 118.

Activators of sphingosine kinase are vast and include growth factors, second messengers and cytokines (for review see 119). After production and signaling events, S1P can subsequently be

degraded by S1P lyase, which yields ethanolaminephosphate and hexadecenal 120. S1P can be

also dephosphorylated by sphingosine-1-phosphate phosphatases (SPP) to yield sphingosine again 121, possibly serving as a buffer for future S1P requirement. Insight in signalling cascades

involving S1P has expanded greatly and S1P has often been implicated in counteracting ceramide-signalling. The lipid interchangeability between ceramide and S1P, grants a

(12)

19

Figure 4. Non-exhaustive representation of sphingolipid metabolism pathways.

Sphinganine-1-phosphate (Sa1P), sphingosylphosphorylcholine (SPC), sphingosine-1-Sphinganine-1-phosphate (S1P), lysophosphatidylcholine (LPC), ceramide-1-phosphate (C1P), glucosylceramide (GlcCer), galactosylceramide (GalCer), lactosylceramide (LacCer), higher order glycosphingolipids (derived from LacCer and GalCer). Predominant sphingolipid synthesis location: endoplasmic reticulum (■) and golgi apparatus (○).

physiologically active regulation system implicated in, for instance; the cardiovascular system, immune system, tumour onset and progression and neurogenesis 122. Signalling events,

mediated by S1P, are often initiated via binding to, and subsequent activation of its specific pleiotropic S1P receptors. In the process of autocrine activation of S1P receptors, one report suggested that, at least in mast cells, synthesized S1P could be transported outside of the cell through the plasma membrane via ATP-binding cassette (ABC) transporters, and consequently

target S1P receptors 123. Next to plasma membrane receptor targeting, there are also

indications for intracellular targets for S1P 124,125. As to date, S1P is the sole sphingolipid

involved in the in figure 3 depicted sphingomyelin-cycle for which receptor coupling has been identified, although indications for novel sphingolipid receptors have been suggested for e.g. SPC 126,127 and C1P 128. In addition to S1P, lysophosphatidic acid (LPA) is another well-defined

(13)

20

lysophospholipid (review see von Meyer zu Heringdorf 129, which was previously believed to

share an equal affinity with S1P for ten lysophospholipid G-protein coupled receptors denoted as EDG-1 to EDG-10. Upon the recognition of a different affinity of S1P and LPA for specific EDG receptors, these receptors have been deorphanized and reclassified according to the IUPHAR guidelines, presently accounting for five S1P receptors (S1P1-5) and five LPA receptors

(LPA1-5) 130,131.

Sphingolipids in cardiovascular regulation

Many sphingomyelin-metabolites have been recognised to circulate in the blood 132. Platelets

express high levels of sphingosine kinase, with low levels of S1P lyase 133. Consequently, these

cells contain high levels of S1P, which is readily released into the circulation upon platelet-activation. Next to platelets, erythrocyte-derived and endothelial cell-derived S1P are the major contributors to S1P presence in the blood 134,135. As a consequence, direct interaction of these

sphingolipids with the endothelium forecasts a cardiovascular signalling potential. Indeed, experimental and clinical evidence is increasingly provided for sphingolipid mediation like S1P and ceramide in maintaining cardiovascular health.

Ceramide functions in the cardiac system are slowly being charted and several cardiac pathologies have been associated with ceramide alterations. For instance, in a model of cardiac lipotoxicity, elevated ceramide presence in cardiac tissue has been postulated to mediate some of the adverse phenotypical changes 136. Moreover, pharmacological inhibition of ceramide

synthesis in this model improved cardiac function and survival. Hence ceramide could be regarded as a cardiotoxin. In a study by Furuya et al., however, exogenous ceramide administration protected against ischemia-induced cerebral infarction and, although performed in cerebral tissue, it could be worthwhile to elaborate on the exact function of ceramide in various cardiac pathophysiological settings 137. It is noteworthy though, that the latter study

was based on the administration of short chain (C2 and C8) cell permeable ceramide species. The beneficial effects of reduced infraction size could be due to further metabolism towards other sphingolipids S1P instead of the administered or further elongated ceramide species. The feasibility of this principle was provided by a study in which addition of C2 ceramide to neuronal cells instantly led to a conversion to sphingosine and S1P 138 and vice versa, thus these

findings mandate a reasonable caution in proving causal effects for specific sphingolipids when administered. For S1P, several studies indicate a cardiac protective role due to its ability to

(14)

21

Figure 5. Major S1PR signaling cascades in the cardiac ( ) and vascular ( ) system. Predominant

G-protein coupling indicated by gray arrows. Although theses arrows indicate a preference of specific receptor-G-protein interaction induced by S1P, a multiplicity of coupling at G-proteins exist, and is determined by several factors like; receptor phosphorylation, G-protein availability, compartmentalization, activity of Regulators of G-proteins (RGS) and several other factors 139-143. These

factors are of influence on the specific downstream effector activation.

activate putative pro-survival and proliferative signalling cascades like for instance eNOS 139 (Fig.

5). Although this scheme clearly depicts the exact role of tissue S1P receptor activation by S1P administration or endogenous inhibition, the functional contribution of plasma S1P to this is

still largely unknown. The majority of S1P-induced effects are mediated by S1P receptors. Although S1P1-5 receptors are present in the mammalian system, the cardiac system expresses merely S1P1, S1P2 and S1P3 receptors, with predominantly S1P1 in cardiomyocytes and S1P3 in

(15)

22

cardiac fibroblasts. These receptors have been shown to be involved in cardiac function 139-143

.

It is postulated by Egome et al. that S1P can exert its cardiac protective actions against ischemia via platelet-juxtacrine uptake of cardiomyocyte-secreted sphingosine, which is subsequently released as newly formed S1P. This S1P can subsequently activate cardiac S1P receptors and concomitantly eNOS 144. Still, another study indicated that patients suffering from myocardial

infarction displayed reduced plasma S1P levels directly after infarction, suggesting an inability of S1P to exert cardioprotection, and this low level was maintained even after the following 5 days 145. Quantification of S1P in plasma of patients with chronic systolic heart failure (primarily

due to ischemic heart disease) demonstrated no alterations in plasma S1P levels, although plasma sphingosine was significantly lowered 146. This would argue against putative beneficial

effects of S1P on cardiac protection after infarction, however, a more detailed study by Sattler et al. could have proven otherwise. This study indeed confirmed a decrease of total free S1P in plasma of patients with acute myocardial infarction. However within a 2-12 hour time-span after infarction, a transient but significant increase in total plasma S1P was detected, with no indication for interference of the clinical intervention, like anticoagulants, on S1P levels 147. It is

likely that due to a predicted high turnover rate for circulating S1P, as indicated by the rapidly decreased plasma S1P values after SK inhibition in vivo 148, the detection window for S1P

alterations is narrow and this possibly contributes to conflicting reports on S1P involvement in cardiac pathologies. Thus, S1P elevation may still provide an endogenous cardiac protection mechanism, albeit temporary, which may even be extrapolated to basal maintenance, since trivial initiators like physical activity were also shown to rapidly elevate plasma S1P 149. Next to

platelet-derived S1P, HDL has been suggested to be a carrier for S1P, since the vast majority of circulating S1P exists bound to either HDL or albumin 150. Indeed, the advantageous effects of

HDL to cardiovascular health have be partly attributed to its S1P component, which upon HDL binding to the scavenger receptor B type I (SR-BI) is postulated to activate endothelial S1P receptors and subsequently eNOS 151. Still, ischemic heart disease patients were found to have

lowered S1P content in HDL, irrespectively of HDL level alterations 152, and especially

non-HDL-bound plasma S1P was found to be elevated in myocardial infarction patients 147, implying that

the beneficial effects of HDL-S1P may poorly contribute to basal cardiovascular maintenance. Although (HDL)S1P-induced NO production may be mediated specifically via endothelial S1P3

receptors, also S1P1 receptor activation has been shown to stimulate NO production 153. Of

interest in this respect is the recent documentation of the crystal structure of S1P1 receptors.

(16)

23

gap close to the plasma membrane that may be easily accessible for membrane-associated S1P

154. This implies that circulating S1P may present significantly slower binding kinetics over

endogenously produced S1P. As such, these structural characteristics provide discriminated signalling kinetics for S1P depending on the delivery route. The importance of S1P signallingin cardiovascular maintenance 155 has been further exemplified in S1P receptor knockout models.

In a mouse model, S1P1 receptor knockout appears to be embryonically lethal, in concordance

with SK1/SK2 double-knockout, due to defects in vascular maturation and neurogenesis. Indeed, S1P1 functions as a vascular network stabilizer, and abrogation of S1P1 signalling causes cellular adherence instability, loss of barrier function and abnormal vascular development 156 In order to investigate S1P

1 signalling in vivo, this led to the generation of

tissue-specific conditional knockout models 157,158. From these models, a clear deficiency in

proper development of the tunica media or pericytes enclosing to embryonic blood vessels became present, thus pointing out the pivotal role of S1P1 in vascular maturation. Although

S1P2 receptor knockout models display less pronounced phenotypic abnormalities,

characterised by normal BP and cardiac function, specific vascular homeostatic regulation defects are present 159,160. These models display cochlear vascular impairment, which is a major

contributor to the inborn deafness 161, decreased vascular resistance and blunted contractile

responses 162. Furthermore, a pathophysiological role for S1P

2 signalling was detected in a

mouse model of heart failure. The intrinsic attempt to maintain BP in heart failure is a redundant elevation of vascular myogentic tone, which actually contributes to further cardiac output reduction due to increased afterload. These elevated contractile responses appeared strongly dependent on the S1P-S1P2 signalling axis 163. As with S1P2 receptor knockout, S1P3

receptor knockout is not associated with embryonic lethality, but partial loss of cardiac chronotropic regulation and vasomotor tone regulation has been reported 164.

In the vascular system, the endothelium predominantly expresses S1P1 receptors, but also S1P3

and low levels, often restricted to particular vascular beds, of S1P2 165,166. Vascular smooth

muscle cells, however, express predominantly S1P2 and S1P3, with lower levels of S1P1 166,167

and with a certain plasticity in receptor expression 168, as exemplified by absent detection of

S1P3 in cultured VSMCs 169. Several factors differentially influence endothelial S1P receptor

expression (e.g. VEGF, ROS), and thus alter S1P-specific vascular regulation 170-173. The

vasomotor control effects of sphingolipids are possibly too elusive to generalise. For S1P, many results point towrads induction of contractile responses in mainly resistance vessels and

(17)

24

vasorelaxation or less pronounced effects in conduit vessels 174. For example, effects of S1P

include vasoconstriction in mesenteric artery 175, human placental artery 176 and rat cerebral

artery 177, as well as vasorelaxation of aorta 178 and coronary artery 179. Pharmacological

elevation of ceramide induced contractions of canine cerebral artery 180. Ceramide contributed

to agonist-induced contractions in rat and human pulmonary artery181. In rat aorta and

mesenteric arteries, however, ceramide induced vasorelaxation 182, 183. Overall, depending on

the pathophysiological condition or specific receptor expression of vascular beds, many sphingolipid species have been appointed Janus-faced properties in vasomotor control. Most of the latter reported effects have been generated using exogenous sphingolipids. In Chapter 2 we focus on endogenous sphingolipid crosstalk in specifically endothelial cells in mediating vasoactive factor secretion, partly with respect to endothelial dysfunction.

(18)

25

References

1. Lawes CM, Vander HS, Rodgers A. Global burden of blood-pressure-related disease, 2001. Lancet 2008; 371(9623):1513-1518.

2. Pickering G. Hypertension. Definitions, natural histories and consequences. Am J Med 1972; 52(5):570-583.

3. Kearney PM, Whelton M, Reynolds K, Whelton PK, He J. Worldwide prevalence of hypertension: a systematic review. J Hypertens 2004; 22(1):11-19.

4. Cutler JA, Sorlie PD, Wolz M, Thom T, Fields LE, Roccella EJ. Trends in hypertension prevalence, awareness, treatment, and control rates in United States adults between 1988-1994 and 1999-2004. Hypertension 2008; 52(5):818-827.

5. Lloyd-Jones D, Adams R, Carnethon M, De SG, Ferguson TB, Flegal K, Ford E, Furie K, Go A, Greenlund K, Haase N, Hailpern S, Ho M, Howard V, Kissela B, Kittner S, Lackland D, Lisabeth L, Marelli A, McDermott M, Meigs J, Mozaffarian D, Nichol G, O'Donnell C, Roger V, Rosamond W, Sacco R, Sorlie P, Stafford R, Steinberger J, Thom T, Wasserthiel-Smoller S, Wong N, Wylie-Rosett J, Hong Y. Heart disease and stroke statistics--2009 update: a report from the American Heart Association Statistics Committee and Stroke Statistics Subcommittee. Circulation 2009; 119(3):e21-181. 6. Centraal Bureau voor Statistieken (CBS). 2012. 2011. Ref Type: Online Source 7. Weder AB. Genetics and hypertension. J Clin Hypertens (Greenwich ) 2007;

9(3):217-223.

8. Fenger M, Linneberg A, Jorgensen T, Madsbad S, Sobye K, Eugen-Olsen J, Jeppesen J. Genetics of the ceramide/sphingosine-1-phosphate rheostat in blood pressure regulation and hypertension. BMC Genet 2011; 12:44.

9. Bakris GL, Ritz E. The message for World Kidney Day 2009: hypertension and kidney disease: a marriage that should be prevented. J Am Soc Hypertens 2009; 3(2):80-83. 10. Sukor N. Secondary hypertension: a condition not to be missed. Postgrad Med J 2011;

87(1032):706-713.

11. Lim KG, Isles CG, Hodsman GP, Lever AF, Robertson JW. Malignant hypertension in women of childbearing age and its relation to the contraceptive pill. Br Med J (Clin Res Ed) 1987; 294(6579):1057-1059.

12. Grossman E, Messerli FH. Drug-induced hypertension: an unappreciated cause of secondary hypertension. Am J Med 2012; 125(1):14-22.

(19)

26

13. Esler MD, Jennings GL, Lambert GW. Release of noradrenaline into the cerebrovascular circulation in patients with primary hypertension. J Hypertens Suppl 1988; 6(4):S494-S496.

14. Grassi G. Role of the sympathetic nervous system in human hypertension. J Hypertens 1998; 16(12 Pt 2):1979-1987.

15. Redgrave J, Rabinowe S, Hollenberg NK, Williams GH. Correction of abnormal renal blood flow response to angiotensin II by converting enzyme inhibition in essential hypertensives. J Clin Invest 1985; 75(4):1285-1290.

16. Beilin LJ, Puddey IB. Alcohol and hypertension: an update. Hypertension 2006; 47(6):1035-1038.

17. Adrogue HJ, Madias NE. Sodium and potassium in the pathogenesis of hypertension. N Engl J Med 2007; 356(19):1966-1978.

18. Burgaz A, Orsini N, Larsson SC, Wolk A. Blood 25-hydroxyvitamin D concentration and hypertension: a meta-analysis. J Hypertens 2011; 29(4):636-645.

19. De SG, Devereux RB, Chinali M, Roman MJ, Best LG, Welty TK, Lee ET, Howard BV. Risk factors for arterial hypertension in adults with initial optimal blood pressure: the Strong Heart Study. Hypertension 2006; 47(2):162-167.

20. Sonne-Holm S, Sorensen TI, Jensen G, Schnohr P. Independent effects of weight change and attained body weight on prevalence of arterial hypertension in obese and non-obese men. BMJ 1989; 299(6702):767-770.

21. Forman JP, Stampfer MJ, Curhan GC. Diet and lifestyle risk factors associated with incident hypertension in women. JAMA 2009; 302(4):401-411.

22. Carnethon MR, Evans NS, Church TS, Lewis CE, Schreiner PJ, Jacobs DR, Jr., Sternfeld B, Sidney S. Joint associations of physical activity and aerobic fitness on the development of incident hypertension: coronary artery risk development in young adults. Hypertension 2010; 56(1):49-55.

23. Egan BM, Zhao Y, Axon RN. US trends in prevalence, awareness, treatment, and control of hypertension, 1988-2008. JAMA 2010; 303(20):2043-2050.

24. Wang NY, Young JH, Meoni LA, Ford DE, Erlinger TP, Klag MJ. Blood pressure change and risk of hypertension associated with parental hypertension: the Johns Hopkins Precursors Study. Arch Intern Med 2008; 168(6):643-648.

25. Hypertension. In: Lilly L.S., editor. Pathophysiology of heart Disease. Wolters Kluwer|Lippincott Williams & Wilkins, 2007: 311-333.

(20)

27

26. Mulvany MJ. Small artery remodeling in hypertension. Curr Hypertens Rep 2002; 4(1):49-55.

27. Intengan HD, Schiffrin EL. Vascular remodeling in hypertension: roles of apoptosis, inflammation, and fibrosis. Hypertension 2001; 38(3 Pt 2):581-587.

28. Control of blood vessels II: extrinsic control. In: Rodney Levick, editor. An Introduction to Cardiovascular Physiology. Hodder Arnold, 2003: 232-250.

29. Control of blood vessels I: intrinsic control. In: Rodney Levick, editor. An Introduction to Cardiovascular Physiology. Hodder Arnold, 2003: 217-231.

30. De Mey JG, Vanhoutte PM. Heterogeneous behavior of the canine arterial and venous wall. Importance of the endothelium. Circ Res 1982; 51(4):439-447.

31. Galley HF, Webster NR. Physiology of the endothelium. Br J Anaesth 2004; 93(1):105-113.

32. Mombouli JV, Vanhoutte PM. Endothelial dysfunction: from physiology to therapy. J Mol Cell Cardiol 1999; 31(1):61-74.

33. Ignarro LJ, Buga GM, Wood KS, Byrns RE, Chaudhuri G. Endothelium-derived relaxing factor produced and released from artery and vein is nitric oxide. Proc Natl Acad Sci U S A 1987; 84(24):9265-9269.

34. Palmer RM, Ferrige AG, Moncada S. Nitric oxide release accounts for the biological activity of endothelium-derived relaxing factor. Nature 1987; 327(6122):524-526. 35. Moncada S, Vane JR. The role of prostacyclin in vascular tissue. Fed Proc 1979;

38(1):66-71.

36. Halushka PV, Mais DE, Morinelli TA. Thromboxane and prostacyclin receptors. Prog Clin Biol Res 1989; 301:21-28.

37. Feletou M, Vanhoutte PM. Endothelium-dependent hyperpolarization of canine coronary smooth muscle. Br J Pharmacol 1988; 93(3):515-524.

38. Feletou M, Vanhoutte PM. EDHF: an update. Clin Sci (Lond) 2009; 117(4):139-155. 39. Feletou M, Vanhoutte PM, Weston AH, Edwards G. EDHF and endothelial potassiun

channels: IKCa and SKCa. Br J Pharmacol 2003; 140(1):225.

40. Yanagisawa M, Kurihara H, Kimura S, Goto K, Masaki T. A novel peptide vasoconstrictor, endothelin, is produced by vascular endothelium and modulates smooth muscle Ca2+ channels. J Hypertens Suppl 1988; 6(4):S188-S191.

(21)

28

41. Masaki T. Possible role of endothelin in endothelial regulation of vascular tone. Annu Rev Pharmacol Toxicol 1995; 35:235-255.

42. Moncada S, Amezcua JL. Prostacyclin, thromboxane A2 interactions in haemostasis and thrombosis. Haemostasis 1979; 8(3-5):252-265.

43. Linder L, Kiowski W, Buhler FR, Luscher TF. Indirect evidence for release of endothelium-derived relaxing factor in human forearm circulation in vivo. Blunted response in essential hypertension. Circulation 1990; 81(6):1762-1767.

44. Panza JA, Quyyumi AA, Brush JE, Jr., Epstein SE. Abnormal endothelium-dependent vascular relaxation in patients with essential hypertension. N Engl J Med 1990; 323(1):22-27.

45. Taddei S, Virdis A, Mattei P, Salvetti A. Vasodilation to acetylcholine in primary and secondary forms of human hypertension. Hypertension 1993; 21(6 Pt 2):929-933. 46. Liszka HA, Mainous AG, III, King DE, Everett CJ, Egan BM. Prehypertension and

cardiovascular morbidity. Ann Fam Med 2005; 3(4):294-299.

47. Winegarden CR. From "prehypertension" to hypertension? Additional evidence. Ann Epidemiol 2005; 15(9):720-725.

48. Plavnik FL, Ajzen SA, Christofalo DM, Barbosa CS, Kohlmann O, Jr. Endothelial function in normotensive and high-normal hypertensive subjects. J Hum Hypertens 2007; 21(6):467-472.

49. Weil BR, Stauffer BL, Greiner JJ, Desouza CA. Prehypertension is associated with impaired nitric oxide-mediated endothelium-dependent vasodilation in sedentary adults. Am J Hypertens 2011; 24(9):976-981.

50. Virdis A, Ghiadoni L, Taddei S. Human endothelial dysfunction: EDCFs. Pflugers Arch 2010; 459(6):1015-1023.

51. Perticone F, Ceravolo R, Pujia A, Ventura G, Iacopino S, Scozzafava A, Ferraro A, Chello M, Mastroroberto P, Verdecchia P, Schillaci G. Prognostic significance of endothelial dysfunction in hypertensive patients. Circulation 2001; 104(2):191-196. 52. Taddei S, Virdis A, Mattei P, Arzilli F, Salvetti A. Endothelium-dependent forearm

vasodilation is reduced in normotensive subjects with familial history of hypertension. J Cardiovasc Pharmacol 1992; 20 Suppl 12:S193-S195.

53. Taddei S, Virdis A, Mattei P, Ghiadoni L, Sudano I, Salvetti A. Defective L-arginine-nitric oxide pathway in offspring of essential hypertensive patients. Circulation 1996; 94(6):1298-1303.

(22)

29

54. d'Uscio LV, Barton M, Shaw S, Moreau P, Luscher TF. Structure and function of small arteries in salt-induced hypertension: effects of chronic endothelin-subtype-A-receptor blockade. Hypertension 1997; 30(4):905-911.

55. Miller MJ, Pinto A, Mullane KM. Impaired endothelium-dependent relaxations in rabbits subjected to aortic coarctation hypertension. Hypertension 1987; 10(2):164-170.

56. Calver A, Collier J, Moncada S, Vallance P. Effect of local intra-arterial NG-monomethyl-L-arginine in patients with hypertension: the nitric oxide dilator mechanism appears abnormal. J Hypertens 1992; 10(9):1025-1031.

57. Panza JA, Casino PR, Kilcoyne CM, Quyyumi AA. Role of endothelium-derived nitric oxide in the abnormal endothelium-dependent vascular relaxation of patients with essential hypertension. Circulation 1993; 87(5):1468-1474.

58. Altiere RJ, Kiritsy-Roy JA, Catravas JD. Acetylcholine-induced contractions in isolated rabbit pulmonary arteries: role of thromboxane A2. J Pharmacol Exp Ther 1986; 236(2):535-541.

59. Miller VM, Vanhoutte PM. Endothelium-dependent contractions to arachidonic acid are mediated by products of cyclooxygenase. Am J Physiol 1985; 248(4 Pt 2):H432-H437.

60. Weil BR, Westby CM, Greiner JJ, Stauffer BL, Desouza CA. Elevated Endothelin-1 Vasoconstrictor Tone in Prehypertensive Adults. Can J Cardiol 2012.

61. Katusic ZS, Vanhoutte PM. Superoxide anion is an endothelium-derived contracting factor. Am J Physiol 1989; 257(1 Pt 2):H33-H37.

62. Ruschitzka F, Noll G, Luscher TF. Angiotensin converting enzyme inhibitors and vascular protection in hypertension. J Cardiovasc Pharmacol 1999; 34 Suppl 1:S3-12. 63. Vanhoutte PM. Endothelial dysfunction: the first step toward coronary arteriosclerosis.

Circ J 2009; 73(4):595-601.

64. Vane JR, Botting RM. Regulatory mechanisms of the vascular endothelium: an update. Pol J Pharmacol 1994; 46(6):499-21.

65. Taddei S, Salvetti A. Endothelial dysfunction in essential hypertension: clinical implications. J Hypertens 2002; 20(9):1671-1674.

66. Williams SB, Cusco JA, Roddy MA, Johnstone MT, Creager MA. Impaired nitric oxide-mediated vasodilation in patients with non-insulin-dependent diabetes mellitus. J Am Coll Cardiol 1996; 27(3):567-574.

(23)

30

67. Taddei S, Virdis A, Ghiadoni L, Mattei P, Sudano I, Bernini G, Pinto S, Salvetti A. Menopause is associated with endothelial dysfunction in women. Hypertension 1996; 28(4):576-582.

68. Heitzer T, Yla-Herttuala S, Luoma J, Kurz S, Munzel T, Just H, Olschewski M, Drexler H. Cigarette smoking potentiates endothelial dysfunction of forearm resistance vessels in patients with hypercholesterolemia. Role of oxidized LDL. Circulation 1996; 93(7):1346-1353.

69. Creager MA, Cooke JP, Mendelsohn ME, Gallagher SJ, Coleman SM, Loscalzo J, Dzau VJ. Impaired vasodilation of forearm resistance vessels in hypercholesterolemic humans. J Clin Invest 1990; 86(1):228-234.

70. Casino PR, Kilcoyne CM, Quyyumi AA, Hoeg JM, Panza JA. The role of nitric oxide in endothelium-dependent vasodilation of hypercholesterolemic patients. Circulation 1993; 88(6):2541-2547.

71. Taddei S, Virdis A, Mattei P, Ghiadoni L, Gennari A, Fasolo CB, Sudano I, Salvetti A. Aging and endothelial function in normotensive subjects and patients with essential hypertension. Circulation 1995; 91(7):1981-1987.

72. Laragh JH. Essential hypertension: new concepts in diagnosis and treatment. Compr Ther 1978; 4(12):10-17.

73. Kristiansen K. Molecular mechanisms of ligand binding, signaling, and regulation within the superfamily of G-protein-coupled receptors: molecular modeling and mutagenesis approaches to receptor structure and function. Pharmacol Ther 2004; 103(1):21-80.

74. Cabrera-Vera TM, Vanhauwe J, Thomas TO, Medkova M, Preininger A, Mazzoni MR, Hamm HE. Insights into G protein structure, function, and regulation. Endocr Rev 2003; 24(6):765-781.

75. Pierce KL, Premont RT, Lefkowitz RJ. Seven-transmembrane receptors. Nat Rev Mol Cell Biol 2002; 3(9):639-650.

76. Ferguson SS. Evolving concepts in G protein-coupled receptor endocytosis: the role in receptor desensitization and signaling. Pharmacol Rev 2001; 53(1):1-24.

77. Marchese A, Paing MM, Temple BR, Trejo J. G protein-coupled receptor sorting to endosomes and lysosomes. Annu Rev Pharmacol Toxicol 2008; 48:601-629.

78. Kenakin T. Functional selectivity through protean and biased agonism: who steers the ship? Mol Pharmacol 2007; 72(6):1393-1401.

79. Higgins B, Williams B. Pharmacological management of hypertension. Clin Med 2007; 7(6):612-616.

(24)

31

80. Mancia G, Grassi G. Management of essential hypertension. Br Med Bull 2010; 94:189-199.

81. Tuomilehto J, Ryynanen OP, Koistinen A, Rastenyte D, Nissinen A, Puska P. Low diastolic blood pressure and mortality in a population-based cohort of 16913 hypertensive patients in North Karelia, Finland. J Hypertens 1998; 16(9):1235-1242. 82. Bangalore S, Messerli FH, Wun CC, Zuckerman AL, DeMicco D, Kostis JB, LaRosa JC.

J-curve revisited: An analysis of blood pressure and cardiovascular events in the Treating to New Targets (TNT) Trial. Eur Heart J 2010; 31(23):2897-2908.

83. Boshuizen HC, Izaks GJ, van BS, Ligthart GJ. Blood pressure and mortality in elderly people aged 85 and older: community based study. BMJ 1998; 316(7147):1780-1784. 84. Kannel WB, Wilson PW, Nam BH, D'Agostino RB, Li J. A likely explanation for the

J-curve of blood pressure cardiovascular risk. Am J Cardiol 2004; 94(3):380-384. 85. Panza JA, Quyyumi AA, Callahan TS, Epstein SE. Effect of antihypertensive treatment

on endothelium-dependent vascular relaxation in patients with essential hypertension. J Am Coll Cardiol 1993; 21(5):1145-1151.

86. De GC, V, Rigamonti A, Fioretti S, Bonomo S, Manfredi B, Ferrario P, Bianchi M, Berti F, Muller EE, Rossoni G. Angiotensin-converting enzyme inhibition and angiotensin AT1-receptor antagonism equally improve endothelial vasodilator function in L-NAME-induced hypertensive rats. Eur J Pharmacol 2005; 516(3):253-259.

87. Dohi Y, Criscione L, Pfeiffer K, Luscher TF. Angiotensin blockade or calcium antagonists improve endothelial dysfunction in hypertension: studies in perfused mesenteric resistance arteries. J Cardiovasc Pharmacol 1994; 24(3):372-379.

88. Tschudi MR, Criscione L, Novosel D, Pfeiffer K, Luscher TF. Antihypertensive therapy augments endothelium-dependent relaxations in coronary arteries of spontaneously hypertensive rats. Circulation 1994; 89(5):2212-2218.

89. Yavuz D, Koc M, Toprak A, Akpinar I, Velioglu A, Deyneli O, Haklar G, Akalin S. Effects of ACE inhibition and AT1-receptor antagonism on endothelial function and insulin sensitivity in essential hypertensive patients. J Renin Angiotensin Aldosterone Syst 2003; 4(3):197-203.

90. Taddei S, Virdis A, Ghiadoni L, Mattei P, Salvetti A. Effects of angiotensin converting enzyme inhibition on endothelium-dependent vasodilatation in essential hypertensive patients. J Hypertens 1998; 16(4):447-456.

91. Ghiadoni L, Virdis A, Magagna A, Taddei S, Salvetti A. Effect of the angiotensin II type 1 receptor blocker candesartan on endothelial function in patients with essential hypertension. Hypertension 2000; 35(1 Pt 2):501-506.

(25)

32

92. The Lipid Bilayer. In: Alberts B, Johnson A, Lewis J, Raff M, Roberts K, Walter P, editors. Molecular Biology of the Cell. Garland Science, 2012.

93. Fahy E, Subramaniam S, Brown HA, Glass CK, Merrill AH, Jr., Murphy RC, Raetz CR, Russell DW, Seyama Y, Shaw W, Shimizu T, Spener F, van MG, VanNieuwenhze MS, White SH, Witztum JL, Dennis EA. A comprehensive classification system for lipids. J Lipid Res 2005; 46(5):839-861.

94. Fonteh AN, Harrington RJ, Huhmer AF, Biringer RG, Riggins JN, Harrington MG. Identification of disease markers in human cerebrospinal fluid using lipidomic and proteomic methods. Dis Markers 2006; 22(1-2):39-64.

95. van MG, de Kroon AI. Lipid map of the mammalian cell. J Cell Sci 2011; 124(Pt 1):5-8. 96. Hannun YA, Luberto C, Argraves KM. Enzymes of sphingolipid metabolism: from

modular to integrative signaling. Biochemistry 2001; 40(16):4893-4903.

97. Marchesini N, Hannun YA. Acid and neutral sphingomyelinases: roles and mechanisms of regulation. Biochem Cell Biol 2004; 82(1):27-44.

98. Takabe K, Paugh SW, Milstien S, Spiegel S. "Inside-out" signaling of sphingosine-1-phosphate: therapeutic targets. Pharmacol Rev 2008; 60(2):181-195.

99. Duan RD, Hertervig E, Nyberg L, Hauge T, Sternby B, Lillienau J, Farooqi A, Nilsson A. Distribution of alkaline sphingomyelinase activity in human beings and animals. Tissue and species differences. Dig Dis Sci 1996; 41(9):1801-1806.

100. Nyberg L, Duan RD, Axelson J, Nilsson A. Identification of an alkaline sphingomyelinase activity in human bile. Biochim Biophys Acta 1996; 1300(1):42-48. 101. Liu B, Obeid LM, Hannun YA. Sphingomyelinases in cell regulation. Semin Cell Dev

Biol 1997; 8(3):311-322.

102. Fatty acid structure and metabolism. In: Gurr MI, Harwood JL, Frayn KN, editors. Lipid Biochemistry - An Introduction. Blackwell, 2002: 13-91.

103. Wijesinghe DS, Massiello A, Subramanian P, Szulc Z, Bielawska A, Chalfant CE. Substrate specificity of human ceramide kinase. J Lipid Res 2005; 46(12):2706-2716. 104. Hannun YA, Obeid LM. The Ceramide-centric universe of lipid-mediated cell

regulation: stress encounters of the lipid kind. J Biol Chem 2002; 277(29):25847-25850.

105. Linn SC, Kim HS, Keane EM, Andras LM, Wang E, Merrill AH, Jr. Regulation of de novo sphingolipid biosynthesis and the toxic consequences of its disruption. Biochem Soc Trans 2001; 29(Pt 6):831-835.

(26)

33

106. Pewzner-Jung Y, Ben-Dor S, Futerman AH. When do Lasses (longevity assurance genes) become CerS (ceramide synthases)?: Insights into the regulation of ceramide synthesis. J Biol Chem 2006; 281(35):25001-25005.

107. Causeret C, Geeraert L, Van der Hoeven G, Mannaerts GP, Van Veldhoven PP. Further characterization of rat dihydroceramide desaturase: tissue distribution, subcellular localization, and substrate specificity. Lipids 2000; 35(10):1117-1125.

108. Hanada K, Kumagai K, Yasuda S, Miura Y, Kawano M, Fukasawa M, Nishijima M. Molecular machinery for non-vesicular trafficking of ceramide. Nature 2003; 426(6968):803-809.

109. Raas-Rothschild A, Pankova-Kholmyansky I, Kacher Y, Futerman AH. Glycosphingolipidoses: beyond the enzymatic defect. Glycoconj J 2004; 21(6):295-304.

110. D'Angelo G, Polishchuk E, Di TG, Santoro M, Di CA, Godi A, West G, Bielawski J, Chuang CC, van der Spoel AC, Platt FM, Hannun YA, Polishchuk R, Mattjus P, De Matteis MA. Glycosphingolipid synthesis requires FAPP2 transfer of glucosylceramide. Nature 2007; 449(7158):62-67.

111. Xu R, Jin J, Hu W, Sun W, Bielawski J, Szulc Z, Taha T, Obeid LM, Mao C. Golgi alkaline ceramidase regulates cell proliferation and survival by controlling levels of sphingosine and S1P. FASEB J 2006; 20(11):1813-1825.

112. Galadari S, Wu BX, Mao C, Roddy P, El BS, Hannun YA. Identification of a novel amidase motif in neutral ceramidase. Biochem J 2006; 393(Pt 3):687-695.

113. Hait NC, Oskeritzian CA, Paugh SW, Milstien S, Spiegel S. Sphingosine kinases, sphingosine 1-phosphate, apoptosis and diseases. Biochim Biophys Acta 2006; 1758(12):2016-2026.

114. Maceyka M, Sankala H, Hait NC, Le SH, Liu H, Toman R, Collier C, Zhang M, Satin LS, Merrill AH, Jr., Milstien S, Spiegel S. SphK1 and SphK2, sphingosine kinase isoenzymes with opposing functions in sphingolipid metabolism. J Biol Chem 2005; 280(44):37118-37129.

115. Pitson SM, Moretti PA, Zebol JR, Lynn HE, Xia P, Vadas MA, Wattenberg BW. Activation of sphingosine kinase 1 by ERK1/2-mediated phosphorylation. EMBO J 2003; 22(20):5491-5500.

116. Pitson SM, Xia P, Leclercq TM, Moretti PA, Zebol JR, Lynn HE, Wattenberg BW, Vadas MA. Phosphorylation-dependent translocation of sphingosine kinase to the plasma membrane drives its oncogenic signalling. J Exp Med 2005; 201(1):49-54.

(27)

34

117. Venkataraman K, Thangada S, Michaud J, Oo ML, Ai Y, Lee YM, Wu M, Parikh NS, Khan F, Proia RL, Hla T. Extracellular export of sphingosine kinase-1a contributes to the vascular S1P gradient. Biochem J 2006; 397(3):461-471.

118. Pyne NJ, Long JS, Lee SC, Loveridge C, Gillies L, Pyne S. New aspects of sphingosine 1-phosphate signaling in mammalian cells. Adv Enzyme Regul 2009; 49(1):214-221. 119. Spiegel S, Milstien S. Sphingosine-1-phosphate: an enigmatic signalling lipid. Nat Rev

Mol Cell Biol 2003; 4(5):397-407.

120. Bandhuvula P, Saba JD. Sphingosine-1-phosphate lyase in immunity and cancer: silencing the siren. Trends Mol Med 2007; 13(5):210-217.

121. Van Veldhoven PP, Mannaerts GP. Sphingosine-phosphate lyase. Adv Lipid Res 1993; 26:69-98.

122. Wymann MP, Schneiter R. Lipid signalling in disease. Nat Rev Mol Cell Biol 2008; 9(2):162-176.

123. Mitra P, Oskeritzian CA, Payne SG, Beaven MA, Milstien S, Spiegel S. Role of ABCC1 in export of sphingosine-1-phosphate from mast cells. Proc Natl Acad Sci U S A 2006; 103(44):16394-16399.

124. Meyer zu HD, Liliom K, Schaefer M, Danneberg K, Jaggar JH, Tigyi G, Jakobs KH. Photolysis of intracellular caged sphingosine-1-phosphate causes Ca2+ mobilization independently of G-protein-coupled receptors. FEBS Lett 2003; 554(3):443-449. 125. Strub GM, Maceyka M, Hait NC, Milstien S, Spiegel S. Extracellular and intracellular

actions of sphingosine-1-phosphate. Adv Exp Med Biol 2010; 688:141-155.

126. Kim KS, Ren J, Jiang Y, Ebrahem Q, Tipps R, Cristina K, Xiao YJ, Qiao J, Taylor KL, Lum H, Anand-Apte B, Xu Y. GPR4 plays a critical role in endothelial cell function and mediates the effects of sphingosylphosphorylcholine. FASEB J 2005; 19(7):819-821. 127. Alewijnse AE, Michel MC. Sphingosine-1-phosphate and sphingosylphosphorylcholine:

two of a kind? Br J Pharmacol 2006; 147(4):347-348.

128. Granado MH, Gangoiti P, Ouro A, Arana L, Gonzalez M, Trueba M, Gomez-Munoz A. Ceramide 1-phosphate (C1P) promotes cell migration Involvement of a specific C1P receptor. Cell Signal 2009; 21(3):405-412.

129. Meyer zu HD, Jakobs KH. Lysophospholipid receptors: signalling, pharmacology and regulation by lysophospholipid metabolism. Biochim Biophys Acta 2007; 1768(4):923-940.

(28)

35

130. Chun J, Goetzl EJ, Hla T, Igarashi Y, Lynch KR, Moolenaar W, Pyne S, Tigyi G. International Union of Pharmacology. XXXIV. Lysophospholipid receptor nomenclature. Pharmacol Rev 2002; 54(2):265-269.

131. Choi JW, Lee CW, Chun J. Biological roles of lysophospholipid receptors revealed by genetic null mice: an update. Biochim Biophys Acta 2008; 1781(9):531-539.

132. Hammad SM, Pierce JS, Soodavar F, Smith KJ, Al Gadban MM, Rembiesa B, Klein RL, Hannun YA, Bielawski J, Bielawska A. Blood sphingolipidomics in healthy humans: impact of sample collection methodology. J Lipid Res 2010; 51(10):3074-3087. 133. Yatomi Y, Yamamura S, Ruan F, Igarashi Y. Sphingosine 1-phosphate induces platelet

activation through an extracellular action and shares a platelet surface receptor with lysophosphatidic acid. J Biol Chem 1997; 272(8):5291-5297.

134. Pappu R, Schwab SR, Cornelissen I, Pereira JP, Regard JB, Xu Y, Camerer E, Zheng YW, Huang Y, Cyster JG, Coughlin SR. Promotion of lymphocyte egress into blood and lymph by distinct sources of sphingosine-1-phosphate. Science 2007; 316(5822):295-298.

135. Venkataraman K, Lee YM, Michaud J, Thangada S, Ai Y, Bonkovsky HL, Parikh NS, Habrukowich C, Hla T. Vascular endothelium as a contributor of plasma sphingosine 1-phosphate. Circ Res 2008; 102(6):669-676.

136. Park TS, Hu Y, Noh HL, Drosatos K, Okajima K, Buchanan J, Tuinei J, Homma S, Jiang XC, Abel ED, Goldberg IJ. Ceramide is a cardiotoxin in lipotoxic cardiomyopathy. J Lipid Res 2008 49(10):2101-2112

137. Furuya K, Ginis I, Takeda H, Chen Y, Hallenbeck JM. Cell permeable exogenous ceramide reduces infarct size in spontaneously hypertensive rats supporting in vitro studies that have implicated ceramide in induction of tolerance to ischemia. J Cereb Blood Flow Metab 2001; 21(3):226-232.

138. Qin J, Berdyshev E, Goya J, Natarajan V, Dawson G. Neurons and oligodendrocytes recycle sphingosine 1-phosphate to ceramide: significance for apoptosis and multiple sclerosis. J Biol Chem 2010; 285(19):14134-14143.

139. Means CK, Brown JH. Sphingosine-1-phosphate receptor signalling in the heart. Cardiovasc Res 2009; 82(2):193-200.

140. Hermans E. Biochemical and pharmacological control of the multiplicity of coupling at G-protein-coupled receptors. Pharmacol Ther 2003; 99(1):25-44. Review

(29)

36

141. Mazurais D, Robert P, Gout B, Berrebi-Bertrand I, Laville MP, Calmels T. Cell type-specific localization of human cardiac S1P receptors. J Histochem Cytochem 2002; 50(5):661-670.

142. Wendler CC, Rivkees SA. Sphingosine-1-phosphate inhibits cell migration and endothelial to mesenchymal cell transformation during cardiac development. Dev Biol 2006; 291(2):264-277.

143. Himmel HM, Meyer zu HD, Graf E, Dobrev D, Kortner A, Schuler S, Jakobs KH, Ravens U. Evidence for Edg-3 receptor-mediated activation of I(K.ACh) by sphingosine-1-phosphate in human atrial cardiomyocytes. Mol Pharmacol 2000; 58(2):449-454. 144. Egom EE, Mohamed TM, Mamas MA, Shi Y, Liu W, Chirico D, Stringer SE, Ke Y,

Shaheen M, Wang T, Chacko S, Wang X, Solaro RJ, Fath-Ordoubadi F, Cartwright EJ, Lei M. Activation of Pak1/Akt/eNOS signaling following sphingosine-1-phosphate release as part of a mechanism protecting cardiomyocytes against ischemic cell injury. Am J Physiol Heart Circ Physiol 2011; 301(4):1487-1495.

145. Knapp M, Baranowski M, Czarnowski D, Lisowska A, Zabielski P, Górski J, Musiał W. Plasma sphingosine-1-phosphate concentration is reduced in patients with myocardial infarction. Med Sci Monit 2009;15(9):490-493.

146. Knapp M, Baranowski M, Lisowska A, Musial W. Decreased free sphingoid base concentration in the plasma of patients with chronic systolic heart failure. Adv Med Sci 2012;1-6.

147. Sattler KJ, Elbasan S, Keul P, Elter-Schulz M, Bode C, Graler MH, Brocker-Preuss M, Budde T, Erbel R, Heusch G, Levkau B. Sphingosine 1-phosphate levels in plasma and HDL are altered in coronary artery disease. Basic Res Cardiol 2010; 105(6):821-832. 148. Choi JW, Lee CW, Chun J. Biological roles of lysophospholipid receptors revealed by

genetic null mice: an update. Biochim Biophys Acta 2008; 1781(9):531-539.

149. Baranowski M, Charmas M, Długołęcka B, Górski J. Exercise increases plasma levels of sphingoid base-1 phosphates in humans. Acta Physiol (Oxf) 2011; 203(3):373-380. 150. Yatomi Y, Ozaki Y, Ohmori T, Igarashi Y. Sphingosine 1-phosphate: synthesis and

release. Prostaglandins 2001; 64(1-4):107-122.

151. Nofer JR, van der Giet M, Tolle M, Wolinska I, von Wnuck LK, Baba HA, Tietge UJ, Godecke A, Ishii I, Kleuser B, Schafers M, Fobker M, Zidek W, Assmann G, Chun J, Levkau B. HDL induces NO-dependent vasorelaxation via the lysophospholipid receptor S1P3. J Clin Invest 2004; 113(4):569-581.

152. Argraves KM, Sethi AA, Gazzolo PJ, Wilkerson BA, Remaley AT, Tybjaerg-Hansen A, Nordestgaard BG, Yeatts SD, Nicholas KS, Barth JL, Argraves WS. S1P, dihydro-S1P

(30)

37

and C24:1-ceramide levels in the HDL-containing fraction of serum inversely correlate with occurrence of ischemic heart disease. Lipids Health Dis 2011; 10:70.

153. Morales-Ruiz M, Lee MJ, Zollner S, Gratton JP, Scotland R, Shiojima I, Walsh K, Hla T, Sessa WC. Sphingosine 1-phosphate activates Akt, nitric oxide production, and chemotaxis through a Gi protein/phosphoinositide 3-kinase pathway in endothelial cells. J Biol Chem 2001; 276(22):19672-19677.

154. Hanson MA, Roth CB, Jo E, Griffith MT, Scott FL, Reinhart G, Desale H, Clemons B, Cahalan SM, Schuerer SC, Sanna MG, Han GW, Kuhn P, Rosen H, Stevens RC. Crystal structure of a lipid G protein-coupled receptor. Science 2012; 335(6070):851-855. 155. Peters SL, Alewijnse AE. Sphingosine-1-phosphate signaling in the cardiovascular

system. Curr Opin Pharmacol 2007; 7(2):186-192.

156. Jung B, Obinata H, Galvani S, Mendelson K, Ding BS, Skoura A, Kinzel B, Brinkmann V, Rafii S, Evans T, Hla T. Flow-regulated endothelial S1P receptor-1 signaling sustains vascular development. Dev Cell 2012; 23(3):600-610.

157. Liu Y, Wada R, Yamashita T, Mi Y, Deng CX, Hobson JP, Rosenfeldt HM, Nava VE, Chae SS, Lee MJ, Liu CH, Hla T, Spiegel S, Proia RL. Edg-1, the G protein-coupled receptor for sphingosine-1-phosphate, is essential for vascular maturation. J Clin Invest 2000; 106(8):951-961.

158. Mizugishi K, Yamashita T, Olivera A, Miller GF, Spiegel S, Proia RL. Essential role for sphingosine kinases in neural and vascular development. Mol Cell Biol 2005; 25(24):11113-11121.

159. Kono M, Belyantseva IA, Skoura A, Frolenkov GI, Starost MF, Dreier JL, Lidington D, Bolz SS, Friedman TB, Hla T, Proia RL. Deafness and stria vascularis defects in S1P2 receptor-null mice. J Biol Chem 2007; 282(14):10690-10696.

160. Lorenz JN, Arend LJ, Robitz R, Paul RJ, MacLennan AJ. Vascular dysfunction in S1P2 sphingosine 1-phosphate receptor knockout mice. Am J Physiol Regul Integr Comp Physiol 2007; 292(1):R440-R446.

161. Kono M, Belyantseva IA, Skoura A, Frolenkov GI, Starost MF, Dreier JL, Lidington D, Bolz SS, Friedman TB, Hla T, Proia RL. Deafness and stria vascularis defects in S1P2 receptor-null mice. J Biol Chem 2007; 282(14):10690-10696.

162. Lorenz JN, Arend LJ, Robitz R, Paul RJ, MacLennan AJ. Vascular dysfunction in S1P2 sphingosine 1-phosphate receptor knockout mice. Am J Physiol Regul Integr Comp Physiol 2007;292(1):440-446.

163. Hanson MA, Roth CB, Jo E, Griffith MT, Scott FL, Reinhart G, Desale H, Clemons B, Cahalan SM, Schuerer SC, Sanna MG, Han GW, Kuhn P, Rosen H, Stevens RC. Crystal structure of a lipid G protein-coupled receptor. Science 2012; 335(6070):851-855.

(31)

38

164. Ishii I, Friedman B, Ye X, Kawamura S, McGiffert C, Contos JJ, Kingsbury MA, Zhang G, Brown JH, Chun J. Selective loss of sphingosine 1-phosphate signaling with no obvious phenotypic abnormality in mice lacking its G protein-coupled receptor, LP(B3)/EDG-3. J Biol Chem 2001; 276(36):33697-33704.

165. Panetti TS. Differential effects of sphingosine 1-phosphate and lysophosphatidic acid on endothelial cells. Biochim Biophys Acta 2002; 1582(1-3):190-196.

166. Waeber C, Blondeau N, Salomone S. Vascular sphingosine-1-phosphate S1P1 and S1P3 receptors. Drug News Perspect 2004; 17(6):365-382.

167. Alewijnse AE, Peters SL, Michel MC. Cardiovascular effects of sphingosine-1-phosphate and other sphingomyelin metabolites. Br J Pharmacol 2004; 143(6):666-684.

168. Kluk MJ, Hla T. Role of the sphingosine 1-phosphate receptor EDG-1 in vascular smooth muscle cell proliferation and migration. Circ Res 2001; 89(6):496-502. 169. Yogi A, Callera GE, Aranha AB, Antunes TT, Graham D, McBride M, Dominiczak A,

Touyz RM. Sphingosine-1-phosphate-induced inflammation involves receptor tyrosine kinase transactivation in vascular cells: upregulation in hypertension. Hypertension. 2011; 57(4):809-818.

170. Igarashi J, Miyoshi M, Hashimoto T, Kubota Y, Kosaka H. Statins induce S1P1 receptors and enhance endothelial nitric oxide production in response to high-density lipoproteins. Br J Pharmacol 2007; 150(4):470-479.

171. Igarashi J, Erwin PA, Dantas AP, Chen H, Michel T. VEGF induces S1P1 receptors in endothelial cells: Implications for cross-talk between sphingolipid and growth factor receptors. Proc Natl Acad Sci U S A 2003; 100(19):10664-10669.

172. Hla T, Maciag T. An abundant transcript induced in differentiating human endothelial cells encodes a polypeptide with structural similarities to G-protein-coupled receptors. J Biol Chem 1990; 265(16):9308-9313.

173. Cai H. Hydrogen peroxide regulation of endothelial function: origins, mechanisms, and consequences. Cardiovasc Res 2005; 68(1):26-36.

174. Levkau B. Sphingosine-1-phosphate in the regulation of vascular tone: a finely tuned integration system of S1P sources, receptors, and vascular responsiveness. Circ Res 2008; 103(3):231-233.

175. Hemmings DG, Xu Y, Davidge ST. Sphingosine 1-phosphate-induced vasoconstriction is elevated in mesenteric resistance arteries from aged female rats. Br J Pharmacol 2004; 143(2):276-284.

Referenties

GERELATEERDE DOCUMENTEN

Figure 2 Expression data matrix of 70 prognostic marker genes from 78 pnmary breast tumors plus seven pairs of matching pairs of primary tumors and distant metastases (6. 10) Each

To define patterns of genes or gene regulatory pathways that drive breast cancer lymph node metastasis, we compared the gene expression profiles of 15 primary breast carcinomas

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), other than for strictly

This thesis describes two ways of gaining insight into the biology of the metastatic process: first, the detection and prognostic role of disseminated breast cancer cells,

In Hoofdstuk 7 laten wij zien dat metastasen op afstand niet alleen hetzelfde genexpressie profiel hebben als de bijbehorende primaire tumor, maar dat ook het moleculaire

In Kapitel 3 zeigen wir, dass Patientinnen mit fortgeschrittenem Brust- krebs signifikant schlechtere Aussichten auf ein progressionsfreies Überleben haben und eine

Detection of circulating breast tumor cells by differential expression of marker genes.. Marker genes

If you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please let the Library know, stating your reasons.. In case of