• No results found

Mast Cell Progenitor Trafficking in Allergic Airway Inflammation

N/A
N/A
Protected

Academic year: 2022

Share "Mast Cell Progenitor Trafficking in Allergic Airway Inflammation"

Copied!
42
0
0

Loading.... (view fulltext now)

Full text

(1)

Digital Comprehensive Summaries of Uppsala Dissertations from the Faculty of Medicine 932

Mast Cell Progenitor Trafficking in Allergic Airway Inflammation

JOAKIM DAHLIN

(2)

Dissertation presented at Uppsala University to be publicly examined in C8:301, BMC, Husargatan 3, Uppsala, Thursday, October 17, 2013 at 09:15 for the degree of Doctor of Philosophy (Faculty of Medicine). The examination will be conducted in English.

Abstract

Dahlin, J. 2013. Mast Cell Progenitor Trafficking in Allergic Airway Inflammation. Acta Universitatis Upsaliensis. Digital Comprehensive Summaries of Uppsala Dissertations from the Faculty of Medicine 932. 41 pp. Uppsala. ISBN 978-91-554-8741-6.

Mast cell progenitors originate from the bone marrow and migrate to the lungs via the blood. During maturation, these cells acquire granules that contain a potent array of bronchoconstrictive mediators. The number of pulmonary mast cells is augmented in asthmatic patients and in mice with allergic airway inflammation, possibly contributing to airway hyperreactivity. An increase in mast cells is likely due to an increased recruitment of committed mast cell progenitors from the blood. However, until now a committed mast cell progenitor population has not been found in adult peripheral blood. We isolated Lin

-

c-kit

hi

ST2

+

integrin β7

hi

CD16/32

hi

progenitors from murine blood and showed that these cells were committed to the mast cell lineage. Based on the expression of FcεRI, these cells were less mature in Th1- prone C57BL/6 mice than in Th2-prone BALB/c mice.

Asthma is associated with elevated levels of IgE. Upon exposure to allergens, IgE immune complexes are formed. In a mouse model of allergic airway inflammation, we showed that intranasal administration of IgE immune complexes to antigen-sensitized mice resulted in an increased number of mast cell progenitors compared with antigen administration alone. The increase in mast cell progenitors was independent of the low-affinity IgE receptor CD23. Rather, signaling through the common FcRγ-chain was required to enhance the number of lung mast cell progenitors. Signaling through FcεRI was likely responsible for the increase. However a role for FcγRIV could not be excluded.

CD11c

+

cells, such as dendritic cells, are important for antigen sensitization. In a mouse model of allergic airway inflammation, these cells are also important for the development of airway hyperreactivity, eosinophilia and Th2 cytokine production in response to antigen challenge.

We showed that CD11c

+

cells are critical for the recruitment of lung mast cell progenitors and the subsequent increase in mast cells. These CD11c

+

cells were needed for the upregulation of endothelial vascular cell adhesion molecule-1 (VCAM-1), which is a prerequisite for the antigen-induced recruitment of lung mast cell progenitors.

Keywords: Mast cells, mast cell progenitors, allergy, asthma, allergic airway inflammation, IgE, lung, CD11c

Joakim Dahlin, Uppsala University, Department of Medical Biochemistry and Microbiology, Box 582, SE-751 23 Uppsala, Sweden.

© Joakim Dahlin 2013 ISSN 1651-6206 ISBN 978-91-554-8741-6

urn:nbn:se:uu:diva-206608 (http://urn.kb.se/resolve?urn=urn:nbn:se:uu:diva-206608)

(3)

When you do things right, people won’t be sure you have done

anything at all

Futurama

(4)
(5)

List of papers

This thesis is based on the following papers, which are referred to in the text by their Roman numerals:

I Dahlin J.S., M.A. Ivarsson, B. Heyman, and J. Hallgren. 2011. IgE Immune Complexes Stimulate an Increase in Lung Mast Cell Pro- genitors in a Mouse Model of Allergic Airway Inflammation. PLoS One 6:e20261.

II Dahlin J.S., R. Feinstein, Y. Cui, B. Heyman, and J. Hallgren. 2012.

CD11c

+

cells are required for antigen-induced increase of mast cells in the lung. The Journal of Immunology 189: 3869-3877.

III Dahlin J.S., B. Heyman, and J. Hallgren. 2013. Committed mast cell progenitors in mouse blood differ in maturity between Th1 and Th2 strains. Allergy. In press.

Reprints were made with permission from the respective publishers.

(6)
(7)

Contents

Introduction ... 9

Background ... 10

The origin of mast cells ... 10

Mast cells in allergic airway inflammation ... 12

Dendritic cells and alveolar macrophages in allergic airway inflammation ... 12

IgE and IgE-binding receptors ... 14

Migration of mast cell progenitors to the lung ... 16

Lung mast cell progenitors ... 16

Integrins, adhesion molecules and chemotaxis ... 17

Cytokines ... 18

IgE antibodies ... 19

Strain-dependent differences ... 19

Present investigation – rationale and aims ... 20

Paper I ... 20

Paper II ... 20

Paper III ... 20

Experimental setup ... 21

Experimental protocols ... 21

Quantification of mast cell progenitors with a limiting dilution assay 21 Results and discussion ... 23

IgE immune complexes stimulate an increase in lung mast cell progenitors in a mouse model of allergic airway inflammation (paper I) ... 23

CD11c

+

cells are required for antigen-induced increase of mast cells in the lung (paper II) ... 24

Committed mast cell progenitors in mouse blood differ in maturity between Th1 and Th2 strains (paper III) ... 26

Concluding remarks and future perspectives ... 29

(8)

Abbreviations

C/EBPα CCAAT/enhancer-binding protein α DTR Diphtheria toxin receptor

MAdCAM-1 Mucosal vascular addressin cell adhesion molecule-1

MCp Mast cell progenitor(s)

MITF Microphthalmia-associated transcription factor

MNC Mononuclear cell(s)

OVA Ovalbumin

VCAM-1 Vascular cell adhesion molecule-1

(9)

Introduction

Mast cells are immune cells that are derived from the bone marrow. These cells enter the blood as immature mast cell progenitors (MCp). Upon reach- ing the peripheral tissues, the cells are allowed to mature and develop their characteristic granules.

Mast cells are infamous for their role in allergic asthma. In allergic asth- matics, mast cells are preloaded with allergen-specific IgE. Allergen expo- sure consequently leads to the formation of IgE immune complexes that crosslink FcεRI on the mast cells’ surface. This crosslinking leads to the release of granule-associated mediators, several of which have potent bron- choconstrictive effects

1

. The increased number of pulmonary mast cells in asthmatics

2,3

likely worsens the symptoms.

Experiments using mouse models of allergic airway inflammation suggest that the elevated number of lung mast cells is due to the recruitment of pro- genitors committed to the mast cell lineage

4

. However, committed MCp have not been found in adult mouse blood in mice or humans. Furthermore, the cellular and molecular mechanisms that govern the increase in lung mast cells are still unclear.

This thesis focuses on the role of IgE immune complexes and CD11c

+

cells in the increase in lung MCp numbers. These topics will be covered in

papers I and II, respectively. In paper III, we identified, isolated and charac-

terized a committed MCp population in murine peripheral blood.

(10)

Background

The origin of mast cells

Mast cells are hematopoietic cells. The most immature hematopoietic cells are stem cells, which have self-renewing potential and are therefore capable of reconstituting all myeloid and lymphoid cells in a transplanted host. Hem- atopoietic stem cells develop into multipotent progenitors that have lost the self-renewing capacity before committing to either the common myeloid progenitor or common lymphoid progenitor lineage

5-7

. The ontogeny of mast cells has mainly been studied in mice, in which the cells belong to the mye- loid lineage, as they can be derived from Sca-1

lo

common myeloid progeni- tors

7

. Further characterization of mast cells’ relationship with other cells is ambiguous as single progenitors that exclusively give rise to 1) mast cells and basophils

8

; 2) mast cells, megakaryocytes and erythrocytes

9

; 3) mast cells and macrophages

9

; and 4) mast cells, macrophages and neutrophils

9

have been found. Therefore, whether mast cells belong to the megakaryo- cyte/erythroid lineage, the granulocyte/monocyte lineage or their own line- age is unclear.

Progenitors that only have the potential to become mast cells, i.e., com- mitted MCp, were first described in fetal mouse blood. These progenitors were identified as Thy-1

lo

c-kit

hi

cells that declined in frequency from gesta- tion day 15.5 until birth

10

. In adult mice, committed MCp have been de- scribed in the bone marrow. In C57BL/6 mice, the cells are identified as Lin

-

c-kit

+

Sca-1

-

Ly6c

-

FcεRI

-

CD27

-

integrin β7

+

ST2

+

cells

11

. Two monoclonal antibodies, BGD6 and AA4, raised against basophilic leukemia cells (RBL- 2H3) have been used to identify committed MCp in the bone marrow of BALB/c mice

12

. In healthy tissues, BGD6 recognizes a 110 kDa mast cell- specific protein using the Fab fragment of the antibody, whereas the Fc part binds CD32

13

. AA4 binds ganglioside GD1b

14

, which is present on mature mast cells but not on their progenitors

12

. In BALB/c bone marrow, progeni- tors committed to the mast cell lineage can therefore be identified as BGD6

+

AA4

-

cells

12

. These cells are further characterized by expression of CD34 and CD13 but a lack of FcεRI.

As mast cells are derived from the bone marrow

15

and given that commit-

ted MCp are found in the bone marrow, it is be reasonable to assume that

committed MCp can be found in the spleen. However, such a progenitor has

(11)

yet to be found

8

. Instead, Lin

-

c-kit

+

integrin β7

hi

CD16/32

hi

bipotent baso- phil/mast cell progenitors have been described

8

.

Although committed MCp in adult blood have not been found in adult blood

8,12

, these cells need to pass through the circulation before reaching their target organ. In the peripheral tissue, such as the intestinal compart- ment, committed MCp are Lin

-

CD45

+

CD34

+

integrin β7

hi

FcεRI

lo

cells

8

. After reaching their target organ, the cells mature into c-kit

+

FcεRI

+

granu- lated cells. The lineage development of mast cells is summarized in Figure 1.

Figure 1. Lineage development of mast cells in adults. Mast cells are derived from hematopoietic stem cells (HSC) in the bone marrow. These cells develop into com- mitted MCp via multipotent progenitors (MPPs) and Sca-1

lo

common myeloid pro- genitors (SL-CMPs). Committed MCp have been described as being present in the bone marrow of both C57BL/6 and BALB/c mice. Bipotent basophil/mast cell pro- genitors (BMCPs) have been described as being present in the spleen of C57BL/6 mice. The MCp likely enter the blood circulation and migrate into the peripheral organs. However, committed MCp have not been found in the blood.

A population of committed MCp has not been isolated in humans. In cord blood, CD34

+

CD38

+

HLA-DR

-

cells have mast cell-forming potential

16

. However, the same population also gives rise to macrophages, neutrophils and eosinophils. In the peripheral blood, MCp can be found in the CD34

+

c- kit

+

CD13

+

cell population

17

. Nevertheless, this population also gives rise to monocytes.

Bone marrow

Spleen

Blood

Peripheral organs

MPP

HSC SL-CMP

BMCP

C57BL/6 Lin

-

Sca-1

-

Ly6c

-

CD27

-

integrin β7

+

ST2

+

c-kit

+

FcεRI

-

BALB/c

AA4

-

BGD6

+

(c-kit

+

FcεRI

-

)

C57BL/6

Lin

-

c-kit

+

integrin β7

hi

CD16/32

hi

(FcεRI

-

)

C57BL/6

Lin

-

CD45

+

CD34

+

integrin β7

hi

FcεRI

lo

Less mature More mature

MCp

?

MCp

(12)

Mast cells in allergic airway inflammation

Upon allergen inhalation challenge in allergic asthmatics, pulmonary mast cells are activated, leading to the release of bronchoconstrictive mediators

18-

21

. The increased number of mast cells in the asthmatic lung

2,3,22

likely ag- gravates the asthmatic symptoms. Increased numbers of mast cells in the lung are also observed in mouse models of allergic airway inflammation

23,24

.

The role of mast cells in allergic airway inflammation has been evaluated in mice. In these models, mice are typically sensitized to ovalbumin (OVA) intraperitoneally and challenged with the same antigen either intranasally or in aerosolized form. Commonly, the inflammatory response in the lungs and airway function are compared between mast cell-deficient and wild-type mice. Traditionally, mast cell-deficient mice with defects in the stem cell factor-kit signaling pathway (W

sh

and W/W

v

mice) have been used. In cer- tain models, the mast cell-deficient mice develop less severe airway reactivi- ty and exhibit less infiltration of inflammatory cells

25-28

. This reduced airway reactivity has been confirmed in transgenic mice transiently devoid of mast cells during the antigen challenge phase

29

. However, in other models, the lack of mast cells does not affect airway reactivity or inflammation

30-32

. The discrepancy between mast cell-dependent and mast cell-independent models seems to be related to the sensitization. It appears that the addition of adju- vant renders a model mast cell independent

33

. However, the reconstitution of mast cells, even in mast cell-independent models, leads to increased airway reactivity

34

, suggesting that mast cells have a potent function in allergic air- way inflammation.

The genetic background of the mice also determines the role of mast cells in inducing airway hyperresponsiveness. For example, W

sh

mice on a C57BL/6 background have reduced airway reactivity compared with wild- type mice

35

. However, using the same protocol, W

sh

mice on a BALB/c background have similar airway hyperresponsiveness compared with wild- type BALB/c mice

35

.

As OVA is rarely implicated in human asthma, mouse models using the natural allergen house dust mite have been developed. In these models, mast cells also have an important role

36

. Recently, new c-kit-independent mast cell-deficient mouse strains have been developed

37-39

. Studies investigating the role of mast cells in allergic airway inflammation in these mouse strains are awaited.

Dendritic cells and alveolar macrophages in allergic airway inflammation

Dendritic cells are important for the priming of naïve T cells. In mouse mod-

els of allergic airway inflammation, dendritic cells are therefore capable of

(13)

sensitizing the host against an allergen

40

. However, these cells are not only important during the antigen sensitization phase. During the antigen chal- lenge phase, dendritic cells are required for the induction of airway eosino- philia, Th2 cytokine production, goblet cell hyperplasia and bronchial hyper- responsiveness

41

.

Dendritic cells in the lung can be divided into conventional, monocyte- derived and plasmacytoid subsets, each with a specific function and localiza- tion in the lung. Conventional dendritic cells can be further subdivided into CD103

+

intraepithelial dendritic cells and CD11b

+

cells, which are present in the lamina propria

42

. Upon contact with antigen, conventional CD11b

+

den- dritic cells migrate to the draining mediastinal lymph nodes and present the antigen to T cells

43

. The inflammatory response induced by the conventional dendritic cells can be counteracted by plasmacytoid dendritic cells, which induce tolerance

44

. In parallel, monocyte-derived (inflammatory) dendritic cells are recruited to the lung via a CCR2-dependent mechanism

43

. These cells produce several chemokines that act on monocytes and eosinophils

43

.

Consistent among the dendritic cell subsets in the lung is the expression of CD11c. Although CD11c is generally considered to be a dendritic cell marker, CD11c is also expressed by alveolar macrophages. Alveolar macro- phages are excellent at endocytosing antigen but are poor inducers of T cell proliferation

43

. These macrophages have been reported to have both protec- tive

45

and pro-inflammatory

46

roles in mouse models of allergic airway in- flammation. The different CD11c

+

cell populations are summarized in Figure 2.

The understanding of CD11c

+

cells’ role in allergic airway inflammation has been greatly enhanced by the generation of CD11c-diphtheria toxin re- ceptor (DTR) mice

41,47

. Cells from the wild-type mice do not normally bind to diphtheria toxin, as these cells express a mutated form of the human diph- theria toxin-binding receptor

48

. In CD11c-DTR mice, a transgenic human diphtheria toxin receptor is expressed under the promoter of cd11c. Admin- istration of diphtheria toxin to CD11c-DTR mice therefore leads to a transi- ent depletion of CD11c

+

cells

49

.

CD11c expression is not completely restricted to dendritic cells and alve-

olar macrophages. For example, plasma cells and NK cells express low lev-

els of CD11c

50,51

. However, expression of CD11c does not automatically

imply that the transgene is expressed. For example, plasmacytoid dendritic

cells, which express low levels of CD11c, lack expression of the transgene

52

.

Consequently, plasmacytoid dendritic cells are insensitive to diphtheria tox-

in.

(14)

Figure 2. CD11c

+

cell populations in the murine lung.

IgE and IgE-binding receptors

In humans, high IgE levels are correlated with an increased risk of asthma

53

. Recent success in treating allergic asthmatics with humanized anti-IgE (omalizumab) further supports IgE’s role in the disease

54

. IgE binds to the high-affinity IgE receptor FcεRI in both humans and mice. In humans, FcεRI is expressed by mast cells, basophils, Langerhans cells, platelets, eosino- phils, monocytes and circulating dendritic cells

55

. In naïve mice, the expres- sion pattern is limited to mast cells and basophils. These cells express FcεRI consisting of a complex of four subunits, known as αβγ

2

. House dust mite provocation and Sendai virus infection of mice can induce FcεRI expression on lung dendritic cells, which is likely limited to monocyte-derived dendritic cells

43,56

. The neutrophils of mice infected with cerebral malaria can also express FcεRI

57

. Murine dendritic cells and neutrophils express the trimeric form, αγ

2

, of FcεRI

56,57

.

Without antigen crosslinking, monomeric IgE increases the survival of mature mast cells via FcεRI

58,59

. Crosslinking of FcεRI leads to the degranu- lation and activation of both mast cells and basophils. FcεRI is therefore potent for the development of systemic anaphylaxis

60

and allergic airway inflammation

61

.

The low-affinity IgE receptor CD23 (FcεRII) is a C-type lectin found in two isoforms. In mice, CD23a is constitutively expressed by B cells

62

and

Alveolar macrophages CD103

+

intraepithelial

dendritic cells CD11b

+

dendritic cells in lamina propria

Monocyte-derived inflammatory dendritic cells

Plasmacytoid dendritic cells

Epithelium Lumen

(15)

follicular dendritic cells

63

, and CD23b is found on intestinal epithelial cells

64

and requires IL-4 for upregulation

65,66

. In humans, CD23a is expressed by B cells, and CD23b is found on several cell types

67

.

CD23 is important for the feedback regulation of the immune response in mice

68

. This phenomenon has been demonstrated by intravenous administra- tion of IgE immune complexes compared with antigen administration alone.

The injected IgE immune complexes are transported to the spleen by CD23- expressing B cells

69

, leading to greater T-cell proliferation and an enhanced antibody response

68

.

In mice, other low-affinity IgE receptors include FcγRIIB, FcγRIII and FcγRIV. Mice deficient in FcγRIIB have enhanced IgE-mediated anaphylax- is, whereas mice lacking FcγRIII show a reduced response

70

. FcγRIV on alveolar macrophages has a minor role in the development of lung inflamma- tion. When mast cells release their mediators into the lung, IgE immune complexes bind to FcγRIV on alveolar macrophages, contributing to the influx of Mac-1

+

Gr-1

+

polymorphonuclear cells

71

.

The binding of IgE immune complexes to the low-affinity receptors FcγRIIB and FcγRIII appears to be dependent on the allotype of the IgE antibodies. An allotype represents the phenomenon of antibodies differing in the constant region between individuals in a species. The criteria for an allo- type also include that the difference needs to be detectable by an antibody.

IgE of allotype b in an immune complex with antigen has no or low affinity for FcγRIIB and FcγRIII, whereas IgE of allotype a binds to these receptors

71

.

Of the IgE-bindning receptors discussed, FcγRIII, FcγRIV and FcεRI re-

quire the common FcRγ-chain for receptor assembly and signaling (Figure

3)

72

. Mice deficient in the FcRγ-chain can therefore be used to evaluate a

potential role for these receptors.

(16)

Figure 3. Schematic view of murine IgE-binding Fc receptors. Note that FcγRIII, FcγRIV and FcεRI rely on the common FcRγ-chain for receptor assembly and sig- naling.

Migration of mast cell progenitors to the lung

Lung mast cell progenitors

MCp contain no or few metachromatic granules (Figure 4) and lack or have low expression of FcεRI. Due to the lack of granules, MCp cannot be identi- fied by histochemical staining techniques. The quantification of tissue MCp therefore often relies on indirect quantification using a limiting dilution as- say (see description in experimental setup).

In naïve mouse lungs, MCp are present on the order of 20–200 cells per mouse

23,73,74

due to constitutive migration of MCp into the lung tissue, re- ferred to as homing. The homing of MCp to the lung is independent of an adaptive immune system

75

. In contrast, MCp numbers in the lung increase over baseline levels in allergic airway inflammation. This migration into the lung tissue is a CD4

+

T cell-mediated process and is called recruitment

73

.

Figure 4. May-Grünwald Giemsa staining of an MCp (left) and a mature mast cell (right). The MCp lack metachromatic granules, whereas the mature mast cells have many.

α β γ γ

α γ γ

α α β γ γ

FcγRIIB FcγRIII FcγRIV FcεRI CD23

MCp Mature mast cell

(17)

If not otherwise mentioned, the protocols for studying MCp recruitment described below share a common structure with a sensitization phase and a challenge phase. During the sensitization, OVA adsorbed to alum is adminis- tered intraperitoneally. Mice are subsequently challenged with OVA in aero- solized form. This protocol promotes a large increase in the frequency and total number of lung MCp. Three days after the first challenge, the total number of MCp increases 28-fold

23

. The short time frame makes it unlikely that the effect is only due to the increased proliferation or survival of the MCp. Rather, MCp are recruited into the tissue. The recruitment of lung MCp is followed by the appearance of intraepithelial mucosal mast cells in the trachea and large bronchi

24,76

.

Many of mast cells’ functions are attributed to the release of their granule contents. MCp, which lack filled granules, are therefore often only regarded as a reservoir of mast cells. This view has recently been challenged, as MCp can produce cytokines such as IL-4 and IL-6 in vivo

77

. MCp may therefore have effector functions by themselves.

In most models of allergic airway inflammation, Th2-prone BALB/c mice have been used to delineate the mechanisms that govern the recruitment of MCp to the lung. The recruitment is controlled differently in C57BL/6 mice in certain instances, which will be described in detail later.

Integrins, adhesion molecules and chemotaxis

For MCp to be recruited into the lung tissue, the lung endothelium needs to express vascular cell adhesion molecule-1 (VCAM-1), but not mucosal vas- cular addressin cell adhesion molecule-1 (MAdCAM-1)

23

. VCAM-1 can interact with integrins α4β1 and α4β7 on the MCp surface and allows entry into the tissue

23

. The β7 integrin subunit can also associate with αE. Howev- er, blocking αE does not affect MCp recruitment into the lung.

The recruitment of MCp to the lung is also regulated by chemokine recep-

tors and their ligands. In the literature, there are 19 chemokine receptors

described: CXCR1–7, CCR1–10, CX3CR1 and XCR1. So far, the involve-

ment of CXCR2, CCR2, CCR3 and CCR5 in MCp recruitment to the lung

has been evaluated. CXCR2, CCR3 and CCR5 are expressed by immature

human mast cells cultured from cord blood

78

. Immature mouse mast cells

derived from the bone marrow express CCR2 and migrate toward CCL2 in

vitro

74,79

. One would anticipate finding chemokine receptors on MCp that are

required for recruitment into the lung. However, the situation is far more

complex.

(18)

lium. This feature is a prerequisite for the antigen-induced upregulation of VCAM-1

24

. CCR3 and CCR5 are dispensable for MCp recruitment

24

. A lack of CCR3 is instead associated with an increased number of intraepithelial mast cells in the trachea

80

.

Studies on humans provide evidence that lung mast cells express CXCR3

81

. In allergic asthma, pulmonary mast cells also express CCR1 and CCR4

82

. Whether the expression of these receptors controls MCp recruit- ment, localization within the lung or both remains to be investigated.

Prostaglandin E

2

, which is produced in the lung tissue upon sensitization and challenge with OVA, can attract in vitro-cultured MCp

83

. This effect is mediated by prostaglandin E

2

receptor 3 on the MCp. Similarly, leukotriene B

4

induces chemotaxis via BLT1

84

. However, the in vivo relevance of pros- taglandin E

2

and leukotriene B

4

to MCp migration needs to be verified. For example, it has not been established whether MCp express prostaglandin E

2

receptor 3 and BLT1 in vivo.

Figure 5. CXCR2 regulate VCAM-1 expression on the lung endothelium. Integrins α4β1 and α4β7 on MCp adhere to VCAM-1. The MCp are then allowed to pass through the tissue endothelium. Type 2 NKT cells produce IL-9, stimulating prolif- eration of the MCp before they mature. CCL2 interaction with CCR2 is also needed for the increase in lung MCp.

Cytokines

Th2 cytokines, such as IL-13, are linked to asthma in patients

85

. In mouse models of allergic asthma, Th2 cytokines promote IgE production, airway hyperresponsiveness and eosinophilia

86

. However, for MCp recruitment, several classical Th2 cytokines are dispensable, including IL-4, IL-5, IL-10 and IL-13

73

. Additionally, many Th1 and Th17 cytokines, such as IL-3, IL-6, IL-12p40, IL-17A and IFNγ, are redundant

73

. So far, the only cytokine that has been found to be critical for the increase in lung MCp is IL-9

73

. As mast cells can produce IL-9, mice lacking mature mast cells but not MCp (i.e., W/W

v

, W

sh

and Kitl

Sl

/Kitl

Sl-d

) have been studied. All of these strains have a

Blood

Mast cell progenitor

Lung

Mature mast cell

VCAM-1

Endothelium

CXCR2

Integrin α4β1 Integrin α4β7

CCR2

CCL2 Type 2

NKT cell IL-9

(19)

normal recruitment of MCp to the lung compared with wild-type mice. This finding suggests that mature mast cells are not involved in MCp recruitment to the lung and therefore are not the source of IL-9. The result also suggests that MCp are not recruited to the lung by chemotactic stem cell factor. The IL-9 is most likely produced by type 2 NKT cells

73

.

In humans, the IL-9 receptor is expressed by lung mast cells

87

and blood MCp

88

. The binding of IL-9 enhances the in vitro proliferation of the MCp

88

. The requirement for IL-9 for the increase in lung MCp is therefore most likely due to the cytokine’s ability to induce proliferation. It is likely that the increase in lung MCp relies partly on integrin-adhesion molecule interaction and partly on proliferation induction by IL-9. This hypothesis is strengthened by the fact that a lack of IL-9 does not completely abolish the increase in MCp in sensitized and challenged animals

73

. Furthermore, VCAM-1 upregu- lation is intact when IL-9 is blocked

73

. Current knowledge about the recruit- ment of lung MCp is summarized in Figure 5.

IgE antibodies

In a model of allergic airway inflammation in which Aspergillus fumigatus was used as the allergen, wild-type and IgE-deficient mice had an equally strong recruitment of MCp to the lung

59

. Nonetheless, the number of mature mast cells was severely reduced in the IgE-deficient mice. This finding sug- gests that IgE induces the survival of mature mast cells but not MCp.

The role of IgE in MCp recruitment has also been indirectly evaluated in an OVA-model in which mice are sensitized intraperitoneally and challenged with aerosolized antigen. Jones et al showed that STAT6-knockout mice, which virtually lack IgE

89

, have normal recruitment of MCp to the lung

73

. In conclusion, IgE is not necessary for the increase in lung MCp.

Strain-dependent differences

Although the requirement for integrins α4β1 and α4β 7 is conserved in the

OVA-induced recruitment of MCp to the lungs in C57BL/6 mice, other

mechanisms differ. In C57BL/6 mice, the production of IL-9 and the pres-

ence of NKT cells are dispensable. Instead, CD25

+

regulatory T cells are

critical for OVA-induced MCp recruitment

90

. Dependency on IL-10R and

TGFβ further strengthens the regulatory T cell pathway’s role in MCp re-

cruitment in C57BL/6 mice

90

.

(20)

Present investigation – rationale and aims

Paper I

Patients with allergic asthma have high levels of allergen-specific IgE and an increased number of mast cells in their airways. Upon allergen exposure, IgE immune complexes are formed in the lung. By using a mouse model of al- lergic airway inflammation, this paper aimed to investigate whether IgE immune complexes in the lung can induce an increase in the number of lung MCp.

Paper II

CD11c

+

cells are needed for airway hyperresponsiveness, lung eosinophilia and the induction of Th2 cytokines in mouse models of allergic airway in- flammation. Here, we investigated the role of CD11c

+

cells in the antigen- induced recruitment of lung MCp and the increase in mature mast cells.

Paper III

In mouse models of allergic airway inflammation, MCp migrate from the

blood to the lung, which causes an increase in pulmonary mast cells. Alt-

hough committed MCp have been postulated to be present in adult peripheral

blood, such cells have not been found. In this study, we identified, isolated

and characterized these cells.

(21)

Experimental setup

Experimental protocols

Mice were sensitized intraperitoneally with OVA adsorbed to alum on days 0 and 7. The injections were followed by antigen challenges on days 17, 18 and 19. In paper I, the mice were challenged intranasally with OVA- trinitrophenyl alone or in an immune complex with IgE. In paper II, the mice were challenged with aerosolized OVA. One day after the last challenge, the mice were euthanized and their lungs were removed and analyzed. In paper II, an analysis of mature mast cells was performed eight days after the last challenge.

Quantification of mast cell progenitors with a limiting dilution assay

Immune cells can often be differentiated based on cell size, the shape of the nucleus and granule contents. This differentiation makes it possible to esti- mate the number of cells of a certain type in an organ using histochemical staining techniques. For example, alveolar macrophages, lymphocytes, neu- trophils, eosinophils, mature mast cells and red blood cells are easily identi- fied in the lung. To further scrutinize different cell subsets, flow cytometry can be applied. However, for this method to be feasible the subset needs to be defined, usually by markers recognized by antibodies. MCp cannot be recognized by histochemical staining techniques, and the quantification of MCp by flow cytometric analysis has not been completely validated in lung tissue. Therefore, an indirect approach based on limiting dilution was used to quantify MCp.

Mononuclear cells (MNC) purified from the lung were plated into 96-well

plates at an initial concentration of 20,000 cells/well with 24 replicates. Sev-

en two-fold serial dilutions of the MNC were also plated with the same

number of replicates. After 10–12 days in culture with IL-3 and SCF, colo-

nies formed in the wells that contained at least one MCp from the beginning.

(22)

!(! = !) = ! ! ! !! /!!

where µ is the mean number of mast cell colonies and ! ∈ ℕ. The frequency of MCp per MNC, ϕ, can be estimated as

! = !/!

where N is the number of MNC in one well. The probability of a well with non-appearing colonies is then

!(! = 0) = ! !!" .

If p is the observed frequency of non-appearing colonies (i.e., an estimate of

!(! = 0)), then

! = −1/! ∙ ln !.

Linear regression can be performed to describe the association between ln ! and every value of N according to the equation

ln ! = −!" + !

The frequency of MCp per MNC can be approximated either based on the slope, ϕ, or as described

91

, by determining the value at which ! = 1/! ≈ 0.37 (in other words when !" = 1) (Figure 6). The total number of MCp per lung is derived by multiplying the frequency of MCp by the total number of isolated MNC.

Figure 6. The frequency of MCp, as estimated by linear regression.

0 5000 10000 15000 20000

-4 -3 -2 -1 0

MNC

ln p

1 MCp per 5789 MNC

173 MCp/106 MNC

(23)

Results and discussion

IgE immune complexes stimulate an increase in lung mast cell progenitors in a mouse model of allergic airway inflammation (paper I)

Asthmatics have increased levels of IgE that form immune complexes upon allergen exposure. Here, we tested the hypothesis that IgE immune complex- es cause an increase in lung MCp.

IgE immune complexes instilled intranasally provoked a large increase in lung MCp in antigen-sensitized mice compared with antigen administration alone. The increased number of MCp was independent of the low-affinity IgE receptor CD23. However, signaling through the FcRγ chain was critical for the recruitment to occur. This finding suggested that the increase is me- diated by FcεRI, FcγRIII or FcγRIV. As the IgE used was of allotype b, which does not bind FcγRIII, the IgE immune complexes likely acted through FcεRI or FcγRIV. Our data were in agreement with the findings of a previous report showing that IgE immune complexes administered intrana- sally induce pulmonary eosinophilia and neutrophilia, which is an effect mediated by FcεRI

92

.

Whether the increase in MCp was due to the recruitment or expansion of preexisting lung MCp was not investigated. It is improbable that the in- creased number of MCp resulted from the enhanced survival of the MCp, as IgE alone did not induce an increased number of MCp. Moreover, a study by Mathias et al supports the fact that IgE does not induce survival in MCp

59

.

The cell that promoted the FcRγ-chain-mediated increase in MCp was not studied. Obvious candidates include FcεRI-expressing mature mast cells, basophils and monocyte-derived dendritic cells. A combined role for FcεRI- expressing cells and FcγRIV-expressing alveolar macrophages cannot, how- ever, be excluded.

Another question is how IgE immune complexes are transported through

the lung epithelium. In mice, intestinal epithelial cells can mediate this

transportation via their CD23 receptors

65

. However, IgE immune complex-

(24)

Patients with asthma have elevated levels of allergen-specific IgE

53

, which forms IgE immune complexes locally in the lung upon allergen expo- sure. Our study suggests that these IgE immune complexes can stimulate an increase in lung MCp. This phenomenon may lead to an increased number of mature mast cells, which could aggravate the asthmatic symptoms.

CD11c + cells are required for antigen-induced increase of mast cells in the lung (paper II)

In this study, we wanted to investigate the role of CD11c

+

cells in lung mast cell accumulation during allergic airway inflammation. Specifically, the role of CD11c

+

cells during the antigen challenge phase was studied. CD11c

+

cells were depleted one day before the first challenge by diphtheria toxin treatment of CD11c-DTR mice. Six to seven days after the injection of diph- theria toxin, the mice died due to DTR expression on radioresistant stromal cells

94

. By transferring CD11c-DTR bone marrow cells into irradiated wild- type mice, lethal side effects were prevented, while all CD11c

+

hematopoiet- ic cells are depleted. In cases in which analysis was performed more than four days after the first diphtheria toxin treatment, bone marrow-chimeric mice were used.

OVA-sensitized CD11c-DTR mice had very few lung MCp, but the num- ber increased tremendously after OVA challenge. This increase in MCp was followed by the appearance of mature mast cells one week later. Similar to patients with atopic uncontrolled asthma, these mast cells were located in the alveolar parenchyma

3

.

The absence of CD11c

+

cells during the challenge phase led to a ham- pered influx of MCp into the lung. Consequently, no increase in mature mast cells was observed after one additional week. The abolished MCp recruit- ment in CD11c

+

cell-depleted mice could be explained by the impaired up- regulation VCAM-1 on the lung endothelium.

The question of how CD11c

+

cells regulate VCAM-1 is more complex. It

has been shown that IL-4 and IL-13 produced by Th2 cells can induce

VCAM-1. However, it is unlikely that these cytokines control VCAM-1

expression in this model, as IL-4 and IL-13 are dispensable for the recruit-

ment of MCp to the lung

73

. Another potent inducer of VCAM-1 is TNF-α

95

.

The CD11c

+

dendritic cells and alveolar macrophages produced TNF-α,

suggesting that TNF-α could be the inducer of VCAM-1. This possibility is

supported by Boyce et al, who showed that human cord blood derived imma-

ture mast cells adhere to TNF-α-treated human vascular endothelium in

vitro

96

. However, stromal expression of CXCR2 also regulates VCAM-1. It

is therefore possible that the ligands of CXCR2, i.e., CXCL1 and CXCL2,

control the expression of VCAM-1, and that several pathways act redundant-

(25)

ly. A possible mechanism by which CD11c

+

cells regulate the increase in mast cells is summarized in Figure 7.

Figure 7. CD11c

+

cells are required to increase the number of lung mast cells in allergic airway inflammation. CD11c

+

dendritic cells and alveolar macrophages induce increased expression of endothelial VCAM-1 upon antigen challenge, most likely due to production of TNF-α. VCAM-1 interacts with integrins α4β1 and α4β7 on MCp, allowing transmigration from the blood to the lung. Inside the lung tissue, the MCp develop into mature mast cells with granules.

To further evaluate which CD11c

+

subpopulation was responsible for the MCp recruitment, different CD11c

+

cell populations were transferred into CD11c

+

cell-depleted mice. However, neither alveolar macrophages nor bone marrow-derived dendritic cells resulted in reconstitution of the CD11c

+

cells in the lung (data not shown). This approach therefore could not be used to evaluate which CD11c

+

cell type that is involved in OVA-induced MCp trafficking.

To rule out the possibility that MCp are depleted by the diphtheria toxin, several experiments were performed. First, wild-type MCp were not depleted by diphtheria toxin. Second, the number of MCp in the bone marrow was similar in CD11c-DTR and wild-type mice treated with diphtheria toxin.

Third, using flow cytometry, it was shown that neither lung-derived mast cells nor splenic basophil/mast cell progenitors express CD11c. Last, diph- theria toxin treatment of CD11c-DTR mice did not affect the number of mast cells in the tongue. To summarize, mast cells and their progenitors do not express CD11c and are not depleted by diphtheria toxin in vivo.

Collington et al showed that CCR2 expression is dispensable on bone marrow-derived cells during MCp recruitment to the lung

74

. Furthermore,

Blood

TNF-α e.g.

CD11c

+

Dendritic cell

Mast cell progenitor

Lung

Mature mast cell

VCAM-1

CD11c

+

Alveolar macrophage

Endothelium

(26)

conventional dendritic cells or alveolar macrophages are needed for the anti- gen-induced MCp recruitment to occur.

Committed mast cell progenitors in mouse blood differ in maturity between Th1 and Th2 strains (paper III)

A population of Lin

-

c-kit

hi

ST2

+

integrin β7

hi

CD16/32

hi

cells was identified in the blood of BALB/c mice. These cells were further divided into FcεRI

-

and FcεRI

+

subpopulations that were isolated using FACS. Cells from each respective population were cultured in medium containing IL-3, IL-5, IL-6, IL-7, IL-9, IL-11, stem cell factor, thrombopoietin, erythropoietin and granu- locyte-macrophage colony-stimulating factor. This treatment allowed differ- entiation into megakaryocytes, erythrocytes, monocytes, eosinophils, neu- trophils, basophils and mast cells

8

. After 4 days in culture, both the FcεRI

-

and the FcεRI

+

fractions of the Lin

-

c-kit

hi

ST2

+

integrin β7

hi

CD16/32

hi

pop- ulation gave rise to c-kit

+

FcεRI

+

immature mast cells. After 14 days, all Lin

-

c-kit

hi

ST2

+

integrin β7

hi

CD16/32

hi

cells exclusively differentiated into c-kit

+

FcεRI

+

mast cells with metachromatic granules.

The mast cell-forming capacity in other populations was limited. For ex- ample, after culturing Lin

-

c-kit

hi

ST2

+

integrin β7

-/lo/int

CD16/32

-/lo/int

for 4 days, the frequency of mast cells was less than 3 %. Instead, the sorted cells mainly turned into c-kit

-

FcεRI

-

cells belonging to the megakaryo- cytic/erythroid lineage (unpublished data).

Flow cytometric analysis of blood from C57BL/6 mice confirmed that Lin

-

c-kit

hi

ST2

+

integrin β7

hi

CD16/32

hi

cells were also present in this strain.

However, a lower proportion of these cells expressed FcεRI than did cells from BALB/c mice, suggesting a less mature phenotype in C57BL/6 mice.

Still, the Lin

-

c-kit

hi

ST2

+

integrin β7

hi

CD16/32

hi

cells from C57BL/6 mice

mainly differentiated into c-kit

+

FcεRI

+

mast cells. An updated view of mast

cell lineage development is shown in Figure 8.

(27)

Figure 8. Bone marrow hematopoietic stem cells (HSCs) develop into Sca-1

lo

com- mon myeloid progenitors (SL-CMPs) via a multipotent progenitor (MPP). Commit- ted MCp are found in the bone marrow in both C57BL/6 and BALB/c mice. Bipo- tent basophil/mast cell progenitors (BMCPs) are found in C57BL/6 mice in the spleen. Committed MCp are found in the blood of both BALB/c and C57BL/6 mice.

These cells migrate into the peripheral organs.

In C57BL/6 mice but not in BALB/c mice, culturing of Lin

-

c-kit

hi

ST2

+

integrin β7

hi

CD16/32

hi

cells for 4 days led to the appearance of less than 7

% c-kit

-

FcεRI

+

cells, beyond the c-kit

+

FcεRI

+

mast cells. Whether these cells were basophils or immature mast cells is still unclear. By day 9, how- ever, these cells were no longer present. This finding either suggests that these cells are basophils that undergo apoptosis or are in an intermediate state before forming mature mast cells. Several studies support the idea that mast cells lack expression of c-kit while expressing IgE receptors during a transitional state in the cells’ development

7,97,98

. Additionally, Arinobu et al reported that Lin

-

c-kit

hi

integrin β7

hi

CD16/32

hi

cells isolated from the spleen of C57BL/6 mice become both c-kit

+

FcεRI

+

mast cells and c-kit

-

FcεRI

+

basophils

8

. This regulation into mast cells or basophils is controlled by the transcription factor CCAAT/enhancer-binding protein α (C/EBPα).

Overexpression of C/EBPα leads to the appearance of basophils, whereas deletion induces the formation of mast cells. Recently, it was also reported that C/EBPα expression inhibits microphthalmia-associated transcription factor (MITF), which is necessary for mast cell development

99

. Culturing bone marrow cells with mutated MITF in medium containing IL-3 results in

Bone marrow

Spleen

Blood

Peripheral organs

MPP

HSC SL-CMP

BMCP

C57BL/6 Lin

-

Sca-1

-

Ly6c

-

CD27

-

integrin β7

+

ST2

+

c-kit

+

FcεRI

-

BALB/c

AA4

-

BGD6

+

(c-kit

+

FcεRI

-

)

C57BL/6

Lin

-

c-kit

+

integrin β7

hi

CD16/32

hi

(FcεRI

-

)

C57BL/6

Lin

-

CD45

+

CD34

+

integrin β7

hi

FcεRI

lo

Less mature More mature

MCp

MCp C57BL/6 & BALB/c

Lin

-

c-kit

hi

ST2

+

integrin β7

hi

CD16/32

hi

(FcεRI

-/+

)

MCp

(28)

As we now report that the Th2-prone BALB/c strain has more mature

MCp than do Th1-prone C57BL/6 mice, the maturation status of MCp may

differ between human individuals as well. In particular, Th2-prone individu-

als may have more mature MCp than do Th1 responders.

(29)

Concluding remarks and future perspectives

Mast cells’ strategic position and their array of bronchoconstrictive media- tors support these cells’ role in allergic asthma. In mouse models of the dis- ease, an increase in lung MCp precedes the appearance of mature mast cells.

In this thesis, the mechanisms leading to an increased number of lung MCp have been explored.

Although IgE is not critically required for MCp recruitment to the lung, paper I described a new pathway in which IgE immune complexes are suffi- cient to induce the increase in lung MCp. Because asthmatics have elevated IgE levels, it is likely that IgE immune complexes are formed locally in the lung. This phenomenon would result in the recruitment of MCp. After matu- ration, the increase in MCp results in an increased number of pulmonary mast cells. Treatment with anti-IgE might therefore not only reduce IgE lev- els but also lead to the impaired recruitment of MCp to the lung. In the long term, this impairment would lead to fewer mature pulmonary mast cells.

In paper II, CD11c

+

cells were found to be critical for the upregulation of endothelial VCAM-1 during the antigen challenge phase. The cells thereby regulated the increase in lung MCp and the subsequent appearance of mature mast cells. CD11c

+

cells were proposed to regulate VCAM-1 via TNF-α.

However, verification of this pathway needs to be performed. A summarized view of the mechanisms that regulate the increase in lung mast cells is illus- trated in Figure 9.

In vitro-derived mast cells are often used to find potential receptors in-

volved in MCp migration. However, in vitro-derived mast cells are not nec-

essarily similar to blood MCp. The identification of circulating MCp would

therefore facilitate the search for critical receptors and mediators that regu-

late the cells’ recruitment to different tissues. In paper III, committed MCp

were identified in the peripheral blood of adult mice for the first time. Char-

acterization of these cells is ongoing. The results will reveal which receptors

are expressed on the surface of MCp and are potentially involved in recruit-

ment to different tissues. As a second step, confirming the role of the recep-

tor(s) in the in vivo recruitment of MCp to the lung will be performed.

(30)

Figure 9. An updated view of the mechanisms that regulate the increase in lung mast cells during allergic airway inflammation. CD11c

+

dendritic cells (DCs) and alveolar macrophages (AMs) upregulate the expression of endothelial VCAM-1, most likely by producing TNF-α. CXCR2 activation also induces higher VCAM-1 expression.

VCAM-1 interacts with integrins α4β1 and α4β7 on the MCp surface, allowing the MCp to migrate through the endothelium. CCR2 binding to its main ligand CCL2 also controls the recruitment of lung MCp. Type 2 NKT cells produce IL-9, which stimulates the expansion of the MCp. The MCp in the tissue matures and develop granules.

In humans, mast cell forming-capacity has been described in the CD34

+

CD13

+

c-kit

+

population of the peripheral blood

17

. This population is not committed to the mast cell lineage, as the cells also give rise to monocytes

17

. As committed MCp could be isolated from mouse blood, it is likely that committed MCp are found within a subset of CD34

+

CD13

+

c-kit

+

cells in human blood. Ongoing experiments will reveal whether such a progenitor exists. Characterization of these human MCp will hopefully reveal new po- tential drug targets.

Blood

TNF-α e.g.

CD11c

+

DC

Mast cell progenitor

Lung

Mature mast cell

VCAM-1 CD11c

+

AM

Endothelium

CXCR2

Integrin α4β1 Integrin α4β7

CCR2

CCL2 Type 2

NKT cell IL-9

(31)

Populärvetenskaplig sammanfattning på svenska

Förekomsten av astma har ökat de senaste årtiondena, och uppskattningsvis lider 300 miljoner personer av sjukdomen. Astmatiska symptom uppkommer ofta till följd av en allergisk reaktion, exempelvis vid exponering av pollen, kvalster eller pälsdjur. Allergiska astmatiker har så kallade IgE-antikroppar som binder till allergener, det vill säga ämnen som personen är överkänslig mot. IgE-antikropparna sitter bundna till en av kroppens egna immunceller – mastcellen. När en astmatiker andas in allergen binder allergenet till IgE- antikropparna på mastcellernas yta. Mastcellerna töms då snabbt på sitt in- nehåll i luftvägarna. Vissa av de ämnen som släpps fria påverkar luftvägarna och gör att de dras samman. Personen får då svårt att andas.

Astmatiker har fler mastceller i luftvägarna än friska personer. Mycket pekar på att ökningen beror på att omogna celler som endast har potential att bilda mastceller, så kallade förutbestämda mastcellsprogenitorer, förflyttar sig från blodet in i lungan. Trots starka indikationer på att så är fallet har man inte tidigare lyckats påvisa förekomsten av förutbestämda mastcellspro- genitorer i blod. I delarbete III identifierade och isolerade vi förutbestämda mastcellsprogenitorer i musblod. Vi visade också att möss som har stor ge- netisk benägenhet att utveckla allergier hade mognare mastcellsprogenitorer än andra möss. Om detta fenomen återspeglas hos allergiska kontra friska människor återstår att se.

Astmatiker har höga nivåer IgE-antikroppar i blodet. I delarbete I upp- täckte vi att allergener som binder till IgE-antikroppar kunde orsaka en ök- ning av omogna mastceller i lungorna hos allergiska möss. Detta styrdes sannolikt av IgE-bindande proteiner som går under namnet FcεRI. Om IgE- antikroppar som binder allergener kan orsaka en ökning av mastcellsprogeni- torer i lungorna även hos astmatiska människor, kan detta på sikt orsaka en ökning av mogna mastceller. Ett ökat antal mogna mastceller skulle i sin tur förvärra de astmatiska symptomen.

I delarbete II undersökte vi betydelsen av alveolära makrofager och dend-

ritiska celler, två vanligt förekommande celltyper i lungan, för ökningen av

(32)

VCAM-1 fungerar som ett slags klister som specifikt bromsar upp omogna mastceller och låter dem passera in i lungan. Ökningen av mastcellsprogeni- torer följdes av ett ökat antal mastceller i lungan.

Våra studier har öppnat upp möjligheten att undersöka mastcellsprogeni-

torer i blod närmare. Vi undersöker nu vilka molekyler på mastcellsprogeni-

torernas yta som styr vandringen från blod till lungan. Våra studier har också

lett till en fördjupad förståelse om vilka celler och molekyler som bidrar till

ett ökat antal mastcellsprogenitorer i lungan vid allergisk luftvägsinflammat-

ion. Dagens astmaläkemedel riktas mot att förhindra effekterna av de ämnen

som mastceller släpper ut i luftvägarna vid en allergisk reaktion. Våra studier

kan förhoppningsvis bidra till utvecklingen av läkemedel som stoppar inflö-

det av mastcellsprogenitorer till lungan.

(33)

Acknowledgements

Jag vill tacka följande personer:

Min handledare Jenny Hallgren, för din oändliga entusiasm gällande pro- jekten. Tack för din ständiga uppmuntran och för alla snabba svar på frågor dygnet runt.

Min biträdande handledare, Birgitta Heyman, som visat vägen när vi tappat fokus. Stort tack för din inspiration, erfarenhet, och visdom!

Alla i gruppen: Anna Rolfsdotter Bergman, för allt peppande när vi skrev lic-uppsatsen och för svar på kliniska frågor. Zhoujie Ding, för alla galna pilotexperiment som vi gjort. Tack också för att du introducerade dumplings i mitt liv. Hui Xu, för all kinesisk mat och för alla Bike Race-tävlingar. An- nika Grahn-Westin, för det oändliga hackandet på cellabb. Behdad Zarne- gar, för alla mastcellsrelaterade diskussioner. Joakim Bergström, för att du alltid livat upp stämningen på labbet och varit tillgänglig för en extrainsatt after work. Lu Zhang, för alla pratstunder när alla andra gått hem!

Tidigare gruppmedlemmar: Christian Rutemark, för alla roliga stunder i och utanför labbet och för alla spratt mot medarbetare. Frida Henningsson, för att du handledde mig under hela exjobbet, och gjorde att jag utan tvekan började doktorera. Tack också för att du alltid kommit till jobbet med ett smittsamt leende och en positiv inställning! Yui Cui, som borde blivit dok- torand hos oss…

Nuvarande eller tidigare kollegor på bmc: Anna-Karin Palm, min lab-

bänksgranne, för ditt sällskap under sena kvällar och för att du stod ut med

all grabbhumor. Lisbeth Fuxler, för att du alltid tog dig tid att hjälpa till på

labbet och för alla pikar om att jag äter mer godis än du. Ida Waern och

Elin Rönnberg, för att ni introducerade mig till doktorandstudier med mid-

nattspulkaåkning på konferenser. Michael Thorpe, för sällskap under mast-

(34)

Hjertner, Tommy Linné, Kersti Larsson, Ananya Roy och alla IMBIM:s administratörer.

Tack till min mamma, pappa, syster (jää!) och Karoline för allt stöd under

hela doktorandperioden!

(35)

References

1. Galli, S.J. & Tsai, M. IgE and mast cells in allergic disease. Nat. Med. 18, 693-704 (2012).

2. Brightling, C.E., et al. Mast-cell infiltration of airway smooth muscle in asthma. N. Engl. J. Med. 346, 1699-1705 (2002).

3. Andersson, C.K., et al. Mast cell-associated alveolar inflammation in patients with atopic uncontrolled asthma. J. Allergy Clin. Immunol. 127, 905-912 (2011).

4. Hallgren, J. & Gurish, M.F. Mast cell progenitor trafficking and maturation. Adv. Exp. Med. Biol. 716, 14-28 (2011).

5. Morrison, S.J. & Weissman, I.L. The long-term repopulating subset of hematopoietic stem cells is deterministic and isolatable by phenotype.

Immunity 1, 661-673 (1994).

6. Akashi, K., Traver, D., Miyamoto, T. & Weissman, I.L. A clonogenic common myeloid progenitor that gives rise to all myeloid lineages. Nature 404, 193-197 (2000).

7. Franco, C.B., Chen, C.C., Drukker, M., Weissman, I.L. & Galli, S.J.

Distinguishing Mast Cell and Granulocyte Differentiation at the Single-Cell Level. Cell Stem Cell 6, 361-368 (2010).

8. Arinobu, Y., et al. Developmental checkpoints of the basophil/mast cell lineages in adult murine hematopoiesis. Proc. Natl. Acad. Sci. U. S. A. 102, 18105-18110 (2005).

9. Suda, T., Suda, J. & Ogawa, M. Single-cell origin of mouse hemopoietic colonies expressing multiple lineages in variable combinations. Proc. Natl.

Acad. Sci. U. S. A. 80, 6689-6693 (1983).

10. Rodewald, H.R., Dessing, M., Dvorak, A.M. & Galli, S.J. Identification of a committed precursor for the mast cell lineage. Science 271, 818-822 (1996).

11. Chen, C.C., Grimbaldeston, M.A., Tsai, M., Weissman, I.L. & Galli, S.J.

Identification of mast cell progenitors in adult mice. Proc. Natl. Acad. Sci.

U. S. A. 102, 11408-11413 (2005).

12. Jamur, M.C., et al. Identification and characterization of undifferentiated mast cells in mouse bone marrow. Blood 105, 4282-4289 (2005).

13. Guiraldelli, M.F., et al. The low affinity IgG receptor Fc gamma RIIB

(36)

14. Guo, N.H., et al. Monoclonal antibody AA4, which inhibits binding of IgE to high affinity receptors on rat basophilic leukemia cells, binds to novel alpha-galactosyl derivatives of ganglioside GD1b. J. Biol. Chem. 264, 13267-13272 (1989).

15. Kitamura, Y., Shimada, M., Hatanaka, K. & Miyano, Y. Development of mast cells from grafted bone marrow cells in irradiated mice. Nature 268, 442 (1977).

16. Kempuraj, D., et al. Characterization of mast cell-committed progenitors present in human umbilical cord blood. Blood 93, 3338-3346 (1999).

17. Kirshenbaum, A.S., et al. Demonstration that human mast cells arise from a progenitor cell population that is CD34(+), c-kit(+), and expresses aminopeptidase N (CD13). Blood 94, 2333-2342 (1999).

18. Casale, T.B., et al. Direct evidence of a role for mast cells in the pathogenesis of antigen-induced bronchoconstriction. J. Clin. Invest. 80, 1507-1511 (1987).

19. Liu, M.C., et al. Immediate and late inflammatory responses to ragweed antigen challenge of the peripheral airways in allergic asthmatics. Cellular, mediator, and permeability changes. Am. Rev. Respir. Dis. 144, 51-58 (1991).

20. Murray, J.J., et al. Release of prostaglandin D2 into human airways during acute antigen challenge. N. Engl. J. Med. 315, 800-804 (1986).

21. Wenzel, S.E., Fowler, A.A., 3rd & Schwartz, L.B. Activation of pulmonary mast cells by bronchoalveolar allergen challenge. In vivo release of histamine and tryptase in atopic subjects with and without asthma. Am. Rev.

Respir. Dis. 137, 1002-1008 (1988).

22. Koshino, T., et al. Identification of basophils by immunohistochemistry in the airways of post-mortem cases of fatal asthma. Clin. Exp. Allergy 23, 919-925 (1993).

23. Abonia, J.P., et al. Alpha-4 integrins and VCAM-1, but not MAdCAM-1, are essential for recruitment of mast cell progenitors to the inflamed lung.

Blood 108, 1588-1594 (2006).

24. Hallgren, J., et al. Pulmonary CXCR2 regulates VCAM-1 and antigen- induced recruitment of mast cell progenitors. Proc. Natl. Acad. Sci. U. S. A.

104, 20478-20483 (2007).

25. Reuter, S., et al. Mast cell-derived tumour necrosis factor is essential for allergic airway disease. Eur. Respir. J. 31, 773-782 (2008).

26. Yu, M., et al. Identification of an IFN-gamma/mast cell axis in a mouse model of chronic asthma. J. Clin. Invest. 121, 3133-3143 (2011).

27. Yu, M., et al. Mast cells can promote the development of multiple features of chronic asthma in mice. J. Clin. Invest. 116, 1633-1641 (2006).

28. Kobayashi, T., et al. An essential role of mast cells in the development of airway hyperresponsiveness in a murine asthma model. J. Immunol. 164, 3855-3861 (2000).

29. Sawaguchi, M., et al. Role of mast cells and basophils in IgE responses and

in allergic airway hyperresponsiveness. J. Immunol. 188, 1809-1818

(2012).

References

Related documents

The two studies above confirmed the correlation between Th2 cells and eosinophils during allergic airway inflammation. ST2 is expressed on the cell surface of both Th2 cells

Although the role of eosinophils and Th cells has been studied extensively, the balance of different Th cells during eosinophilic inflammation and the corresponding local

After a few hours of exposure in the swine confinement building healthy, naïve, non-allergic volunteers develop increased bronchial responsiveness, intense airway

The aim of this thesis was to characterize the presence and function of TLRs in different cell types involved in upper airway inflammation, with special emphasis on

Since recruitment of white blood cell to lung is one of the characteristics of allergic asthma, we further investigated if mast cells attract certain white blood cells

The aim of this study is to further elucidate the role of mMCP-6 in allergic airway inflammation and to investigate the mechanism behind the attenuated allergic response in

[r]

In this study, the feasibility of mesoporous silica particles (MSPs) as carriers for anti-inflammatory drugs in the treatment of airway inflammation was investigated.. Two