• No results found

Immunotolerance during bacterial pneumonia and sepsis - Chapter 1: General introduction and outline of this thesis

N/A
N/A
Protected

Academic year: 2021

Share "Immunotolerance during bacterial pneumonia and sepsis - Chapter 1: General introduction and outline of this thesis"

Copied!
11
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)

UvA-DARE (Digital Academic Repository)

Immunotolerance during bacterial pneumonia and sepsis

Hoogerwerf, J.J.

Publication date

2010

Link to publication

Citation for published version (APA):

Hoogerwerf, J. J. (2010). Immunotolerance during bacterial pneumonia and sepsis.

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)

7

1

1

General Introduction

C

C

h

h

a

a

p

p

t

t

e

e

r

r

(3)

8

Chapter 1

Introduction

Infectious diseases are a major cause of morbidity and mortality worldwide. Massive

use of antibiotics promotes pathogen resistance and as a consequence, the incidence

of drug-resistant bacteria is increasing (WHO; The world health report 2000, Health

Systems: improving performance). Therefore, it is of the utmost importance to expand

our comprehension of host responses against invading pathogens in order to develop

new treatment strategies. This thesis focuses on the immune response against

bacteria during (nosocomial) pneumonia and sepsis.

Bacterial pneumonia

Bacterial pneumonia is one of the most common infectious diseases and the most

frequent source of sepsis

1

. Depending on the circumstances in which the patient

acquires pneumonia, community-acquired pneumonia can be distinguished from

hospital-acquired (nosocomial) pneumonia occurring in patients with pre-existing

conditions. The most frequent causative pathogen in community-acquired pneumonia

is Streptococcus pneumoniae

2

, whereas Pseudomonas aeruginosa and Klebsiella

pneumoniae are prominent bacteria causing nosocomial pneumonia

3

.

The host response against bacterial pneumonia

The airways are in direct contact with the outside environment and therefore

continuously exposed to respiratory pathogens. The first line of defense in the upper

respiratory tract is formed by physical mechanisms like coughing and sneezing. When

respiratory pathogens overcome these structural defenses and enter the alveolar

space, the innate immune response is primarily responsible for the elimination of

these pathogens. Upon recognition of invading pathogens, innate immune cells like

respiratory epithelial cells and resident alveolar macrophages will then orchestrate an

innate immune response leading to the secretion of cytokines, chemokines and

antimicrobial peptides

4

(Figure 1.1). Moreover, alveolar macrophages are able to bind

and phagocytose pathogens and subsequently kill them intracellularly. The secreted

cytokines and chemokines mediate recruitment and activation of neutrophils from the

circulation to the site of inflammation in the lung. Recruited neutrophils effectively

phagocytose and eliminate pathogens

5,6

(Figure 1.1). Besides the elimination of

pathogens, alveolar macrophages are able to phagocytose apoptotic neutrophils and

thereby contribute to the resolution of pneumonia

7

. Furthermore, the innate immune

response is thought to orchestrate the adaptive immune response that primarily

consists of T- and B-cell responses that provide specific memory of infection

8

.

(4)

Introduction

9

Figure 1.1 Normal (left side) and inflamed (right side) alveolus (adapted from 9

).

Recognition of pathogens by Toll-like receptors

In the alveolar space, innate immune cells distinguish potential pathogens from self,

using receptors that recognize highly conserved motifs (pathogen-associated

molecular patterns; PAMPs) on pathogens that are not found in higher eukaryotes.

The receptors recognizing these PAMPs have been termed “pattern recognition

receptors” or PRRs. Among other receptor families, Toll-like receptors (TLRs) occupy a

central position as PRRs in the initiation of cellular innate immune responses

5,10

. TLRs

are distinguished from other PRRs by their ability to recognize, but moreover, to

discriminate between different classes of pathogens. Presently, thirteen TLRs are

described, of which TLR2 and TLR4 are of great importance in bacterial pneumonia.

(5)

10

Chapter 1

TLR4 recognizes lipopolysaccharide (LPS), part of the outer membrane of

gram-negative bacteria

11

, whereas TLR2 recognizes lipoteichoic acid (LTA), a major

constituent of gram-positive bacteria

12-17

(Figure 1.2). Although many investigations

have been published on the effects of LPS in humans

18,19

, the human response to LTA

in vivo has never been studied. Knowledge of the effects of LTA in humans is

important considering the prominent place of gram-positive pathogens in both

community-acquired and nosocomial infections.

Figure 1.2 Overview of PAMPs as part of the membrane of gram-positive and gram-negative bacteria (adapted from 20

)

Coagulation and fibrinolysis

The acute inflammatory response is frequently accompanied by activation of

coagulation and inhibition of fibrinolysis in the bronchoalveolar space during

pneumonia

21-24

. These hemostatic effects can be considered host-protective in

containing inflammation to the site of infection

25

. However, procoagulant activity can

also be disadvantageous by modulating inflammatory activity, leading to excessive

activation of inflammation in the alveolar compartment during pneumonia

26

. LPS have

been demonstrated to reproduce the hemostatic alterations of pneumonia in the

lungs of healthy humans when administered in the airways by bronchial

instillation

27-29

. In contrast, knowledge of the hemostatic balance in inflammation

caused by gram-positive pathogens is limited.

Sepsis

Sepsis is one of the leading causes of death in the Western world and its mortality

rate remains unacceptably high between 20-40%

30

. Sepsis is a heterogenous clinical

(6)

Introduction

11

syndrome broadly defined as the systemic host response to an infection. Although any

bacterial infection can progress and cause systemic inflammation, respiratory tract

infections are the most common source for sepsis

31,32

. Furthermore, patients with

sepsis are prone to develop nosocomial infections, in particular pneumonia, which has

a large impact on outcome.

Immunotolerance in sepsis

Until recently, the high mortality rate of sepsis was thought to be the result of an

uncontrolled hyperinflammatory response of the host to an infection. However,

failure of clinical trials with anti-inflammatory strategies in sepsis patients and the

development of animal models more closely imitating clinical sepsis have led to the

reconsideration of the pathogenesis of sepsis. Sepsis is currently considered a

misbalance between hyperinflammatory responses and immunotolerance (Figure

1.3).

Figure 1.3 Misbalance of hyperinflammation and immunotolerance in the host response during sepsis (adapted from 20

).

Hyperinflammation is designed to eliminate invading pathogens, but is at the same

time responsible for tissue damage. In contrast, immunotolerance is believed to

dampen excessive inflammation and subsequent tissue damage, but may contribute

(7)

12

Chapter 1

to the susceptibility of septic patients to nosocomial infections

20,33-35

. Clear evidence

of immunotolerance in sepsis comes from studies showing hyporesponsiveness of

immunocompetent cells upon recurrent exposures to microbial agents or products

(often referred to as tolerance to LPS)

36-38

.

Various mechanisms are thought to contribute to immunotolerance, among which

anti-inflammatory cytokines such as interleukin (IL)-10 and transforming

growth-factor (TGF)-β. Likewise, deregulated apoptosis of lymphocytes, dendritic cells,

monocytes/macrophages and granulocytes, has been implicated to play a role in

immunotolerance

20,33,34,39,40

(Figure 1.3).

Alongside upregulation of anti-inflammatory mediators and deregulated apoptosis of

immune cells, inhibitors of TLRs such as MyD88 short, A20, interleukin-1

receptor-associated kinase (IRAK)-M and ST2 are thought to play a role in the immunotolerance

in septic patients

41-43

(Figure 1.3 and 1.4).

ST2

The receptor ST2 emerges as a

transmembrane variant (ST2L) and a

soluble secreted variant (sST2).

Originally described as a Th2 marker

44

,

several other cell-types also express

ST2 including mast cells

45

, eosinophils

46

and macrophages

47

. ST2L is linked to

important Th2 effector functions

48-51

,

but concomitantly, ST2L has been

shown to play an important negative

regulatory function in TLR signaling

43

(Figure 1.4). Therefore, ST2 is thought

to play a role in the

immuno-suppression in septic patients. Soluble

ST2 probably acts as a decoy receptor

by binding IL-33 (ligand of ST2L),

thereby inhibiting signaling by ST2L

52,53

.

Figure 1.4 Overview of extra- and intracellular Toll-like receptor regulators54

(8)

Introduction

13

IRAK-M

IRAK-M is an intracellular proximal inhibitor of TLR signaling expressed by epithelial

cells and macrophages in the lung. IRAK-M inhibits the IRAK-1/IRAK-4 complex and

thereby mitigates intracellular responses elicited by all MyD88 dependent receptors

55

.

Considering its central position in the regulation of TLR signaling and its expression in

the two most prominent resident cells in the bronchoalveolar space, IRAK-M likely

plays an important role in the host response to bacterial infection. Importantly, in

septic mice, enhanced IRAK-M expression in pulmonary macrophages resulted in a

strongly impaired host defense response during secondary (i.e. following sepsis)

Pseudomonas pneumonia, suggesting that IRAK-M contributes to immunotolerance

42

.

Outline of this thesis

The general aim of this thesis is to enhance our knowledge of the host response to

bacterial pneumonia and sepsis and to increase our insight into the underlying

mechanisms of immunotolerance as a feature of patients with sepsis. In the first part

we used a model of 1) lung inflammation: bronchial instillation of LTA or LPS in the

human lung in healthy volunteers, in order to mimick the pulmonary response during

gram-positive or gram-negative pneumonia respectively; and 2) lung infection:

K. pneumoniae pneumonia in mice. Chapter 2 describes the inflammatory host

response to LTA versus known LPS-induced responses in the human bronchoalveolar

space. In chapter 3 the effects of LTA on hemostasis in the human lung was described

and compared with the known hemostatic effects to LPS. In chapter 4 we investigated

the effect of in vivo LPS bronchial instillation on the responsiveness of alveolar

macrophages to further stimulation with bacterial products. Chapter 5 reports on the

role of TLR-inhibitor IRAK-M in the host response during gram-negative pneumonia. In

the next part the influence of apoptosis and TLR-inhibitor ST2 was investigated in

sepsis. Chapter 6 describes the gene expression profiles of apoptosis regulators in

purified leukocyte subsets in human sepsis. The extent of soluble ST2 (a decoy

receptor for TLR inhibitor ST2) release during human sepsis was investigated in

Chapter 7. Last, the role of ST2 in modulating host defense in the lung during sepsis

was investigated using a murine model of cecal ligation and puncture (CLP)-induced

sepsis followed by secondary challenge with intranasal P. aeruginosa (chapter 8).

(9)

14

Chapter 1

References

1. Laterre PF, Garber G, Levy H, Wunderink R, Kinasewitz GT, Sollet JP, Maki DG, Bates B, Yan SC, Dhainaut JF. Severe community-acquired pneumonia as a cause of severe sepsis: data from the PROWESS study. Crit Care Med 2005;33:952-961.

2. Bartlett JG, Mundy LM. Community-acquired pneumonia. N Engl J Med 1995;333:1618-1624. 3. Kofteridis DP, Papadakis JA, Bouros D, Nikolaides P, Kioumis G, Levidiotou S, Maltezos E, Kastanakis S,

Kartali S, Gikas A. Nosocomial lower respiratory tract infections: prevalence and risk factors in 14 Greek hospitals. Eur J Clin Microbiol Infect Dis 2004;23:888-891.

4. Martin TR, Frevert CW. Innate immunity in the lungs. Proc Am Thorac Soc 2005;2:403-411. 5. Akira S, Uematsu S, Takeuchi O. Pathogen recognition and innate immunity. Cell 2006;124:783-801. 6. Beutler B, Jiang Z, Georgel P, Crozat K, Croker B, Rutschmann S, Du X, Hoebe K. Genetic analysis of

host resistance: Toll-like receptor signaling and immunity at large. Annu Rev Immunol 2006;24: 353-389.

7. Knapp S, Schultz MJ, van der Poll T. Pneumonia models and innate immunity to respiratory bacterial pathogens. Shock 2005;24 Suppl 1:12-18.

8. Pasare C, Medzhitov R. Toll-like receptors: linking innate and adaptive immunity. Adv Exp Med Biol 2005;560:11-18.

9. Ware LB, Matthay MA. The acute respiratory distress syndrome. N Engl J Med 2000;342:1334-1349. 10. Beutler B. Inferences, questions and possibilities in Toll-like receptor signalling. Nature 2004;430:

257-263.

11. Beutler B, Rietschel ET. Innate immune sensing and its roots: the story of endotoxin. Nat Rev Immunol 2003;3:169-176.

12. Lehner MD, Morath S, Michelsen KS, Schumann RR, Hartung T. Induction of cross-tolerance by lipopolysaccharide and highly purified lipoteichoic acid via different Toll-like receptors independent of paracrine mediators. J Immunol 2001;166:5161-5167.

13. Opitz B, Schroder NW, Spreitzer I, Michelsen KS, Kirschning CJ, Hallatschek W, Zahringer U, Hartung T, Gobel UB, Schumann RR. Toll-like receptor-2 mediates Treponema glycolipid and lipoteichoic acid-induced NF-kappaB translocation. J Biol Chem 2001;276:22041-22047.

14. Triantafilou M, Manukyan M, Mackie A, Morath S, Hartung T, Heine H, Triantafilou K. Lipoteichoic acid and toll-like receptor 2 internalization and targeting to the Golgi are lipid raft-dependent. J Biol Chem 2004;279:40882-40889.

15. Schroder NW, Morath S, Alexander C, Hamann L, Hartung T, Zahringer U, Gobel UB, Weber JR, Schumann RR. Lipoteichoic acid (LTA) of Streptococcus pneumoniae and Staphylococcus aureus activates immune cells via Toll-like receptor (TLR)-2, lipopolysaccharide-binding protein (LBP), and CD14, whereas TLR-4 and MD-2 are not involved. J Biol Chem 2003;278:15587-15594.

16. Knapp S, Wieland CW, van 't Veer C, Takeuchi O, Akira S, Florquin S, van der Poll T. Toll-like receptor 2 plays a role in the early inflammatory response to murine pneumococcal pneumonia but does not contribute to antibacterial defense. J Immunol 2004;172:3132-3138.

17. Han SH, Kim JH, Martin M, Michalek SM, Nahm MH. Pneumococcal lipoteichoic acid (LTA) is not as potent as staphylococcal LTA in stimulating Toll-like receptor 2. Infect Immun 2003;71:5541-5548. 18. Thorn J. The inflammatory response in humans after inhalation of bacterial endotoxin: a review.

Inflamm Res 2001;50:254-261.

19. Lowry SF. Human endotoxemia: a model for mechanistic insight and therapeutic targeting. Shock 2005;24 Suppl 1:94-100.

20. van der Poll T, Opal SM. Host-pathogen interactions in sepsis. Lancet Infect Dis 2008;8:32-43. 21. Gunther A, Mosavi P, Heinemann S, Ruppert C, Muth H, Markart P, Grimminger F, Walmrath D,

Temmesfeld-Wollbruck B, Seeger W. Alveolar fibrin formation caused by enhanced procoagulant and depressed fibrinolytic capacities in severe pneumonia. Comparison with the acute respiratory distress syndrome. Am J Respir Crit Care Med 2000;161:454-462.

22. Ware LB, Fang X, Matthay MA. Protein C and thrombomodulin in human acute lung injury. Am J

(10)

Introduction

15

23. Rijneveld AW, Florquin S, Bresser P, Levi M, De Waard V, Lijnen R, Van Der Zee JS, Speelman P, Carmeliet P, Van Der Poll T. Plasminogen activator inhibitor type-1 deficiency does not influence the outcome of murine pneumococcal pneumonia. Blood 2003;102:934-939.

24. Rijneveld AW, Weijer S, Bresser P, Florquin S, Vlasuk GP, Rote WE, Spek CA, Reitsma PH, van der Zee JS, Levi M, van der Poll T. Local activation of the tissue factor-factor VIIa pathway in patients with pneumonia and the effect of inhibition of this pathway in murine pneumococcal pneumonia. Crit Care

Med 2006;34:1725-1730.

25. Opal SM. Phylogenetic and functional relationships between coagulation and the innate immune response. Crit Care Med 2000;28:S77-80.

26. Levi M, Schultz MJ, Rijneveld AW, van der Poll T. Bronchoalveolar coagulation and fibrinolysis in endotoxemia and pneumonia. Crit Care Med 2003;31:S238-242.

27. Maris NA, de Vos AF, Bresser P, van der Zee JS, Jansen HM, Levi M, van der Poll T. Salmeterol enhances pulmonary fibrinolysis in healthy volunteers. Crit Care Med 2007;35:57-63.

28. Maris NA, de Vos AF, Dessing MC, Spek CA, Lutter R, Jansen HM, van der Zee JS, Bresser P, van der Poll T. Antiinflammatory effects of salmeterol after inhalation of lipopolysaccharide by healthy volunteers. Am J Respir Crit Care Med 2005;172:878-884.

29. van der Poll T, Levi M, Nick JA, Abraham E. Activated protein C inhibits local coagulation after intrapulmonary delivery of endotoxin in humans. Am J Respir Crit Care Med 2005;171:1125-1128. 30. Riedemann NC, Guo RF, Ward PA. The enigma of sepsis. J Clin Invest 2003;112:460-467. 31. Angus DC, Wax RS. Epidemiology of sepsis: an update. Crit Care Med 2001;29:S109-116. 32. Wheeler AP, Bernard GR. Treating patients with severe sepsis. N Engl J Med 1999;340:207-214. 33. Hotchkiss RS, Karl IE. The pathophysiology and treatment of sepsis. N Engl J Med 2003;348:138-150. 34. Hotchkiss RS, Nicholson DW. Apoptosis and caspases regulate death and inflammation in sepsis. Nat

Rev Immunol 2006;6:813-822.

35. Volk HD, Reinke P, Docke WD. Clinical aspects: from systemic inflammation to 'immunoparalysis'.

Chem Immunol 2000;74:162-177.

36. Adib-Conquy M, Adrie C, Moine P, Asehnoune K, Fitting C, Pinsky MR, Dhainaut JF, Cavaillon JM. NF-kappaB expression in mononuclear cells of patients with sepsis resembles that observed in lipopolysaccharide tolerance. Am J Respir Crit Care Med 2000;162:1877-1883.

37. Fitting C, Dhawan S, Cavaillon JM. Compartmentalization of tolerance to endotoxin. J Infect Dis 2004; 189:1295-1303.

38. West MA, Heagy W. Endotoxin tolerance: A review. Crit Care Med 2002;30:S64-S73.

39. Wesche DE, Lomas-Neira JL, Perl M, Chung CS, Ayala A. Leukocyte apoptosis and its significance in sepsis and shock. J Leukoc Biol 2005;78:325-337.

40. Brown KA, Brain SD, Pearson JD, Edgeworth JD, Lewis SM, Treacher DF. Neutrophils in development of multiple organ failure in sepsis. Lancet 2006;368:157-169.

41. Liew FY, Xu D, Brint EK, O'Neill LA. Negative regulation of toll-like receptor-mediated immune responses. Nat Rev Immunol 2005;5:446-458.

42. Deng JC, Cheng G, Newstead MW, Zeng X, Kobayashi K, Flavell RA, Standiford TJ. Sepsis-induced suppression of lung innate immunity is mediated by IRAK-M. J Clin Invest 2006;116:2532-2542. 43. Brint EK, Xu D, Liu H, Dunne A, McKenzie AN, O'Neill LA, Liew FY. ST2 is an inhibitor of interleukin 1

receptor and Toll-like receptor 4 signaling and maintains endotoxin tolerance. Nat Immunol 2004; 5:373-379.

44. Xu D, Chan WL, Leung BP, Huang F, Wheeler R, Piedrafita D, Robinson JH, Liew FY. Selective expression of a stable cell surface molecule on type 2 but not type 1 helper T cells. J Exp Med 1998; 187:787-794.

45. Moritz DR, Rodewald HR, Gheyselinck J, Klemenz R. The IL-1 receptor-related T1 antigen is expressed on immature and mature mast cells and on fetal blood mast cell progenitors. J Immunol 1998;161:4866-4874.

46. Cherry WB, Yoon J, Bartemes KR, Iijima K, Kita H. A novel IL-1 family cytokine, IL-33, potently activates human eosinophils. J Allergy Clin Immunol 2008;121:1484-1490.

47. Oshikawa K, Yanagisawa K, Tominaga S, Sugiyama Y. ST2 protein induced by inflammatory stimuli can modulate acute lung inflammation. Biochem Biophys Res Commun 2002;299:18-24.

(11)

16

Chapter 1

48. Lohning M, Grogan JL, Coyle AJ, Yazdanbakhsh M, Meisel C, Gutierrez-Ramos JC, Radbruch A, Kamradt T. T1/ST2 expression is enhanced on CD4+ T cells from schistosome egg-induced granulomas: analysis of Th cell cytokine coexpression ex vivo. J Immunol 1999;162:3882-3889.

49. Lohning M, Stroehmann A, Coyle AJ, Grogan JL, Lin S, Gutierrez-Ramos JC, Levinson D, Radbruch A, Kamradt T. T1/ST2 is preferentially expressed on murine Th2 cells, independent of interleukin 4, interleukin 5, and interleukin 10, and important for Th2 effector function. Proc Natl Acad Sci U S A 1998;95:6930-6935.

50. Coyle AJ, Lloyd C, Tian J, Nguyen T, Erikkson C, Wang L, Ottoson P, Persson P, Delaney T, Lehar S, Lin S, Poisson L, Meisel C, Kamradt T, Bjerke T, Levinson D, Gutierrez-Ramos JC. Crucial role of the interleukin 1 receptor family member T1/ST2 in T helper cell type 2-mediated lung mucosal immune responses. J Exp Med 1999;190:895-902.

51. Townsend MJ, Fallon PG, Matthews DJ, Jolin HE, McKenzie AN. T1/ST2-deficient mice demonstrate the importance of T1/ST2 in developing primary T helper cell type 2 responses. J Exp Med 2000;191:1069-1076.

52. Hayakawa H, Hayakawa M, Kume A, Tominaga S. Soluble ST2 blocks interleukin-33 signaling in allergic airway inflammation. J Biol Chem 2007;282:26369-26380.

53. Sanada S, Hakuno D, Higgins LJ, Schreiter ER, McKenzie AN, Lee RT. IL-33 and ST2 comprise a critical biomechanically induced and cardioprotective signaling system. J Clin Invest 2007;117:1538-1549. 54. Akira S, Takeda K. Toll-like receptor signalling. Nat Rev Immunol 2004;4:499-511.

55. Kobayashi K, Hernandez LD, Galan JE, Janeway CA, Jr., Medzhitov R, Flavell RA. IRAK-M is a negative regulator of Toll-like receptor signaling. Cell 2002;110:191-202.

Referenties

GERELATEERDE DOCUMENTEN

Middelen worden gebundeld en goedkopere con- tractenn worden afgesloten, maar de effectiviteit van scholen is niet toegeno- men,, aangezien autonomievergroting nauwelijks tot

Een aantal andere mogelijkheden die het FBS biedt enn die redeÜjk veel worden gebruikt zijn het voeren van een eigen taakverde- lingsbeleidd (30%), het voeren van een

De termm 'losse koppeling' wordt ook gebruikt om de professionele bureaucratie te typeren,, waar het onderwijskundige en beheersmatige terrein (twee subsyste- menn die

Binnen het onderwijs kunnen diverse functione- lee doelen worden onderscheiden, zoals de innovatiefunctie, de opvangfunctie, dee werkgelegenheidsfunctie en

Uitt het nonrespons onderzoek blijkt dat de resultaten van het survey die te makenn hebben met de beleidsbenutting voor wat betreft de taak- en functieverbreding,, het reserveren

Dit biedt uiteinde- lijkk de mogelijkheid om scholen te onderscheiden die redelijk tot veel beleids- ruimtee benutten (categorie 3 en 4) en scholen die in enige mate tot weinig

Veran- deringenn die zich voordoen in de organisatiestructuur zijn, naast een verande- ringring in de besluitvormingsverdeling voor strategische besluiten (meer geza-

Doordat de schoolleider van schooll F in het bovenschools management zit en dus minder tijd heeft voor dee eigen school worden binnen school F meer schooltaken gedelegeerd naar