19
UvA-DARE is a service provided by the library of the University of Amsterdam (http://dare.uva.nl) UvA-DARE (Digital Academic Repository) Cytokines in atherosclerosis: an intricate balance Boshuizen, M.C.S. Link to publication Citation for published version (APA): Boshuizen, M. C. S. (2016). Cytokines in atherosclerosis: an intricate balance. 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. Download date: 02 Mar 2020

UvA-DARE (Digital Academic Repository) Cytokines in atherosclerosis… · foam cells, including oxysterols like oxLDL 38, oxygen radicals like nitric oxide 36 and several cytokines,

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: UvA-DARE (Digital Academic Repository) Cytokines in atherosclerosis… · foam cells, including oxysterols like oxLDL 38, oxygen radicals like nitric oxide 36 and several cytokines,

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

UvA-DARE (Digital Academic Repository)

Cytokines in atherosclerosis: an intricate balance

Boshuizen, M.C.S.

Link to publication

Citation for published version (APA):Boshuizen, M. C. S. (2016). Cytokines in atherosclerosis: an intricate balance.

General rightsIt 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 regulationsIf you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please let the Library know, statingyour reasons. In case of a legitimate complaint, the Library will make the material inaccessible and/or remove it from the website. Please Askthe 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.

Download date: 02 Mar 2020

Page 2: UvA-DARE (Digital Academic Repository) Cytokines in atherosclerosis… · foam cells, including oxysterols like oxLDL 38, oxygen radicals like nitric oxide 36 and several cytokines,

Chapter 1

General Introducti on

13358- Boshuizen-Binnenwerk na proefdruk.indd 9 4-3-2016 16:17:13

Page 3: UvA-DARE (Digital Academic Repository) Cytokines in atherosclerosis… · foam cells, including oxysterols like oxLDL 38, oxygen radicals like nitric oxide 36 and several cytokines,

Chapter 1

10

13358- Boshuizen-Binnenwerk na proefdruk.indd 10 4-3-2016 16:17:13

Page 4: UvA-DARE (Digital Academic Repository) Cytokines in atherosclerosis… · foam cells, including oxysterols like oxLDL 38, oxygen radicals like nitric oxide 36 and several cytokines,

1

General Introduction

11

IntroductionThe improvement in quality of life has increased the average life-expectancy over the last few years in many parts of the world. This is, however, accompanied by a strong increase in diseases associated with ageing, such as neurodegenerative diseases, cancer and cardiovascular diseases (CVDs). CVD is the main global cause of mortality, with 17.3 million deaths per year, a number that is expected to grow rapidly 1. Atherosclerosis is the underlying pathology in the majority of clinical manifestations of CVD. Atherosclerotic disease progression is normally without any clinical symptoms. However, when clinical symptoms arise, they are mostly acute and can be the result of blood flow-obstructing stenoses or because of plaque rupture or erosion. Plaque rupture or erosion results in thrombus formation which can obstruct blood flow in the heart or in the brain, leading to myocardial infarction or stroke respectively 2.Several risk factors are known to promote atherogenesis and its clinical manifestations, of which smoking, dyslipidemia, hypertension and obesity strongly contribute to disease 3. With the incidence of obesity and the metabolic syndrome growing worldwide, a serious threat for the increased development of CVDs, including atherosclerosis, has emerged 4-6. Lipid-lowering strategies have thus far been the main therapeutic approach to reduce atherosclerosis 7, 8. But anti-inflammatory strategies now also seem promising in reducing atherogenesis in experimental settings 9. Currently, clinical trials are being performed in which the relevance of blocking inflammatory agents in atherosclerosis development are tested 10-12. However, these therapies will likely not be enough to fully eradicate atherosclerosis and its cardiovascular complications. As atherosclerosis is an extremely complex and multifactorial disease, better understanding of the disease process is needed to develop more successful therapies.

Atherogenesis Atherosclerosis is a complex lipid-driven inflammatory disease in which numerous cell types and inflammatory mediators are involved 13, 14. The initiating step in atherogenesis is endothelial dysfunction, which can be induced by shear stress or by traditional risk factors such as dyslipidemia or hypertension. A result of endothelial dysfunction is increased permeability to circulating lipoproteins, such as low-density lipoproteins (LDL) 15, 16. LDL particles accumulate in the intima, were they are prone to become modified, often resulting in oxidized LDL (oxLDL), which is the most abundant lipoprotein form present in atherosclerotic lesions 17. OxLDL on its turn induces inflammatory responses by further activating the endothelium by the expression of adhesion molecules such as vascular cell adhesion molecule-1 (VCAM-1) and intercellular adhesion molecule-1 (ICAM-1), and by the expression of several cytokines and chemokines such as monocyte chemoattractant protein-1 (MCP-1) 2, 18. Consequently, monocytes are recruited to the arterial wall, where they migrate into the intima, differentiate into macrophages and ingest the modified lipoproteins 19, 20. This uptake is mainly regulated via scavenger receptor A and CD36 and leads to lipid-loaded foam cells 21, 22. Macrophages and foam cells are critical components of atherosclerotic lesions as they enhance inflammation via secretion of inflammatory cytokines and chemokines which recruits more inflammatory cells like monocytes, T cells and neutrophils 13, 23. Macrophages are very plastic cells, as they can switch from one phenotype to another thereby adopting different polarization states 24. Numerous factors in the plaque microenvironment can induce macrophage polarization, resulting in a very heterogenic cell population 25. A

13358- Boshuizen-Binnenwerk na proefdruk.indd 11 4-3-2016 16:17:13

Page 5: UvA-DARE (Digital Academic Repository) Cytokines in atherosclerosis… · foam cells, including oxysterols like oxLDL 38, oxygen radicals like nitric oxide 36 and several cytokines,

Chapter 1

12

simplified classification describes that plaque-derived signals, like cytokines, can induce either a pro-inflammatory M1 macrophage phenotype or an anti-inflammatory M2 phenotype. M1 macrophages are elicited by stimulation with IFNγ or LPS and secrete several inflammatory cytokines such as tumor necrosis factor (TNF), interleukin (IL)-1β, IL-6 and IL-12, and produce reactive oxygen species. M2 macrophages can be induced upon stimulation with IL-4, IL-13 and IL-10 and respond by secretion of anti-inflammatory cytokines like IL-10 and TGF-β 19, 24. As can be deducted from these characteristics, M1 macrophages may promote atherosclerosis, while M2 macrophages may dampen atherosclerosis development. Different macrophage populations have been described in human atherosclerotic lesions 26. Interestingly, M1 macrophages were observed in the rupture-prone shoulder regions of plaques, while M2 macrophages were found in more stable plaque areas 27. Macrophage polarization may therefore be an important determinant of plaque stability.Besides macrophages, other immune cells also tightly regulate atherosclerosis development. Concerning innate immunity, neutrophils have received increasing attention during the last few years and they are now known to influence atherogenesis by the secretion of several pro-inflammatory mediators and by affecting plaque stability 28, 29. Dendritic cells (DCs) link innate and adaptive immunity and are found to accumulate in atherosclerotic lesions. However, different DC subsets have been identified which differ in their location and immune functions and thus also exert different actions throughout disease development 30. The adaptive immune system also strongly influences atherosclerosis development. On the one hand a pro-atherosclerotic role for T cells was initially described, when T cell deficient mice showed a strong decrease in lesion development 31. However, several T cell subsets are now identified, with varying pro- and anti-atherogenic properties. Some of those have already a well-described role in atherogenesis, while other subsets are just being discovered 32. On the other hand, B cells were initially described as being atheroprotective 33. However, upon the finding that the B cell population also consist of different subsets, different atherogenic effects were attributed to each subset 34, 35. With all those different immune cell subsets in mind, it is clear the immune response is central in controlling atherosclerosis development.When plaques advance, non-immune cells like vascular smooth muscle cells (VSMCs) come into play as well as they migrate from the media to the intima, where they form a plaque-stabilizing fibrous cap surrounding the pro-inflammatory core of the lesion. In addition, collagen is secreted to create further plaque stability 23. Plaque stability is challenged by the death of lesional macrophages as apoptotic macrophages expel their lipid content into the intima 36, 37. Several apoptosis inducers are known to play a role in apoptosis of lipid-loaded foam cells, including oxysterols like oxLDL 38, oxygen radicals like nitric oxide 36 and several cytokines, for instance the interferons 39-42. Those potential apoptosis inducers all have the property to activate caspases and to induce ER stress 43, 44. In early phases of atherosclerosis, apoptotic cells are efficiently phagocytosed by macrophages. However, when atherosclerotic lesions progress this clearance becomes inefficient as macrophages are already fully lipid-loaded. This results in secondary necrosis of the already death macrophages, leading to a plaque destabilizing necrotic core 38. The plaque fibrous cap and the necrotic core are the two most important determinants of plaque stability, which are both under the influence of inflammatory processes. So, the equilibrium between these lesional components and the inflammatory milieu determines plaque stability and thus, disease outcome.

Interferons In 1957 the interferons (IFNs) were the first cytokines to be described and were found to

13358- Boshuizen-Binnenwerk na proefdruk.indd 12 4-3-2016 16:17:13

Page 6: UvA-DARE (Digital Academic Repository) Cytokines in atherosclerosis… · foam cells, including oxysterols like oxLDL 38, oxygen radicals like nitric oxide 36 and several cytokines,

1

General Introduction

13

interfere with viral infection 45. Nowadays they are known as class 2 cytokines with various effects influencing both innate and adaptive immunity during numerous infections. They are considered to be important immunomodulatory cytokines, but in contrast to their protective capacities, interferons can also have detrimental effects on disease outcome 46-

48. Their role in disease is very context specific and depends among others on the cellular source of the interferons and the subsequent induction of specific interferon stimulated genes (ISGs), processes that thus should be tightly controlled. The interferons consist of 3 distinct family members. The type I interferon (T1 IFNs) family consist of several members of which IFNα and IFNβ are best characterized. T1 IFNs can be secreted by almost all cells, with the plasmacytoid dendritic cells (pDCs) being their main producers 46, 49. The only type II IFN (T2 IFN), IFNγ, is secreted by lymphocytes, activated macrophages and natural killer cells 50. The type III interferons were recently discovered and were found to have similar biological activity as the T1 IFNs. Most cells can secrete the type III IFNs but its receptor is mostly restricted to the epithelium 51. All three interferon families signal via a different heterodimeric receptor complex 52, 53, which is described in more detail in Chapter 2 54. IFN signaling results in induction of the expression of hundreds of ISGs, of which some are regulated by a single IFN family, while others are induced by all 3 classes of IFNs. Multiple connections exist between the different IFN families to induce a proper immune response 55, 56. For example, T1 IFNs can induce STAT1 homodimerization to stimulate IFNγ activated sequences, while T2 IFNs can activate the IFN-stimulated gene factor 3 complex to induce transcription of T1 IFN-target genes 57, 58.

Interferons in viral diseaseClassically, the T1 IFNs are known to be induced upon viral infections. The presence of viral components, mostly nucleic acids, triggers activation of pattern recognition receptors, such as toll-like receptors (TLRs), RIG-1-like receptors (RLRs) and NOD-like receptors (NLRs). These receptors are located either at the cell surface, in the cytosol or in endosomal compartments 59-62. Activation of those receptors results in rapid induction of T1 IFNs, which mostly requires the action of the transcription factors interferon regulatory factors (IRF) 3 and 7 63-66. The T1 IFNs are produced in the early phases of an infection, where pDCs are essential for their first production, while in later stages of infection other cell types produce T1 IFNs as well 49,

67. It has been shown that upon viral infection, 60% of the transcriptional activity of pDCs is associated with induction of T1 IFN genes 68. This rapid production of T1 IFNs is needed to hamper viral replication and spread either via direct production of ISGs such as myxovirus resistance 1 (MX1) and IFN-induced transmembrane proteins (IFITMs) 69, or via the indirect activation of antiviral functions of other cells such as macrophages, NK cells and T cells 70. As a result antigen presentation and cytokine production in innate cells is stimulated 71, while effector T cell responses are also enhanced 70. With those actions in mind, it is not surprising that several viruses have developed mechanisms to block T1 IFN antiviral activities 72, 73. Although the T1 IFNs have crucial antiviral actions, T1 IFNs can also have detrimental effects upon viral infection, which is among others mediated by the induction of immunosuppression 48, 74. In lymphocytic choriomeningitis virus (LCMV) and human immunodeficient virus (HIV) infections, the T1 IFNs exert protective effects during the early phases of infection. However, when these infections cannot be cleared and become chronic, persistent T1 IFN activity may become harmful. In chronic LCMV infections, T1 IFN activity induces the immunosuppressive cytokine IL-10 and the programmed cell death 1 ligand 1 (PDL1), which together suppress T cell function and therefore fail to clear infection 75, 76. HIV patients are

13358- Boshuizen-Binnenwerk na proefdruk.indd 13 4-3-2016 16:17:13

Page 7: UvA-DARE (Digital Academic Repository) Cytokines in atherosclerosis… · foam cells, including oxysterols like oxLDL 38, oxygen radicals like nitric oxide 36 and several cytokines,

Chapter 1

14

known to have a T1 IFN signaling profile 77, which has recently been associated with an increase in lymphocyte suppression and apoptosis, thereby enhancing disease progression 78, 79. The immunosuppressive actions of the T1 IFNs might be part of a coping mechanism to reduce host morbidity.Even though IFNγ is less well known as an antiviral cytokine, IFNγ has been proven to be essential for antiviral immunity as well 80-82. During a viral infection, IFNγ can be induced by earlier produced cytokines such as the T1 IFNs, IL-12 and IL-18 83, 84. This process is tightly controlled as the T1 IFNs can also negatively regulate IFNγ production, which seems mediated by STAT1 levels 85. Upon production of IFNγ, several antiviral mediators are produced of which protein kinase dsRNA-regulated (PKR) is an essential one. PKR is known to block viral protein synthesis and to induce apoptosis to limit viral spread 86, 87. Altogether, an equilibrium must be present in which the appropriate production of interferons is required to neutralize viral infections. A concise overview of the actions of IFNs in viral disease is presented in figure 1a.

Interferons in bacterial diseaseAlthough the T1 IFNs were initially known to have potent antiviral activities, it is now appreciated that they have important antibacterial effects as well. Since T1 IFNs were quite long considered to be unimportant in bacterial infections, they received little attention here and a better understanding is still needed to determine how exactly the T1 IFNs act as antibacterial mediators. An already well-described phenomenon is the induction of T1 IFNs by macrophages and DCs upon TLR4 activation by lipopolysaccharide (LPS) 88-

90. The secreted T1 IFNs then signal in an autocrine fashion to induce the production of ISGs, including the gene encoding inducible Nitric Oxide Synthase (iNOS). Nitric oxide (NO) production is essential in antibacterial immunity as many bacteria are sensitive to NO-mediated killing and its induction by T1 IFNs has been known for many years now 91, 92. As in viral disease, the T1 IFNs might have either disease inhibiting or disease promoting effects in bacterial infections, but the exact regulation of disease outcome is still poorly understood 48, 93. Next to NO production, the production of IFNγ by Th1 cells and NK cells is suggested to be an important mediator in T1 IFNs-induced antibacterial activity 94. IFNγ has always been classified as an antibacterial mediator. Upon infection IFNγ is secreted by Th1 cells and NK cells, which among others skews macrophages to a pro-inflammatory M1 phenotype. Activated M1 macrophages secrete IL-12 and IL-18 to stimulate microbicidal effector functions, including enhanced phagocytosis and the production of reactive oxygen species such as NO 50. The relevance of NO production in macrophage defense mechanisms is widely recognized 95. However, for maximal microbicidal NO production, IFNγ priming of macrophages is needed to enhance responses to bacterial LPS 96-98. The need for this priming mechanism is demonstrated by the resistance of IFNγ receptor (IFNGR) deficient mice to LPS-induced shock 99. In addition, bacterial infection models have shown the importance of IFNγ, as deficiency of IFNGR or T-bet, the regulator of Th1 differentiation, resulted in increased susceptibility and an increased severity of infection 47, 100. This susceptibility to bacterial disease was confirmed in IFNγ and IFNGR-deficient patients as well 101, 102. Again, a balance between the T1 IFNs and IFNγ may be of importance, which can influence the outcome of for instance mycobacterial infections 103-105. A concise overview of IFNs actions in bacterial disease is presented in figure 1a.

Interferons in autoimmune diseaseAs mentioned above, the interferons are crucial mediators of cellular defense mechanisms.

13358- Boshuizen-Binnenwerk na proefdruk.indd 14 4-3-2016 16:17:13

Page 8: UvA-DARE (Digital Academic Repository) Cytokines in atherosclerosis… · foam cells, including oxysterols like oxLDL 38, oxygen radicals like nitric oxide 36 and several cytokines,

1

General Introduction

15

However, their actions can also result in autoimmunity. The T1 IFNs for instance have been implicated in the pathogenesis of systemic lupus erythematosus (SLE). Peripheral blood monocytes from patients with SLE show an increased T1 IFNs signature 106 and the T1 IFNs are known to stimulate disease induction and progression 107. In addition, SLE patients may have an up to 50-fold increased risk for CVDs, which is amongst others attributed to this increased T1 IFN signature 108-111. Currently, clinical trials are being performed in which IFNα- and IFNAR-blocking strategies are being evaluated 112. The agents tested so far have proven to be safe, however great caution is required regarding the risk for developing infections. With the crosstalk of the T1 and T2 IFNs in mind, IFNγ also plays an established role in SLE pathogenesis 113. IFNγ-blocking therapies, like for IFNα, have thus far received little attention in the fight against SLE, probably due to the increased susceptibility towards infections. Nevertheless, IFNγ-antibody treatment of patients with progressive multiple sclerosis (MS) did show delay in disease progression. This indicates that neutralizing IFNγ could be a potential new treatment in progressive MS, outweighing the possible increased risk for infections 114. In Crohn’s disease, where IFNγ plays a prominent role, IFNγ-targeting therapies have also proven to be successful as early signs of clinical efficacy have been observed 115, 116. In addition to the abovementioned autoimmune diseases, future research should point out whether other diseases, like rheumatoid arthritis, would benefit from interferon-targeted therapies as well. A concise overview of IFNs actions in autoimmune disease is shown in figure 1b.

Interferons as therapeutic agents In contrast to the detrimental role for the T1 IFNs in autoimmunity, IFNβ is used as an effective treatment against relapsing remitting MS. This treatment has been shown to reduce MS relapses and delay the onset of progressive MS 117, 118. In hepatitis C virus (HCV) IFNα therapy has been the standard treatment for many years now, even though its effectiveness is rather limited. A shift in treatment regimen from interferon-based treatments towards more direct anti-viral therapies is now suggested as this seems to be more effective 119-

121. Also in myeloproliferative diseases and other types of cancer the T1 IFNs have been used as successful treatments 122, 123. The induction of an immune response to tumors by IFNγ and IFNγ’s anti-proliferative effects have been the subject in several clinical cancer trials as well 124, 125. Despite these beneficial indications, several attempts to treat cancer with recombinant IFNγ have so far resulted in mixed effects. This suggests that IFNγ can have both cytotoxic as well as cytoproliferative actions, likely depending on its surrounding environment 126. Because of IFNγ’s numerous inflammatory actions, this cytokine has clinically been tested as a treatment for several other diseases as well, including tuberculosis, invasive fungal infections and in HCV 127, 128. Despite all the valuable therapeutic applications of the interferons, it is important to keep their many immunomodulatory actions in mind, as those might influence the patients’ immune response.

13358- Boshuizen-Binnenwerk na proefdruk.indd 15 4-3-2016 16:17:14

Page 9: UvA-DARE (Digital Academic Repository) Cytokines in atherosclerosis… · foam cells, including oxysterols like oxLDL 38, oxygen radicals like nitric oxide 36 and several cytokines,

Chapter 1

16

virus/bacterium

a). IFNs in host protec�on

T1 IFNsISGs

UPRRs

U

infec�ouspar�cles

UIFNAR

T1 IFNsISGs

UIFNAR

DC

macrophage

IFNs

ISGs

ISGs B cell

IFNs

an�bodyproduc�on

YYY

IFNs

Ag presentation

IFNs

Ag presentation

NK cell

T cell

M1 macrophage skewing:- NO production- pro-in�ammatory cytokines e.g. TNF, IL-6, IL-12- phagocytosis

b). IFNs in autoimmunity

B cell

YYY

T cell

autoan�bodyproduc�on

YY

Y

Immune complexes

pDC

IL-10

immunosuppression

innate immune cell

cell death and release of self an�gens

Tissue injury

macrophage

T1 IFNs

IFNγ

IFNγ

infected cell

Figure 1. Overview of interferons in host protection and autoimmunity. a). Upon infection, infected cells produce IFNs, resulting in the expression of interferon stimulated genes (ISGs) in both infected and neighboring cells. One of the functions of ISGs is to limit the spread of infectious particles. Upon IFN stimulation or after sensing of infectious particles with pattern-recognition receptors (PRRs), macrophages and dendritic cells (DCs) also start producing IFNs and ISGs. In addition, they respond by increased antigen presentation and by the production of inflammatory mediators. Adaptive immunity is also influenced by the IFNs as effector functions of B and T cells

13358- Boshuizen-Binnenwerk na proefdruk.indd 16 4-3-2016 16:17:14

Page 10: UvA-DARE (Digital Academic Repository) Cytokines in atherosclerosis… · foam cells, including oxysterols like oxLDL 38, oxygen radicals like nitric oxide 36 and several cytokines,

1

General Introduction

17

are enhanced. b). The chronic IFN production that is present in several autoimmune diseases is continued in part by persistent stimulation of antigen-presenting cells, including macrophages and plasmacytoid dendritic cells (pDCs), by self antigens and damage-associated molecular pattern molecules (DAMPs). Continued IFN production increases T and B cell effector function, leading to autoantibody production, resulting in the characteristic tissue injury. Chronic stimulation by IFNs also increases production of IL-10 by innate immune cells such as macrophages, which suppresses inflammatory responses, likely to reduce host morbidity. IFNAR, interferon α/β receptor; Ag presentation, antigen presentation; NO, nitric oxide.

Interferons in atherosclerosisThe interferons are also known to play a substantial role in atherosclerosis. A growing body of evidence is present showing that the interferons act as pro-atherosclerotic mediators 41,

54, 129-139. Studies on the role of the T1 IFNs in atherosclerosis development have only just started, while the role of IFNγ in atherosclerosis is already longer established. Their actions are prominent throughout both the initiation and progression of the disease as they are for instance known to promote leukocyte recruitment towards atherosclerotic lesions and to stimulate lesional apoptosis 54. The way in which the interferons influence atherosclerosis development is extensively described in Chapter 2. In any case, the actions of this family of cytokines in atherogenesis are extremely complex, and the way in which they mediate atherosclerosis development can therefore be considered to be an intricate balance.

Interleukin-10Interleukin-10 (IL-10) is, like the interferons, a class 2 cytokine. It was discovered in 1989 as a cytokine that inhibits the production of pro-inflammatory cytokines by Th1 cells 140. Indeed, mice deficient in IL-10 have increased Th1 cell responses, resulting for instance in enhanced bacterial clearance upon infection 141, 142. Subsequently, it was found that IL-10 affects not only Th1 cells but several immune cells, like macrophages and DCs, thereby dampening inflammatory responses 143, 144. IL-10 is produced by nearly all leukocytes, adding to the complexity of this immunomodulatory cytokine 52, 144. T regulatory cells (Tregs) use IL-10 as their hallmark cytokine to limit inflammation and autoimmunity and nowadays B regulatory cells (Bregs) are investigated for their role in self-tolerance via the usage of IL-10 145, 146. Another important function of IL-10 is the suppression of antigen presentation by macrophages and DCs via an autocrine feedback loop 140, 147. As an essential immunosuppressive cytokine, IL-10 thus limits inflammatory responses at different levels in both innate and adaptive immunity.IL-10’s actions are mediated after binding to its cell surface receptor, IL-10R, which consist of two chains, IL-10R1 and IL-10R2. In response to IL-10 binding, signal transducer and activator of transcription (STAT)3, STAT1 and STAT5 are recruited to the receptor complex where they become phosphorylated for their activation. Homo- and heterodimers of these STAT molecules translocate to the nucleus to drive gene transcription of for instance suppressors of cytokine signaling (SOCS)-1 and SOCS-3 143. SOCS-1 is a negative regulator of inflammatory cytokine production and thus an important IL-10 target 148. Another mechanism by which IL-10 suppresses inflammation is by inhibition of NFκb activity, a transcription factor necessary for inflammatory cytokine production 149.IL-10-directed therapies have been tested in several clinical trials to treat autoimmune diseases. An important role for IL-10 in intestinal homeostasis has been described, which is impaired in inflammatory bowel disease (IBD) 150, 151. Despite protective effects of IL-10 in mouse intestinal inflammation models, clinical trials using IL-10 therapy in IBD have mainly been negative 144, 152. IL-10 administration in MS, where reduced secretion of IL-10 by Bregs

13358- Boshuizen-Binnenwerk na proefdruk.indd 17 4-3-2016 16:17:14

Page 11: UvA-DARE (Digital Academic Repository) Cytokines in atherosclerosis… · foam cells, including oxysterols like oxLDL 38, oxygen radicals like nitric oxide 36 and several cytokines,

Chapter 1

18

has been reported, also appeared ineffective 144, 153. In contrast to deficient production of IL-10 in several autoimmune diseases, overproduction of IL-10 by monocytes and B cells has been implicated in SLE disease pathogenesis 154. Here, neutralization of IL-10 has shown potential in a small cohort of SLE patients 155. But altogether, therapeutic targeting of IL-10 in autoimmune diseases still seems elusive as its widespread immunomodulatory actions are highly complex.

IL-10 in atherosclerosisWith the strong anti-inflammatory properties of IL-10 in mind, IL-10 is thought to act in an anti-atherogenic manner. IL-10 has been identified in the human atherosclerotic lesion, where it seems to modulate the inflammatory response and protect from too much apoptotic cell death 156. Since this discovery, IL-10 has been a target for intervention in experimental atherosclerosis. These experimental data further confirmed the anti-atherogenic role for IL-10, as deficiency of IL-10 157, 158 or of its downstream signaling molecules 159, 160 promoted atherosclerosis, whereas gene therapy for IL-10 161-164 decreased atherosclerosis development. These observed atheroprotective effects of IL-10 are not only attributed to anti-inflammatory activities, but also to its regulation of macrophage lipid handling. IL-10-mediated effects on cholesterol metabolism are primarily linked to increased cholesterol efflux, resulting in decreased macrophage foam cell formation 165, 166. In addition, IL-10 is also involved in stabilization of the atherosclerotic lesion as it provides pro-survival factors to leukocytes 156, 163, 167 and limits extracellular matrix degradation 157, 158, 168. Altogether, IL-10 can thus indeed be considered as an anti-atherogenic mediator. But as its atheroprotective actions are tightly controlled and still not fully understood, future research is needed to investigate the potential of IL-10-based therapies in atherosclerosis.

Aim of thesisIn recent years progress has been made in understanding the contribution of the interferons and IL-10 in both immunity and atherosclerosis, as can be deducted from the above mentioned sections. Accumulating in vitro and in vivo research indicates that the interferons can be considered as pro-atherosclerotic cytokines, whereas IL-10 is considered to be anti-atherogenic. Nevertheless, their atherosclerotic actions are very complicated and tightly balanced. More research is therefore needed to fully elucidate their atherosclerotic effects. To this end, the aim of this thesis is 1) to assess the functional contribution of interferons in atherogenesis with regards to foam cell formation and foam cell inflammatory responses and 2) to investigate whether interferon and IL-10 modulation can be used to diminish atherosclerosis.

Chapter 2 provides an overview regarding the role of interferons in atherosclerosis and discusses whether interferons could be regarded as new molecular targets in atherosclerosis treatment. In Chapter 3 the effects of IFNβ on macrophage foam cell formation are characterized, as to date a direct link between the T1 IFNs and this essential atherosclerotic process is still missing. We could demonstrate that IFNβ promotes macrophage foam cell formation by increasing SR-A-mediated cholesterol influx and decreasing ABCA1-mediated efflux. In Chapter 4 the role of IFNβ in foam cell inflammatory responses is assessed, as lipid loading of monocytes and macrophages resulted in downregulation of interferon signaling pathways. This made us question whether this also results in a decreased interferon response and a decrease in the accompanying inflammatory response. We observed macrophage

13358- Boshuizen-Binnenwerk na proefdruk.indd 18 4-3-2016 16:17:14

Page 12: UvA-DARE (Digital Academic Repository) Cytokines in atherosclerosis… · foam cells, including oxysterols like oxLDL 38, oxygen radicals like nitric oxide 36 and several cytokines,

1

General Introduction

19

hyporesponsiveness towards IFNβ under high cholesterol conditions, which might be relevant for IFNβ-mediated inflammatory activities. In Chapter 5 blockade of the IFNα/β receptor subunit 1 was investigated in a combined mouse model for atherosclerosis and arteriogenesis. We were able to stimulate arteriogenesis without enhancing atherosclerosis burden in our mice, which had been a downside of previous pro-arteriogenic stimuli so far. The pro-atherosclerotic role of IFNγ was already established many years ago. In vivo atherosclerosis studies to date focused on the systemic role of IFNγ, making it impossible to conclude which cell types exactly are involved here. Therefore we examined the effects of IFNγ on myeloid cells in atherogenesis, which is described in Chapter 6. From this study we could conclude that myeloid IFNγ signaling is dispensable for atherosclerosis development. A similar rationale formed the basis for the study described in Chapter 7. In this chapter the role of myeloid specific IL-10 signaling in atherosclerosis was studied. So far, the anti-atherogenic role of IL-10 was already well established, but the contribution of cell-specific effects still remained unclear. We could show that myeloid cells do not contribute to the atheroprotective actions of IL-10 and that myeloid IL-10 signaling even acts pro-atherogenic. In addition, we observed a new link between myeloid IL-10 signaling and cholesterol metabolism. Finally, in Chapter 8 all results obtained in this thesis and its implications are summarized and discussed.

References1. Mozaffarian D, Benjamin EJ, Go AS, Arnett DK, Blaha MJ, Cushman M, de Ferranti S, Despres JP, Fullerton HJ, Howard VJ, Huffman MD, Judd SE, Kissela BM, Lackland DT, Lichtman JH, Lisabeth LD, Liu S, Mackey RH, Matchar DB, McGuire DK, Mohler ER, 3rd, Moy CS, Muntner P, Mussolino ME, Nasir K, Neumar RW, Nichol G, Palaniappan L, Pandey DK, Reeves MJ, Rodriguez CJ, Sorlie PD, Stein J, Towfighi A, Turan TN, Virani SS, Willey JZ, Woo D, Yeh RW, Turner MB, American Heart Association Statistics C and Stroke Statistics S. Heart disease and stroke statistics--2015 update: a report from the American Heart Association. Circulation. 2015;131:e29-322.2. Hansson GK. Inflammation, atherosclerosis, and coronary artery disease. The New England journal of medicine. 2005;352:1685-95.3. Ezzati M, Obermeyer Z, Tzoulaki I, Mayosi BM, Elliott P and Leon DA. Contributions of risk factors and medical care to cardiovascular mortality trends. Nature reviews Cardiology. 2015.4. Alexopoulos N, Katritsis D and Raggi P. Visceral adipose tissue as a source of inflammation and promoter of atherosclerosis. Atherosclerosis. 2014;233:104-12.5. Luna-Luna M, Medina-Urrutia A, Vargas-Alarcon G, Coss-Rovirosa F, Vargas-Barron J and Perez-Mendez O. Adipose Tissue in Metabolic Syndrome: Onset and Progression of Atherosclerosis. Archives of medical research. 2015;46:392-407.6. Rocha VZ and Libby P. Obesity, inflammation, and atherosclerosis. Nature reviews Cardiology. 2009;6:399-409.7. Ferrieres J. Effects on coronary atherosclerosis by targeting low-density lipoprotein cholesterol with statins. American journal of cardiovascular drugs : drugs, devices, and other interventions. 2009;9:109-15.8. Nicholls SJ, Borgman M, Nissen SE, Raichlen JS, Ballantyne C, Barter P, Chapman MJ, Erbel R and Libby P. Impact of statins on progression of atherosclerosis: rationale and design of SATURN (Study of Coronary Atheroma by InTravascular Ultrasound: effect of Rosuvastatin versus Atorvastatin). Current medical research and opinion. 2011;27:1119-29.9. Moubayed SP, Heinonen TM and Tardif JC. Anti-inflammatory drugs and atherosclerosis. Current opinion in lipidology. 2007;18:638-44.10. Back M and Hansson GK. Anti-inflammatory therapies for atherosclerosis. Nature reviews Cardiology. 2015;12:199-211.11. Everett BM, Pradhan AD, Solomon DH, Paynter N, Macfadyen J, Zaharris E, Gupta M, Clearfield M, Libby P, Hasan AA, Glynn RJ and Ridker PM. Rationale and design of the Cardiovascular Inflammation Reduction Trial: a test of the inflammatory hypothesis of atherothrombosis. American heart journal. 2013;166:199-207 e15.12. Ridker PM, Thuren T, Zalewski A and Libby P. Interleukin-1beta inhibition and the prevention of recurrent cardiovascular events: rationale and design of the Canakinumab Anti-inflammatory Thrombosis Outcomes Study (CANTOS). American heart journal. 2011;162:597-605.13. Hansson GK and Libby P. The immune response in atherosclerosis: a double-edged sword. Nature reviews

13358- Boshuizen-Binnenwerk na proefdruk.indd 19 4-3-2016 16:17:14

Page 13: UvA-DARE (Digital Academic Repository) Cytokines in atherosclerosis… · foam cells, including oxysterols like oxLDL 38, oxygen radicals like nitric oxide 36 and several cytokines,

Chapter 1

20

Immunology. 2006;6:508-19.14. Libby P, Ridker PM and Hansson GK. Progress and challenges in translating the biology of atherosclerosis. Nature. 2011;473:317-25.15. Kinlay S, Libby P and Ganz P. Endothelial function and coronary artery disease. Current opinion in lipidology. 2001;12:383-9.16. Tabas I, Williams KJ and Boren J. Subendothelial lipoprotein retention as the initiating process in atherosclerosis: update and therapeutic implications. Circulation. 2007;116:1832-44.17. Steinberg D. Low density lipoprotein oxidation and its pathobiological significance. The Journal of biological chemistry. 1997;272:20963-6.18. Leitinger N. Oxidized phospholipids as modulators of inflammation in atherosclerosis. Current opinion in lipidology. 2003;14:421-30.19. Moore KJ, Sheedy FJ and Fisher EA. Macrophages in atherosclerosis: a dynamic balance. Nature reviews Immunology. 2013;13:709-21.20. Moore KJ and Tabas I. Macrophages in the pathogenesis of atherosclerosis. Cell. 2011;145:341-55.21. Kunjathoor VV, Febbraio M, Podrez EA, Moore KJ, Andersson L, Koehn S, Rhee JS, Silverstein R, Hoff HF and Freeman MW. Scavenger receptors class A-I/II and CD36 are the principal receptors responsible for the uptake of modified low density lipoprotein leading to lipid loading in macrophages. The Journal of biological chemistry. 2002;277:49982-8.22. Kzhyshkowska J, Neyen C and Gordon S. Role of macrophage scavenger receptors in atherosclerosis. Immunobiology. 2012;217:492-502.23. Lusis AJ. Atherosclerosis. Nature. 2000;407:233-41.24. Stoger JL, Goossens P and de Winther MP. Macrophage heterogeneity: relevance and functional implications in atherosclerosis. Current vascular pharmacology. 2010;8:233-48.25. Wolfs IM, Donners MM and de Winther MP. Differentiation factors and cytokines in the atherosclerotic plaque micro-environment as a trigger for macrophage polarisation. Thrombosis and haemostasis. 2011;106:763-71.26. Waldo SW, Li Y, Buono C, Zhao B, Billings EM, Chang J and Kruth HS. Heterogeneity of human macrophages in culture and in atherosclerotic plaques. The American journal of pathology. 2008;172:1112-26.27. Stoger JL, Gijbels MJ, van der Velden S, Manca M, van der Loos CM, Biessen EA, Daemen MJ, Lutgens E and de Winther MP. Distribution of macrophage polarization markers in human atherosclerosis. Atherosclerosis. 2012;225:461-8.28. Doring Y, Drechsler M, Soehnlein O and Weber C. Neutrophils in atherosclerosis: from mice to man. Arterioscler Thromb Vasc Biol 2015;35:288-95.29. Hartwig H, Silvestre Roig C, Daemen M, Lutgens E and Soehnlein O. Neutrophils in atherosclerosis. A brief overview. Hamostaseologie. 2015;35:121-7.30. Zernecke A. Dendritic cells in atherosclerosis: evidence in mice and humans. Arterioscler Thromb Vasc Biol 2015;35:763-70.31. Emeson EE, Shen ML, Bell CG and Qureshi A. Inhibition of atherosclerosis in CD4 T-cell-ablated and nude (nu/nu) C57BL/6 hyperlipidemic mice. The American journal of pathology. 1996;149:675-85.32. Andersson J, Libby P and Hansson GK. Adaptive immunity and atherosclerosis. Clinical immunology. 2010;134:33-46.33. Major AS, Fazio S and Linton MF. B-lymphocyte deficiency increases atherosclerosis in LDL receptor-null mice. Arteriosclerosis, thrombosis, and vascular biology. 2002;22:1892-8.34. Kyaw T, Tay C, Khan A, Dumouchel V, Cao A, To K, Kehry M, Dunn R, Agrotis A, Tipping P, Bobik A and Toh BH. Conventional B2 B cell depletion ameliorates whereas its adoptive transfer aggravates atherosclerosis. Journal of immunology. 2010;185:4410-9.35. Miller AM, Xu D, Asquith DL, Denby L, Li Y, Sattar N, Baker AH, McInnes IB and Liew FY. IL-33 reduces the development of atherosclerosis. The Journal of experimental medicine. 2008;205:339-46.36. Tabas I. Consequences and therapeutic implications of macrophage apoptosis in atherosclerosis: the importance of lesion stage and phagocytic efficiency. Arterioscler Thromb Vasc Biol 2005;25:2255-64.37. Tabas I. Macrophage death and defective inflammation resolution in atherosclerosis. Nature reviews Immunology. 2010;10:36-46.38. Tabas I. Apoptosis and plaque destabilization in atherosclerosis: the role of macrophage apoptosis induced by cholesterol. Cell death and differentiation. 2004;11 Suppl 1:S12-6.39. Bai Y, Ahmad U, Wang Y, Li JH, Choy JC, Kim RW, Kirkiles-Smith N, Maher SE, Karras JG, Bennett CF, Bothwell AL, Pober JS and Tellides G. Interferon-gamma induces X-linked inhibitor of apoptosis-associated factor-1 and Noxa expression and potentiates human vascular smooth muscle cell apoptosis by STAT3 activation. The Journal of biological chemistry. 2008;283:6832-42.40. Bernabei P, Coccia EM, Rigamonti L, Bosticardo M, Forni G, Pestka S, Krause CD, Battistini A and Novelli F. Interferon-gamma receptor 2 expression as the deciding factor in human T, B, and myeloid cell proliferation or

13358- Boshuizen-Binnenwerk na proefdruk.indd 20 4-3-2016 16:17:14

Page 14: UvA-DARE (Digital Academic Repository) Cytokines in atherosclerosis… · foam cells, including oxysterols like oxLDL 38, oxygen radicals like nitric oxide 36 and several cytokines,

1

General Introduction

21

death. Journal of leukocyte biology. 2001;70:950-60.41. Goossens P, Gijbels MJ, Zernecke A, Eijgelaar W, Vergouwe MN, van der Made I, Vanderlocht J, Beckers L, Buurman WA, Daemen MJ, Kalinke U, Weber C, Lutgens E and de Winther MP. Myeloid type I interferon signaling promotes atherosclerosis by stimulating macrophage recruitment to lesions. Cell metabolism. 2010;12:142-53.42. Niessner A, Sato K, Chaikof EL, Colmegna I, Goronzy JJ and Weyand CM. Pathogen-sensing plasmacytoid dendritic cells stimulate cytotoxic T-cell function in the atherosclerotic plaque through interferon-alpha. Circulation. 2006;114:2482-9.43. Seimon T and Tabas I. Mechanisms and consequences of macrophage apoptosis in atherosclerosis. Journal of lipid research. 2009;50 Suppl:S382-7.44. Tabas I. Macrophage apoptosis in atherosclerosis: consequences on plaque progression and the role of endoplasmic reticulum stress. Antioxidants & redox signaling. 2009;11:2333-9.45. Isaacs A and Lindenmann J. Virus interference. I. The interferon. Proceedings of the Royal Society of London Series B, Biological sciences. 1957;147:258-67.46. Borden EC, Sen GC, Uze G, Silverman RH, Ransohoff RM, Foster GR and Stark GR. Interferons at age 50: past, current and future impact on biomedicine. Nature reviews Drug discovery. 2007;6:975-90.47. Lazarevic V and Glimcher LH. T-bet in disease. Nature immunology. 2011;12:597-606.48. McNab F, Mayer-Barber K, Sher A, Wack A and O'Garra A. Type I interferons in infectious disease. Nature reviews Immunology. 2015;15:87-103.49. Gilliet M, Cao W and Liu YJ. Plasmacytoid dendritic cells: sensing nucleic acids in viral infection and autoimmune diseases. Nature reviews Immunology. 2008;8:594-606.50. Schroder K, Hertzog PJ, Ravasi T and Hume DA. Interferon-gamma: an overview of signals, mechanisms and functions. Journal of leukocyte biology. 2004;75:163-89.51. Donnelly RP and Kotenko SV. Interferon-lambda: a new addition to an old family. Journal of interferon & cytokine research. 2010;30:555-64.52. Pestka S, Krause CD and Walter MR. Interferons, interferon-like cytokines, and their receptors. Immunological reviews. 2004;202:8-32.53. Stark GR and Darnell JE, Jr. The JAK-STAT pathway at twenty. Immunity. 2012;36:503-14.54. Boshuizen MC and de Winther MP. Interferons as Essential Modulators of Atherosclerosis. Arterioscler Thromb Vasc Biol 2015;35:1579-88.55. Gough DJ, Messina NL, Hii L, Gould JA, Sabapathy K, Robertson AP, Trapani JA, Levy DE, Hertzog PJ, Clarke CJ and Johnstone RW. Functional crosstalk between type I and II interferon through the regulated expression of STAT1. PLoS biology. 2010;8:e1000361.56. Taniguchi T and Takaoka A. A weak signal for strong responses: interferon-alpha/beta revisited. Nature reviews Molecular cell biology. 2001;2:378-86.57. Matsumoto M, Tanaka N, Harada H, Kimura T, Yokochi T, Kitagawa M, Schindler C and Taniguchi T. Activation of the transcription factor ISGF3 by interferon-gamma. Biological chemistry. 1999;380:699-703.58. van Boxel-Dezaire AH, Rani MR and Stark GR. Complex modulation of cell type-specific signaling in response to type I interferons. Immunity. 2006;25:361-72.59. Goubau D, Deddouche S and Reis e Sousa C. Cytosolic sensing of viruses. Immunity. 2013;38:855-69.60. Leber JH, Crimmins GT, Raghavan S, Meyer-Morse NP, Cox JS and Portnoy DA. Distinct TLR- and NLR-mediated transcriptional responses to an intracellular pathogen. PLoS pathogens. 2008;4:e6.61. Moynagh PN. TLR signalling and activation of IRFs: revisiting old friends from the NF-kappaB pathway. Trends in immunology. 2005;26:469-76.62. Paludan SR and Bowie AG. Immune sensing of DNA. Immunity. 2013;38:870-80.63. Ciancanelli MJ, Huang SX, Luthra P, Garner H, Itan Y, Volpi S, Lafaille FG, Trouillet C, Schmolke M, Albrecht RA, Israelsson E, Lim HK, Casadio M, Hermesh T, Lorenzo L, Leung LW, Pedergnana V, Boisson B, Okada S, Picard C, Ringuier B, Troussier F, Chaussabel D, Abel L, Pellier I, Notarangelo LD, Garcia-Sastre A, Basler CF, Geissmann F, Zhang SY, Snoeck HW and Casanova JL. Infectious disease. Life-threatening influenza and impaired interferon amplification in human IRF7 deficiency. Science. 2015;348:448-53.64. Honda K, Takaoka A and Taniguchi T. Type I interferon [corrected] gene induction by the interferon regulatory factor family of transcription factors. Immunity. 2006;25:349-60.65. Sakaguchi S, Negishi H, Asagiri M, Nakajima C, Mizutani T, Takaoka A, Honda K and Taniguchi T. Essential role of IRF-3 in lipopolysaccharide-induced interferon-beta gene expression and endotoxin shock. Biochemical and biophysical research communications. 2003;306:860-6.66. Tamura T, Yanai H, Savitsky D and Taniguchi T. The IRF family transcription factors in immunity and oncogenesis. Annual review of immunology. 2008;26:535-84.67. Colonna M, Trinchieri G and Liu YJ. Plasmacytoid dendritic cells in immunity. Nature immunology. 2004;5:1219-26.68. Ito T, Kanzler H, Duramad O, Cao W and Liu YJ. Specialization, kinetics, and repertoire of type 1 interferon responses by human plasmacytoid predendritic cells. Blood. 2006;107:2423-31.

13358- Boshuizen-Binnenwerk na proefdruk.indd 21 4-3-2016 16:17:15

Page 15: UvA-DARE (Digital Academic Repository) Cytokines in atherosclerosis… · foam cells, including oxysterols like oxLDL 38, oxygen radicals like nitric oxide 36 and several cytokines,

Chapter 1

22

69. Yan N and Chen ZJ. Intrinsic antiviral immunity. Nature immunology. 2012;13:214-22.70. Crouse J, Kalinke U and Oxenius A. Regulation of antiviral T cell responses by type I interferons. Nature reviews Immunology. 2015;15:231-42.71. Biron CA. Interferons alpha and beta as immune regulators--a new look. Immunity. 2001;14:661-4.72. Bowie AG and Unterholzner L. Viral evasion and subversion of pattern-recognition receptor signalling. Nature reviews Immunology. 2008;8:911-22.73. Randall RE and Goodbourn S. Interferons and viruses: an interplay between induction, signalling, antiviral responses and virus countermeasures. The Journal of general virology. 2008;89:1-47.74. Stifter SA and Feng CG. Interfering with immunity: detrimental role of type I IFNs during infection. Journal of immunology. 2015;194:2455-65.75. Teijaro JR, Ng C, Lee AM, Sullivan BM, Sheehan KC, Welch M, Schreiber RD, de la Torre JC and Oldstone MB. Persistent LCMV infection is controlled by blockade of type I interferon signaling. Science. 2013;340:207-11.76. Wilson EB, Yamada DH, Elsaesser H, Herskovitz J, Deng J, Cheng G, Aronow BJ, Karp CL and Brooks DG. Blockade of chronic type I interferon signaling to control persistent LCMV infection. Science. 2013;340:202-7.77. Pulliam L, Calosing C, Sun B, Grunfeld C and Rempel H. Monocyte activation from interferon-alpha in HIV infection increases acetylated LDL uptake and ROS production. Journal of interferon & cytokine research. 2014;34:822-8.78. Herbeuval JP, Nilsson J, Boasso A, Hardy AW, Kruhlak MJ, Anderson SA, Dolan MJ, Dy M, Andersson J and Shearer GM. Differential expression of IFN-alpha and TRAIL/DR5 in lymphoid tissue of progressor versus nonprogressor HIV-1-infected patients. Proceedings of the National Academy of Sciences of the United States of America. 2006;103:7000-5.79. Herbeuval JP and Shearer GM. HIV-1 immunopathogenesis: how good interferon turns bad. Clinical immunology. 2007;123:121-8.80. Finke D, Brinckmann UG, ter Meulen V and Liebert UG. Gamma interferon is a major mediator of antiviral defense in experimental measles virus-induced encephalitis. Journal of virology. 1995;69:5469-74.81. Huang S, Hendriks W, Althage A, Hemmi S, Bluethmann H, Kamijo R, Vilcek J, Zinkernagel RM and Aguet M. Immune response in mice that lack the interferon-gamma receptor. Science. 1993;259:1742-5.82. Shrestha B, Wang T, Samuel MA, Whitby K, Craft J, Fikrig E and Diamond MS. Gamma interferon plays a crucial early antiviral role in protection against West Nile virus infection. Journal of virology. 2006;80:5338-48.83. Malmgaard L and Paludan SR. Interferon (IFN)-alpha/beta, interleukin (IL)-12 and IL-18 coordinately induce production of IFN-gamma during infection with herpes simplex virus type 2. The Journal of general virology. 2003;84:2497-500.84. Sareneva T, Matikainen S, Kurimoto M and Julkunen I. Influenza A virus-induced IFN-alpha/beta and IL-18 synergistically enhance IFN-gamma gene expression in human T cells. Journal of immunology. 1998;160:6032-8.85. Nguyen KB, Cousens LP, Doughty LA, Pien GC, Durbin JE and Biron CA. Interferon alpha/beta-mediated inhibition and promotion of interferon gamma: STAT1 resolves a paradox. Nature immunology. 2000;1:70-6.86. Donze O, Dostie J and Sonenberg N. Regulatable expression of the interferon-induced double-stranded RNA dependent protein kinase PKR induces apoptosis and fas receptor expression. Virology. 1999;256:322-9.87. Clemens MJ. PKR--a protein kinase regulated by double-stranded RNA. The international journal of biochemistry & cell biology. 1997;29:945-9.88. Decker T, Stockinger S, Karaghiosoff M, Muller M and Kovarik P. IFNs and STATs in innate immunity to microorganisms. The Journal of clinical investigation. 2002;109:1271-7.89. Noppert SJ, Fitzgerald KA and Hertzog PJ. The role of type I interferons in TLR responses. Immunology and cell biology. 2007;85:446-57.90. Sheikh F, Dickensheets H, Gamero AM, Vogel SN and Donnelly RP. An essential role for IFN-beta in the induction of IFN-stimulated gene expression by LPS in macrophages. Journal of leukocyte biology. 2014;96:591-600.91. Devitt A, Lund PA, Morris AG and Pearce JH. Induction of alpha/beta interferon and dependent nitric oxide synthesis during Chlamydia trachomatis infection of McCoy cells in the absence of exogenous cytokine. Infection and immunity. 1996;64:3951-6.92. Gao JJ, Filla MB, Fultz MJ, Vogel SN, Russell SW and Murphy WJ. Autocrine/paracrine IFN-alphabeta mediates the lipopolysaccharide-induced activation of transcription factor Stat1alpha in mouse macrophages: pivotal role of Stat1alpha in induction of the inducible nitric oxide synthase gene. Journal of immunology. 1998;161:4803-10.93. Carrero JA. Confounding roles for type I interferons during bacterial and viral pathogenesis. International immunology. 2013;25:663-9.94. Decker T, Muller M and Stockinger S. The yin and yang of type I interferon activity in bacterial infection. Nature reviews Immunology. 2005;5:675-87.95. MacMicking J, Xie QW and Nathan C. Nitric oxide and macrophage function. Annual review of immunology. 1997;15:323-50.96. Green SJ, Nacy CA, Schreiber RD, Granger DL, Crawford RM, Meltzer MS and Fortier AH. Neutralization of gamma interferon and tumor necrosis factor alpha blocks in vivo synthesis of nitrogen oxides from L-arginine

13358- Boshuizen-Binnenwerk na proefdruk.indd 22 4-3-2016 16:17:15

Page 16: UvA-DARE (Digital Academic Repository) Cytokines in atherosclerosis… · foam cells, including oxysterols like oxLDL 38, oxygen radicals like nitric oxide 36 and several cytokines,

1

General Introduction

23

and protection against Francisella tularensis infection in Mycobacterium bovis BCG-treated mice. Infection and immunity. 1993;61:689-98.97. Held TK, Weihua X, Yuan L, Kalvakolanu DV and Cross AS. Gamma interferon augments macrophage activation by lipopolysaccharide by two distinct mechanisms, at the signal transduction level and via an autocrine mechanism involving tumor necrosis factor alpha and interleukin-1. Infection and immunity. 1999;67:206-12.98. Lorsbach RB, Murphy WJ, Lowenstein CJ, Snyder SH and Russell SW. Expression of the nitric oxide synthase gene in mouse macrophages activated for tumor cell killing. Molecular basis for the synergy between interferon-gamma and lipopolysaccharide. The Journal of biological chemistry. 1993;268:1908-13.99. Car BD, Eng VM, Schnyder B, Ozmen L, Huang S, Gallay P, Heumann D, Aguet M and Ryffel B. Interferon gamma receptor deficient mice are resistant to endotoxic shock. The Journal of experimental medicine. 1994;179:1437-44.100. Lu B, Ebensperger C, Dembic Z, Wang Y, Kvatyuk M, Lu T, Coffman RL, Pestka S and Rothman PB. Targeted disruption of the interferon-gamma receptor 2 gene results in severe immune defects in mice. Proceedings of the National Academy of Sciences of the United States of America. 1998;95:8233-8.101. van de Vosse E, Hoeve MA and Ottenhoff TH. Human genetics of intracellular infectious diseases: molecular and cellular immunity against mycobacteria and salmonellae. The Lancet Infectious diseases. 2004;4:739-49.102. Wu UI and Holland SM. Host susceptibility to non-tuberculous mycobacterial infections. The Lancet Infectious diseases. 2015.103. McNab FW, Ewbank J, Howes A, Moreira-Teixeira L, Martirosyan A, Ghilardi N, Saraiva M and O'Garra A. Type I IFN induces IL-10 production in an IL-27-independent manner and blocks responsiveness to IFN-gamma for production of IL-12 and bacterial killing in Mycobacterium tuberculosis-infected macrophages. Journal of immunology. 2014;193:3600-12.104. Redford PS, Mayer-Barber KD, McNab FW, Stavropoulos E, Wack A, Sher A and O'Garra A. Influenza A virus impairs control of Mycobacterium tuberculosis coinfection through a type I interferon receptor-dependent pathway. The Journal of infectious diseases. 2014;209:270-4.105. Teles RM, Graeber TG, Krutzik SR, Montoya D, Schenk M, Lee DJ, Komisopoulou E, Kelly-Scumpia K, Chun R, Iyer SS, Sarno EN, Rea TH, Hewison M, Adams JS, Popper SJ, Relman DA, Stenger S, Bloom BR, Cheng G and Modlin RL. Type I interferon suppresses type II interferon-triggered human anti-mycobacterial responses. Science. 2013;339:1448-53.106. Baechler EC, Batliwalla FM, Karypis G, Gaffney PM, Ortmann WA, Espe KJ, Shark KB, Grande WJ, Hughes KM, Kapur V, Gregersen PK and Behrens TW. Interferon-inducible gene expression signature in peripheral blood cells of patients with severe lupus. Proceedings of the National Academy of Sciences of the United States of America. 2003;100:2610-5.107. Crow MK. Type I interferon in the pathogenesis of lupus. Journal of immunology. 2014;192:5459-68.108. Al Gadban MM, Alwan MM, Smith KJ and Hammad SM. Accelerated vascular disease in systemic lupus erythematosus: role of macrophage. Clinical immunology. 2015;157:133-44.109. Kahlenberg JM and Kaplan MJ. The interplay of inflammation and cardiovascular disease in systemic lupus erythematosus. Arthritis research & therapy. 2011;13:203.110. Sozzani S, Bosisio D, Scarsi M and Tincani A. Type I interferons in systemic autoimmunity. Autoimmunity. 2010;43:196-203.111. Wu GC, Liu HR, Leng RX, Li XP, Li XM, Pan HF and Ye DQ. Subclinical atherosclerosis in patients with systemic lupus erythematosus: A systemic review and meta-analysis. Autoimmunity reviews. 2015.112. Lauwerys BR, Ducreux J and Houssiau FA. Type I interferon blockade in systemic lupus erythematosus: where do we stand? Rheumatology. 2014;53:1369-76.113. Pollard KM, Cauvi DM, Toomey CB, Morris KV and Kono DH. Interferon-gamma and systemic autoimmunity. Discovery medicine. 2013;16:123-31.114. Skurkovich S, Boiko A, Beliaeva I, Buglak A, Alekseeva T, Smirnova N, Kulakova O, Tchechonin V, Gurova O, Deomina T, Favorova OO, Skurkovic B and Gusev E. Randomized study of antibodies to IFN-gamma and TNF-alpha in secondary progressive multiple sclerosis. Multiple sclerosis. 2001;7:277-84.115. Cui D, Huang G, Yang D, Huang B and An B. Efficacy and safety of interferon-gamma-targeted therapy in Crohn's disease: a systematic review and meta-analysis of randomized controlled trials. Clinics and research in hepatology and gastroenterology. 2013;37:507-13.116. Bouma G and Strober W. The immunological and genetic basis of inflammatory bowel disease. Nature reviews Immunology. 2003;3:521-33.117. Group IMS. Interferon beta-1b is effective in relapsing-remitting multiple sclerosis. I. Clinical results of a multicenter, randomized, double-blind, placebo-controlled trial. The IFNB Multiple Sclerosis Study Group. Neurology. 1993;43:655-61.118. Rudick RA and Cutter G. Interferon-beta for multiple sclerosis: Long-term benefits? Annals of neurology. 2007;61:283-5.119. Kamal SM. Pharmacogenetics of hepatitis C: transition from interferon-based therapies to direct-acting

13358- Boshuizen-Binnenwerk na proefdruk.indd 23 4-3-2016 16:17:15

Page 17: UvA-DARE (Digital Academic Repository) Cytokines in atherosclerosis… · foam cells, including oxysterols like oxLDL 38, oxygen radicals like nitric oxide 36 and several cytokines,

Chapter 1

24

antiviral agents. Hepatic medicine : evidence and research. 2014;6:61-77.120. akamoto S, Kanda T, Shirasawa H and Yokosuka O. Antiviral therapies for chronic hepatitis C virus infection with cirrhosis. World journal of hepatology. 2015;7:1133-41.121. Reddy KR, Zeuzem S, Zoulim F, Weiland O, Horban A, Stanciu C, Villamil FG, Andreone P, George J, Dammers E, Fu M, Kurland D, Lenz O, Ouwerkerk-Mahadevan S, Verbinnen T, Scott J and Jessner W. Simeprevir versus telaprevir with peginterferon and ribavirin in previous null or partial responders with chronic hepatitis C virus genotype 1 infection (ATTAIN): a randomised, double-blind, non-inferiority phase 3 trial. The Lancet Infectious diseases. 2015;15:27-35.122. Bekisz J, Sato Y, Johnson C, Husain SR, Puri RK and Zoon KC. Immunomodulatory effects of interferons in malignancies. Journal of interferon & cytokine research. 2013;33:154-61.123. Pizzi M, Silver RT, Barel A and Orazi A. Recombinant interferon-alpha in myelofibrosis reduces bone marrow fibrosis, improves its morphology and is associated with clinical response. Modern pathology : an official journal of the United States and Canadian Academy of Pathology, Inc. 2015;28:1315-23.124. Dunn GP, Ikeda H, Bruce AT, Koebel C, Uppaluri R, Bui J, Chan R, Diamond M, White JM, Sheehan KC and Schreiber RD. Interferon-gamma and cancer immunoediting. Immunologic research. 2005;32:231-45.125. Ikeda H, Old LJ and Schreiber RD. The roles of IFN gamma in protection against tumor development and cancer immunoediting. Cytokine & growth factor reviews. 2002;13:95-109.126. Zaidi MR and Merlino G. The two faces of interferon-gamma in cancer. Clinical cancer research : an official journal of the American Association for Cancer Research. 2011;17:6118-24.127. Katayama K, Kasahara A, Sasaki Y, Kashiwagi T, Naito M, Masuzawa M, Katoh M, Yoshihara H, Kamada T, Mukuda T, Hijioka T, Hori M and Hayashi N. Immunological response to interferon-gamma priming prior to interferon-alpha treatment in refractory chronic hepatitis C in relation to viral clearance. Journal of viral hepatitis. 2001;8:180-5.128. Miller CH, Maher SG and Young HA. Clinical Use of Interferon-gamma. Annals of the New York Academy of Sciences. 2009;1182:69-79.129. Buono C, Come CE, Stavrakis G, Maguire GF, Connelly PW and Lichtman AH. Influence of interferon-gamma on the extent and phenotype of diet-induced atherosclerosis in the LDLR-deficient mouse. Arterioscler Thromb Vasc Biol 2003;23:454-60.130. Daissormont IT, Christ A, Temmerman L, Sampedro Millares S, Seijkens T, Manca M, Rousch M, Poggi M, Boon L, van der Loos C, Daemen M, Lutgens E, Halvorsen B, Aukrust P, Janssen E and Biessen EA. Plasmacytoid dendritic cells protect against atherosclerosis by tuning T-cell proliferation and activity. Circulation research. 2011;109:1387-95.131. Doring Y, Manthey HD, Drechsler M, Lievens D, Megens RT, Soehnlein O, Busch M, Manca M, Koenen RR, Pelisek J, Daemen MJ, Lutgens E, Zenke M, Binder CJ, Weber C and Zernecke A. Auto-antigenic protein-DNA complexes stimulate plasmacytoid dendritic cells to promote atherosclerosis. Circulation. 2012;125:1673-83.132. Gupta S, Pablo AM, Jiang X, Wang N, Tall AR and Schindler C. IFN-gamma potentiates atherosclerosis in ApoE knock-out mice. The Journal of clinical investigation. 1997;99:2752-61.133. Koga M, Kai H, Yasukawa H, Kato S, Yamamoto T, Kawai Y, Kusaba K, Seki Y, Kai M, Egashira K, Kataoka Y and Imaizumi T. Postnatal blocking of interferon-gamma function prevented atherosclerotic plaque formation in apolipoprotein E-knockout mice. Hypertension research : official journal of the Japanese Society of Hypertension. 2007;30:259-67.134. Koga M, Kai H, Yasukawa H, Yamamoto T, Kawai Y, Kato S, Kusaba K, Kai M, Egashira K, Kataoka Y and Imaizumi T. Inhibition of progression and stabilization of plaques by postnatal interferon-gamma function blocking in ApoE-knockout mice. Circulation research. 2007;101:348-56.135. Levy Z, Rachmani R, Trestman S, Dvir A, Shaish A, Ravid M and Harats D. Low-dose interferon-alpha accelerates atherosclerosis in an LDL receptor-deficient mouse model. European journal of internal medicine. 2003;14:479-483.136. Macritchie N, Grassia G, Sabir SR, Maddaluno M, Welsh P, Sattar N, Ialenti A, Kurowska-Stolarska M, McInnes IB, Brewer JM, Garside P and Maffia P. Plasmacytoid dendritic cells play a key role in promoting atherosclerosis in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol 2012;32:2569-79.137. Niwa T, Wada H, Ohashi H, Iwamoto N, Ohta H, Kirii H, Fujii H, Saito K and Seishima M. Interferon-gamma produced by bone marrow-derived cells attenuates atherosclerotic lesion formation in LDLR-deficient mice. Journal of atherosclerosis and thrombosis. 2004;11:79-87.138. Whitman SC, Ravisankar P and Daugherty A. IFN-gamma deficiency exerts gender-specific effects on atherogenesis in apolipoprotein E-/- mice. Journal of interferon & cytokine research. 2002;22:661-70.139. Whitman SC, Ravisankar P, Elam H and Daugherty A. Exogenous interferon-gamma enhances atherosclerosis in apolipoprotein E-/- mice. The American journal of pathology. 2000;157:1819-24.140. Fiorentino DF, Bond MW and Mosmann TR. Two types of mouse T helper cell. IV. Th2 clones secrete a factor that inhibits cytokine production by Th1 clones. The Journal of experimental medicine. 1989;170:2081-95.141. Yang X, Gartner J, Zhu L, Wang S and Brunham RC. IL-10 gene knockout mice show enhanced Th1-like protective immunity and absent granuloma formation following Chlamydia trachomatis lung infection. Journal of

13358- Boshuizen-Binnenwerk na proefdruk.indd 24 4-3-2016 16:17:15

Page 18: UvA-DARE (Digital Academic Repository) Cytokines in atherosclerosis… · foam cells, including oxysterols like oxLDL 38, oxygen radicals like nitric oxide 36 and several cytokines,

1

General Introduction

25

immunology. 1999;162:1010-7.142. Murray PJ and Young RA. Increased antimycobacterial immunity in interleukin-10-deficient mice. Infection and immunity. 1999;67:3087-95.143. Moore KW, de Waal Malefyt R, Coffman RL and O'Garra A. Interleukin-10 and the interleukin-10 receptor. Annual review of immunology. 2001;19:683-765.144. Saxena A, Khosraviani S, Noel S, Mohan D, Donner T and Hamad AR. Interleukin-10 paradox: A potent immunoregulatory cytokine that has been difficult to harness for immunotherapy. Cytokine. 2015;74:27-34.145. Guichelaar T, ten Brink CB, van Kooten PJ, Berlo SE, Broeren CP, van Eden W and Broere F. Autoantigen-specific IL-10-transduced T cells suppress chronic arthritis by promoting the endogenous regulatory IL-10 response. Journal of immunology. 2008;180:1373-81.146. Mauri C and Bosma A. Immune regulatory function of B cells. Annual review of immunology. 2012;30:221-41.147. Ding L and Shevach EM. IL-10 inhibits mitogen-induced T cell proliferation by selectively inhibiting macrophage costimulatory function. Journal of immunology. 1992;148:3133-9.148. Ding Y, Chen D, Tarcsafalvi A, Su R, Qin L and Bromberg JS. Suppressor of cytokine signaling 1 inhibits IL-10-mediated immune responses. Journal of immunology. 2003;170:1383-91.149. Schottelius AJ, Mayo MW, Sartor RB and Baldwin AS, Jr. Interleukin-10 signaling blocks inhibitor of kappaB kinase activity and nuclear factor kappaB DNA binding. The Journal of biological chemistry. 1999;274:31868-74.150. Schreiber S, Heinig T, Thiele HG and Raedler A. Immunoregulatory role of interleukin 10 in patients with inflammatory bowel disease. Gastroenterology. 1995;108:1434-44.151. Shah N, Kammermeier J, Elawad M and Glocker EO. Interleukin-10 and interleukin-10-receptor defects in inflammatory bowel disease. Current allergy and asthma reports. 2012;12:373-9.152. Shouval DS, Biswas A, Goettel JA, McCann K, Conaway E, Redhu NS, Mascanfroni ID, Al Adham Z, Lavoie S, Ibourk M, Nguyen DD, Samsom JN, Escher JC, Somech R, Weiss B, Beier R, Conklin LS, Ebens CL, Santos FG, Ferreira AR, Sherlock M, Bhan AK, Muller W, Mora JR, Quintana FJ, Klein C, Muise AM, Horwitz BH and Snapper SB. Interleukin-10 receptor signaling in innate immune cells regulates mucosal immune tolerance and anti-inflammatory macrophage function. Immunity. 2014;40:706-19.153. Hirotani M, Niino M, Fukazawa T, Kikuchi S, Yabe I, Hamada S, Tajima Y and Sasaki H. Decreased IL-10 production mediated by Toll-like receptor 9 in B cells in multiple sclerosis. Journal of neuroimmunology. 2010;221:95-100.154. Mongan AE, Ramdahin S and Warrington RJ. Interleukin-10 response abnormalities in systemic lupus erythematosus. Scandinavian journal of immunology. 1997;46:406-12.155. Llorente L, Richaud-Patin Y, Garcia-Padilla C, Claret E, Jakez-Ocampo J, Cardiel MH, Alcocer-Varela J, Grangeot-Keros L, Alarcon-Segovia D, Wijdenes J, Galanaud P and Emilie D. Clinical and biologic effects of anti-interleukin-10 monoclonal antibody administration in systemic lupus erythematosus. Arthritis and rheumatism. 2000;43:1790-800.156. Mallat Z, Heymes C, Ohan J, Faggin E, Leseche G and Tedgui A. Expression of interleukin-10 in advanced human atherosclerotic plaques: relation to inducible nitric oxide synthase expression and cell death. Arterioscler Thromb Vasc Biol 1999;19:611-6.157. Caligiuri G, Rudling M, Ollivier V, Jacob MP, Michel JB, Hansson GK and Nicoletti A. Interleukin-10 deficiency increases atherosclerosis, thrombosis, and low-density lipoproteins in apolipoprotein E knockout mice. Molecular medicine. 2003;9:10-7.158. Potteaux S, Esposito B, van Oostrom O, Brun V, Ardouin P, Groux H, Tedgui A and Mallat Z. Leukocyte-derived interleukin 10 is required for protection against atherosclerosis in low-density lipoprotein receptor knockout mice. Arteriosclerosis, thrombosis, and vascular biology. 2004;24:1474-8.159. Kanters E, Gijbels MJ, van der Made I, Vergouwe MN, Heeringa P, Kraal G, Hofker MH and de Winther MP. Hematopoietic NF-kappaB1 deficiency results in small atherosclerotic lesions with an inflammatory phenotype. Blood. 2004;103:934-40.160. Khan JA, Cao M, Kang BY, Liu Y, Mehta JL and Hermonat PL. AAV/hSTAT3-gene delivery lowers aortic inflammatory cell infiltration in LDLR KO mice on high cholesterol. Atherosclerosis. 2010;213:59-66.161. Liu Y, Li D, Chen J, Xie J, Bandyopadhyay S, Zhang D, Nemarkommula AR, Liu H, Mehta JL and Hermonat PL. Inhibition of atherogenesis in LDLR knockout mice by systemic delivery of adeno-associated virus type 2-hIL-10. Atherosclerosis. 2006;188:19-27.162. Namiki M, Kawashima S, Yamashita T, Ozaki M, Sakoda T, Inoue N, Hirata K, Morishita R, Kaneda Y and Yokoyama M. Intramuscular gene transfer of interleukin-10 cDNA reduces atherosclerosis in apolipoprotein E-knockout mice. Atherosclerosis. 2004;172:21-9.163. Pinderski LJ, Fischbein MP, Subbanagounder G, Fishbein MC, Kubo N, Cheroutre H, Curtiss LK, Berliner JA and Boisvert WA. Overexpression of interleukin-10 by activated T lymphocytes inhibits atherosclerosis in LDL receptor-deficient Mice by altering lymphocyte and macrophage phenotypes. Circulation research. 2002;90:1064-71.164. Von Der Thusen JH, Kuiper J, Fekkes ML, De Vos P, Van Berkel TJ and Biessen EA. Attenuation of atherogenesis by systemic and local adenovirus-mediated gene transfer of interleukin-10 in LDLr-/- mice. FASEB journal : official publication of the Federation of American Societies for Experimental Biology. 2001;15:2730-2.

13358- Boshuizen-Binnenwerk na proefdruk.indd 25 4-3-2016 16:17:15

Page 19: UvA-DARE (Digital Academic Repository) Cytokines in atherosclerosis… · foam cells, including oxysterols like oxLDL 38, oxygen radicals like nitric oxide 36 and several cytokines,

Chapter 1

26

165. Halvorsen B, Holm S, Yndestad A, Scholz H, Sagen EL, Nebb H, Holven KB, Dahl TB and Aukrust P. Interleukin-10 increases reverse cholesterol transport in macrophages through its bidirectional interaction with liver X receptor alpha. Biochemical and biophysical research communications. 2014;450:1525-30.166. Han X, Kitamoto S, Lian Q and Boisvert WA. Interleukin-10 facilitates both cholesterol uptake and efflux in macrophages. The Journal of biological chemistry. 2009;284:32950-8.167. Han X, Kitamoto S, Wang H and Boisvert WA. Interleukin-10 overexpression in macrophages suppresses atherosclerosis in hyperlipidemic mice. FASEB journal : official publication of the Federation of American Societies for Experimental Biology. 2010;24:2869-80.168. Holven KB, Halvorsen B, Bjerkeli V, Damas JK, Retterstol K, Morkrid L, Ose L, Aukrust P and Nenseter MS. Impaired inhibitory effect of interleukin-10 on the balance between matrix metalloproteinase-9 and its inhibitor in mononuclear cells from hyperhomocysteinemic subjects. Stroke; a journal of cerebral circulation. 2006;37:1731-6.

13358- Boshuizen-Binnenwerk na proefdruk.indd 26 4-3-2016 16:17:15