• No results found

The role of intracellular thyroid hormone metabolism in innate immune cells - Chapter 3: The thyroid hormone inactivating enzyme type 3 deiodinase is present in bactericidal granules and the cytoplasm of human neutro

N/A
N/A
Protected

Academic year: 2021

Share "The role of intracellular thyroid hormone metabolism in innate immune cells - Chapter 3: The thyroid hormone inactivating enzyme type 3 deiodinase is present in bactericidal granules and the cytoplasm of human neutro"

Copied!
23
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)

The role of intracellular thyroid hormone metabolism in innate immune cells

van der Spek, A.H.

Publication date

2018

Document Version

Other version

License

Other

Link to publication

Citation for published version (APA):

van der Spek, A. H. (2018). The role of intracellular thyroid hormone metabolism in innate

immune cells.

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)

516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek Processed on: 16-5-2018 Processed on: 16-5-2018 Processed on: 16-5-2018

Processed on: 16-5-2018 PDF page: 34PDF page: 34PDF page: 34PDF page: 34

ENZYME TYPE 3 DEIODINASE IS PRESENT

IN BACTERICIDAL GRANULES AND THE

CYTOPLASM OF HUMAN NEUTROPHILS

Anne H. van der Spek, Flavia F. Bloise, Wikky Tichgelaar, Monica Dentice,

Domenico Salvatore, Nicole N. van der Wel, Eric Fliers, Anita Boelen

Endocrinology 2016 Aug;157(8):3293-305

516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek Processed on: 16-5-2018 Processed on: 16-5-2018 Processed on: 16-5-2018

Processed on: 16-5-2018 PDF page: 35PDF page: 35PDF page: 35PDF page: 35

ENZYME TYPE 3 DEIODINASE IS PRESENT

IN BACTERICIDAL GRANULES AND THE

CYTOPLASM OF HUMAN NEUTROPHILS

Anne H. van der Spek, Flavia F. Bloise, Wikky Tichgelaar, Monica Dentice,

Domenico Salvatore, Nicole N. van der Wel, Eric Fliers, Anita Boelen

(3)

516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek Processed on: 16-5-2018 Processed on: 16-5-2018 Processed on: 16-5-2018

Processed on: 16-5-2018 PDF page: 36PDF page: 36PDF page: 36PDF page: 36

36

Abstract

Neutrophils are important effector cells of the innate immune system. Thyroid hormone is thought to play an important role in their function. Intracellular thyroid hormone levels are regulated by the deiodinating enzymes. The thyroid hormone inactivating type 3 deiodinase (D3) is expressed in infiltrating murine neutrophils and D3 knockout mice show impaired bacterial killing upon infection. This suggests that D3 plays an important role in the bacterial killing capacity of neutrophils. The mechanism behind this effect is unknown. We aimed to assess the presence of D3 in human neutrophils, and determine its subcellular localization using confocal and electron microscopy as this could give important clues about its function in these cells. D3 appeared to be present in the cytoplasm and in myeloperoxidase (MPO) containing azurophilic granules and as well as lactoferrin containing specific granules within human neutrophils. This subcellular localization did not change upon activation of the cells. D3 is observed intracellularly during neutrophil extracellular trap (NET) formation, followed by a reduction of D3 staining after release of the NETs into the extracellular space. At the transcriptional level, human neutrophils expressed additional essential elements of thyroid hormone metabolism, including thyroid hormone transporters and thyroid hormone receptors. Here we demonstrate the presence and subcellular location of D3 in human neutrophils for the first time and propose a model in which D3 plays a role in the bacterial killing capacity of neutrophils either through generation of iodide for the MPO system or through modulation of intracellular thyroid hormone bioavailability.

516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek Processed on: 16-5-2018 Processed on: 16-5-2018 Processed on: 16-5-2018

Processed on: 16-5-2018 PDF page: 37PDF page: 37PDF page: 37PDF page: 37

37

3

Introduction

Thyroid hormone metabolism is known to undergo profound changes during illness and infection, collectively known as non thyroidal illness syndrome (NTIS) (Docter et al., 1993, Boelen et al., 2011). These changes are characterized by low serum triiodothyronine (T3) and, in prolonged illness, low serum thyroxine (T4) concentrations (Bello et al., 2010, Gangemi et al., 2008). NTIS is present in a wide range of diseases including critical illness (Bello et al., 2010), myocardial infarction (Lazzeri et al., 2012, Ozcan et al., 2014), Crohn’s disease (Liu et al., 2013b) and stroke (Alevizaki et al., 2007) and is correlated with both disease severity and outcome. Besides alterations in systemic thyroid hormone (TH) levels, NTIS is also known to affect TH metabolism at the tissue and cellular level, resulting in changes in local T3 concentration (Boelen et al., 2011). One of the cell types in which TH metabolism appears to play a role during infection is neutrophils (Boelen et al., 2005).

Neutrophils are important effector cells of the innate immune system that phagocytose and kill bacteria and other pathogens. They are the most abundant type of blood leukocyte and, as the first cells to arrive at the site of infection, a key component of the inflammatory response (Bardoel et al., 2014, Borregaard, 2010, Kolaczkowska and Kubes, 2013). Research from the 1970s has shown that phagocytosing human leukocytes can actively degrade significant amounts of TH, both T3 and T4 (Woeber and Ingbar, 1973, Woeber et al., 1972, Klebanoff and Green, 1973). The mechanism behind this is not fully understood. TH can be metabolized by the deiodinase enzymes. Deiodinases regulate the availability of biologically active TH at the cellular and tissue level by removing an iodine atom from the TH molecule, resulting in the production of free iodide (I-) (Bianco and Kim, 2006, Bianco et al., 2002). The activating deiodinases types 1 and 2 convert the prohormone T4 to the active hormone T3 while the TH inactivating deiodinase type 3 (D3) is responsible for the intracellular conversion of T3 and T4 to the biologically inactive diiodothyronine (T2) and reverse (r)T3 respectively. The expression of the different types of deiodinases is cell type specific (Bianco and Kim, 2006, Bianco et al., 2002).

Various mouse models have suggested that D3 plays a role in neutrophils during infection and inflammation. Infiltrating neutrophils in sections of inflamed lung from mice with pulmonary infection with Streptococcus pneumoniae markedly expressed D3 protein (Boelen et al., 2008). Furthermore, in a model for chronic local inflammation in which mice were subcutaneously injected with turpentine resulting in a sterile abscess, infiltrating neutrophils showed strong D3 protein expression while D3 activity was significantly increased in hind limb tissue containing the abscess compared to control tissue (Boelen et al., 2005). Finally, D3 knockout mice showed impaired bacterial killing following Streptococcus pneumoniae infection compared to wildtype mice (Boelen et al., 2009). These results suggest that D3 plays an important role in the bacterial killing capacity of neutrophils.

Although D3 is known to be present in infiltrating murine neutrophils, its presence in human neutrophils has not been previously studied. Furthermore, nothing is currently known on its subcellular location in neutrophils. The location of D3 within the cell could give important clues as to its functional role. We aimed to determine the presence and subcellular location of D3 in resting and activated human

(4)

516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek Processed on: 16-5-2018 Processed on: 16-5-2018 Processed on: 16-5-2018

Processed on: 16-5-2018 PDF page: 36PDF page: 36PDF page: 36PDF page: 36

36

Abstract

Neutrophils are important effector cells of the innate immune system. Thyroid hormone is thought to play an important role in their function. Intracellular thyroid hormone levels are regulated by the deiodinating enzymes. The thyroid hormone inactivating type 3 deiodinase (D3) is expressed in infiltrating murine neutrophils and D3 knockout mice show impaired bacterial killing upon infection. This suggests that D3 plays an important role in the bacterial killing capacity of neutrophils. The mechanism behind this effect is unknown. We aimed to assess the presence of D3 in human neutrophils, and determine its subcellular localization using confocal and electron microscopy as this could give important clues about its function in these cells. D3 appeared to be present in the cytoplasm and in myeloperoxidase (MPO) containing azurophilic granules and as well as lactoferrin containing specific granules within human neutrophils. This subcellular localization did not change upon activation of the cells. D3 is observed intracellularly during neutrophil extracellular trap (NET) formation, followed by a reduction of D3 staining after release of the NETs into the extracellular space. At the transcriptional level, human neutrophils expressed additional essential elements of thyroid hormone metabolism, including thyroid hormone transporters and thyroid hormone receptors. Here we demonstrate the presence and subcellular location of D3 in human neutrophils for the first time and propose a model in which D3 plays a role in the bacterial killing capacity of neutrophils either through generation of iodide for the MPO system or through modulation of intracellular thyroid hormone bioavailability.

516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek Processed on: 16-5-2018 Processed on: 16-5-2018 Processed on: 16-5-2018

Processed on: 16-5-2018 PDF page: 37PDF page: 37PDF page: 37PDF page: 37

37

3

Introduction

Thyroid hormone metabolism is known to undergo profound changes during illness and infection, collectively known as non thyroidal illness syndrome (NTIS) (Docter et al., 1993, Boelen et al., 2011). These changes are characterized by low serum triiodothyronine (T3) and, in prolonged illness, low serum thyroxine (T4) concentrations (Bello et al., 2010, Gangemi et al., 2008). NTIS is present in a wide range of diseases including critical illness (Bello et al., 2010), myocardial infarction (Lazzeri et al., 2012, Ozcan et al., 2014), Crohn’s disease (Liu et al., 2013b) and stroke (Alevizaki et al., 2007) and is correlated with both disease severity and outcome. Besides alterations in systemic thyroid hormone (TH) levels, NTIS is also known to affect TH metabolism at the tissue and cellular level, resulting in changes in local T3 concentration (Boelen et al., 2011). One of the cell types in which TH metabolism appears to play a role during infection is neutrophils (Boelen et al., 2005).

Neutrophils are important effector cells of the innate immune system that phagocytose and kill bacteria and other pathogens. They are the most abundant type of blood leukocyte and, as the first cells to arrive at the site of infection, a key component of the inflammatory response (Bardoel et al., 2014, Borregaard, 2010, Kolaczkowska and Kubes, 2013). Research from the 1970s has shown that phagocytosing human leukocytes can actively degrade significant amounts of TH, both T3 and T4 (Woeber and Ingbar, 1973, Woeber et al., 1972, Klebanoff and Green, 1973). The mechanism behind this is not fully understood. TH can be metabolized by the deiodinase enzymes. Deiodinases regulate the availability of biologically active TH at the cellular and tissue level by removing an iodine atom from the TH molecule, resulting in the production of free iodide (I-) (Bianco and Kim, 2006, Bianco et al., 2002). The activating deiodinases types 1 and 2 convert the prohormone T4 to the active hormone T3 while the TH inactivating deiodinase type 3 (D3) is responsible for the intracellular conversion of T3 and T4 to the biologically inactive diiodothyronine (T2) and reverse (r)T3 respectively. The expression of the different types of deiodinases is cell type specific (Bianco and Kim, 2006, Bianco et al., 2002).

Various mouse models have suggested that D3 plays a role in neutrophils during infection and inflammation. Infiltrating neutrophils in sections of inflamed lung from mice with pulmonary infection with Streptococcus pneumoniae markedly expressed D3 protein (Boelen et al., 2008). Furthermore, in a model for chronic local inflammation in which mice were subcutaneously injected with turpentine resulting in a sterile abscess, infiltrating neutrophils showed strong D3 protein expression while D3 activity was significantly increased in hind limb tissue containing the abscess compared to control tissue (Boelen et al., 2005). Finally, D3 knockout mice showed impaired bacterial killing following Streptococcus pneumoniae infection compared to wildtype mice (Boelen et al., 2009). These results suggest that D3 plays an important role in the bacterial killing capacity of neutrophils.

Although D3 is known to be present in infiltrating murine neutrophils, its presence in human neutrophils has not been previously studied. Furthermore, nothing is currently known on its subcellular location in neutrophils. The location of D3 within the cell could give important clues as to its functional role. We aimed to determine the presence and subcellular location of D3 in resting and activated human

(5)

516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek Processed on: 16-5-2018 Processed on: 16-5-2018 Processed on: 16-5-2018

Processed on: 16-5-2018 PDF page: 38PDF page: 38PDF page: 38PDF page: 38

38

neutrophils to provide important insights into the role of local TH metabolism in the bacterial killing machinery of these essential cells of the innate immune system.

Material and Methods

Neutrophil isolation

Venous blood was obtained from healthy male and female volunteers following written informed consent. This study was approved by the local medical ethical committee of the Academic Medical Center of the University of Amsterdam in accordance with the principles of the Declaration of Helsinki (version Fortaleza, 2013).

Neutrophils were isolated as described previously (Kuijpers et al., 1991, Roos and de Boer, 1986, Roos et al., 2014). Neutrophils were suspended in HEPES buffered medium (Roos et al., 2014) and kept at room temperature (RT) until use. Cell count and viability was assessed using trypan blue. Neutrophil purity was assessed by flow cytometry staining for CD16 (neutrophils) and CD49d (eosinophils) and was always at least 90%.

Neutrophil stimulation

Neutrophils were incubated with stimuli in a shaking water bath at 37⁰C for 15 minutes. Phorbol myristate acetate (PMA) (Sigma-Aldrich) was stored as a stock solution in DMSO and diluted in HEPES-medium at least 1000x immediately prior to use. Zymosan (Sigma-Aldrich) was opsonized with human serum and added to cells at a ratio of 20 particles per cell. Neutrophil extracellular traps (NETs) were generated and visualized as described in Brinkmann et al. (Brinkmann et al., 2010). Briefly, neutrophils were allowed to adhere to glass coverslips after which they were stimulated with PMA for up to 3 hours in a 37⁰C incubator with 5% CO2. Coverslips were then stained and imaged as described under Confocal Microscopy.

Western Blot

Cell lysates of freshly isolated unstimulated neutrophils were produced as previously described (Roos et al., 2014). Lysates were stored at -20⁰C until use. Cell lysates were loaded on a 10% SDS-PAGE gel. Gels were blotted on to PVDF membrane and processed as described previously (de Vries et al., 2014b). Primary antibodies used were polyclonal rabbit anti-D3 #676 (dilution 1:500, kindly provided by prof. dr. T.J.Visser, Erasmus Medical Center, Rotterdam, the Netherlands) and polyclonal goat anti-actin I-19 (dilution 1:5000; Santa Cruz Biotechnology). The #676 D3 antibody was raised against the synthetic peptide (C)RYDEQLHGARPRRV (human D3 amino acid residues 265–278) (Kuiper et al., 2003). Secondary antibodies were horseradish peroxidase-conjugated goat-anti-rabbit (1:10,000) and rabbit-anti-goat (1:20,000) antibodies (Dako). 516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek Processed on: 16-5-2018 Processed on: 16-5-2018 Processed on: 16-5-2018

Processed on: 16-5-2018 PDF page: 39PDF page: 39PDF page: 39PDF page: 39

39

3

Confocal Microscopy

Freshly isolated neutrophils were fixed with 4% paraformaldehyde (PFA) in phosphate buffered saline (PBS) at RT for 10 minutes. Cells were blocked with 10% normal goat serum (Abcam) and human FC receptor block (BD Biosciences) in saponin buffer (0,5% [wt/vol] saponin and 0,5% [wt/vol] bovine serum albumin in PBS) for 15 minutes. Neutrophils were incubated for 1 hour at RT with primary antibodies in saponin buffer followed by incubation with appropriate secondary antibodies in saponin buffer for 30 minutes at RT. The stained cell suspension was mounted on a glass slide with Prolong Gold antifade reagent with DAPI (Life Technologies). Slides were imaged by a confocal laser-scanning system (Leica TCS SP8 X) using the Leica DMI6000 inverted microscope and the 63x/1.40 Oil CS2 objective. Images were analyzed using the Leica LAS X software (version 1.1).

The primary D3 antibody used was #718 (dilution 1:500), raised against the synthetic peptide KPEPEVELNSEGEEVP (human D3 amino acid residues 53-68) (Huang et al., 2003). Other primary antibodies used were anti-EEA1 (1:300, BD Biosciences), FITC-conjugated anti-myeloperoxidase (dilution 1:12.5, clone CLB-MPO-1/1, Novus Biologicals), anti-Lactoferrin (1:500, clone 2B8, Abcam), anti-matrix metallopeptidase 9 (1:75, clone 56-2A4, Abcam) and anti-human albumin (1:500, clone HSA-9, Sigma-Aldrich). Secondary antibodies used were Alexa Fluor 568-conjugated goat-anti-rabbit and/or Alexa Fluor 647-conjugated goat-anti-mouse IgG1 (1:500; both Life Technologies). All stainings included a control without primary antibody to assess background staining.

To assess the specificity of D3 staining, the #718 antibody was preincubated with the corresponding peptide. This resulted in the disappearance of all D3 staining (Supplemental Figure 3.1 A-B), supporting the specificity of the staining. The #718 antibody also gave clear staining of SK-N-AS cells, a human neuroblastoma cell line known to contain D3 (Supplemental Figure 3.1 C-D) (Jo et al., 2012, Freitas et al., 2010). The SK-N-AS cell line was a kind gift of dr. F. Ponds (Dept. of Gastroenterology, Academic Medical Center, Amsterdam) and was cultured as described previously (Freitas et al., 2010).

Electron Microscopy

Isolated neutrophils were incubated in a 37⁰C shaking water bath (5x106 cells/tube) for 15 minutes with PMA (20ng/ml), serum opsonized zymosan or no stimulus. After stimulation samples were fixed and processed as described in van der Wel et al. (van der Wel et al., 2007). In short: following fixation in 2% PFA for 24 hours, samples were pelleted in 12% gelatin, cut in 2-3 mm2 blocks and incubated in 2.3M sucrose overnight. Blocks were snap frozen in liquid nitrogen and 60 nm thick sections were cut at -120⁰C using the Leica microtome UC6 with cryochamber Ultracut EM FC6 and a diamond cryo-immuno knife (35° Diatome). Immunogold labelling was performed using primary antibodies anti-D3 (#718; 1:25), polyclonal rabbit anti-MPO (1:14.000; Dako) and anti-Lactoferrin (1:200; Cappel Laboratories) and 10 or 15 nm gold labelled protein (Aurion) or with rabbit anti-mouse bridging serum (1:200; Dako) and protein-A-conjugated to 10 nm or 15 nm gold (Utrecht University, the Netherlands). Immunogold labelled grids were analyzed using a Tecnai 12 with 2k×2k CCD Camera (Veleta, Olympus).

(6)

516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek Processed on: 16-5-2018 Processed on: 16-5-2018 Processed on: 16-5-2018

Processed on: 16-5-2018 PDF page: 38PDF page: 38PDF page: 38PDF page: 38

38

neutrophils to provide important insights into the role of local TH metabolism in the bacterial killing machinery of these essential cells of the innate immune system.

Material and Methods

Neutrophil isolation

Venous blood was obtained from healthy male and female volunteers following written informed consent. This study was approved by the local medical ethical committee of the Academic Medical Center of the University of Amsterdam in accordance with the principles of the Declaration of Helsinki (version Fortaleza, 2013).

Neutrophils were isolated as described previously (Kuijpers et al., 1991, Roos and de Boer, 1986, Roos et al., 2014). Neutrophils were suspended in HEPES buffered medium (Roos et al., 2014) and kept at room temperature (RT) until use. Cell count and viability was assessed using trypan blue. Neutrophil purity was assessed by flow cytometry staining for CD16 (neutrophils) and CD49d (eosinophils) and was always at least 90%.

Neutrophil stimulation

Neutrophils were incubated with stimuli in a shaking water bath at 37⁰C for 15 minutes. Phorbol myristate acetate (PMA) (Sigma-Aldrich) was stored as a stock solution in DMSO and diluted in HEPES-medium at least 1000x immediately prior to use. Zymosan (Sigma-Aldrich) was opsonized with human serum and added to cells at a ratio of 20 particles per cell. Neutrophil extracellular traps (NETs) were generated and visualized as described in Brinkmann et al. (Brinkmann et al., 2010). Briefly, neutrophils were allowed to adhere to glass coverslips after which they were stimulated with PMA for up to 3 hours in a 37⁰C incubator with 5% CO2. Coverslips were then stained and imaged as described under Confocal Microscopy.

Western Blot

Cell lysates of freshly isolated unstimulated neutrophils were produced as previously described (Roos et al., 2014). Lysates were stored at -20⁰C until use. Cell lysates were loaded on a 10% SDS-PAGE gel. Gels were blotted on to PVDF membrane and processed as described previously (de Vries et al., 2014b). Primary antibodies used were polyclonal rabbit anti-D3 #676 (dilution 1:500, kindly provided by prof. dr. T.J.Visser, Erasmus Medical Center, Rotterdam, the Netherlands) and polyclonal goat anti-actin I-19 (dilution 1:5000; Santa Cruz Biotechnology). The #676 D3 antibody was raised against the synthetic peptide (C)RYDEQLHGARPRRV (human D3 amino acid residues 265–278) (Kuiper et al., 2003). Secondary antibodies were horseradish peroxidase-conjugated goat-anti-rabbit (1:10,000) and rabbit-anti-goat (1:20,000) antibodies (Dako). 516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek Processed on: 16-5-2018 Processed on: 16-5-2018 Processed on: 16-5-2018

Processed on: 16-5-2018 PDF page: 39PDF page: 39PDF page: 39PDF page: 39

39

3

Confocal Microscopy

Freshly isolated neutrophils were fixed with 4% paraformaldehyde (PFA) in phosphate buffered saline (PBS) at RT for 10 minutes. Cells were blocked with 10% normal goat serum (Abcam) and human FC receptor block (BD Biosciences) in saponin buffer (0,5% [wt/vol] saponin and 0,5% [wt/vol] bovine serum albumin in PBS) for 15 minutes. Neutrophils were incubated for 1 hour at RT with primary antibodies in saponin buffer followed by incubation with appropriate secondary antibodies in saponin buffer for 30 minutes at RT. The stained cell suspension was mounted on a glass slide with Prolong Gold antifade reagent with DAPI (Life Technologies). Slides were imaged by a confocal laser-scanning system (Leica TCS SP8 X) using the Leica DMI6000 inverted microscope and the 63x/1.40 Oil CS2 objective. Images were analyzed using the Leica LAS X software (version 1.1).

The primary D3 antibody used was #718 (dilution 1:500), raised against the synthetic peptide KPEPEVELNSEGEEVP (human D3 amino acid residues 53-68) (Huang et al., 2003). Other primary antibodies used were anti-EEA1 (1:300, BD Biosciences), FITC-conjugated anti-myeloperoxidase (dilution 1:12.5, clone CLB-MPO-1/1, Novus Biologicals), anti-Lactoferrin (1:500, clone 2B8, Abcam), anti-matrix metallopeptidase 9 (1:75, clone 56-2A4, Abcam) and anti-human albumin (1:500, clone HSA-9, Sigma-Aldrich). Secondary antibodies used were Alexa Fluor 568-conjugated goat-anti-rabbit and/or Alexa Fluor 647-conjugated goat-anti-mouse IgG1 (1:500; both Life Technologies). All stainings included a control without primary antibody to assess background staining.

To assess the specificity of D3 staining, the #718 antibody was preincubated with the corresponding peptide. This resulted in the disappearance of all D3 staining (Supplemental Figure 3.1 A-B), supporting the specificity of the staining. The #718 antibody also gave clear staining of SK-N-AS cells, a human neuroblastoma cell line known to contain D3 (Supplemental Figure 3.1 C-D) (Jo et al., 2012, Freitas et al., 2010). The SK-N-AS cell line was a kind gift of dr. F. Ponds (Dept. of Gastroenterology, Academic Medical Center, Amsterdam) and was cultured as described previously (Freitas et al., 2010).

Electron Microscopy

Isolated neutrophils were incubated in a 37⁰C shaking water bath (5x106 cells/tube) for 15 minutes with PMA (20ng/ml), serum opsonized zymosan or no stimulus. After stimulation samples were fixed and processed as described in van der Wel et al. (van der Wel et al., 2007). In short: following fixation in 2% PFA for 24 hours, samples were pelleted in 12% gelatin, cut in 2-3 mm2 blocks and incubated in 2.3M sucrose overnight. Blocks were snap frozen in liquid nitrogen and 60 nm thick sections were cut at -120⁰C using the Leica microtome UC6 with cryochamber Ultracut EM FC6 and a diamond cryo-immuno knife (35° Diatome). Immunogold labelling was performed using primary antibodies anti-D3 (#718; 1:25), polyclonal rabbit anti-MPO (1:14.000; Dako) and anti-Lactoferrin (1:200; Cappel Laboratories) and 10 or 15 nm gold labelled protein (Aurion) or with rabbit anti-mouse bridging serum (1:200; Dako) and protein-A-conjugated to 10 nm or 15 nm gold (Utrecht University, the Netherlands). Immunogold labelled grids were analyzed using a Tecnai 12 with 2k×2k CCD Camera (Veleta, Olympus).

(7)

516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek Processed on: 16-5-2018 Processed on: 16-5-2018 Processed on: 16-5-2018

Processed on: 16-5-2018 PDF page: 40PDF page: 40PDF page: 40PDF page: 40

40

their immunogold labelling. Granules were scored as either negative, MPO or LF positive, D3 positive or double positive for D3 and either MPO or LF. A total of 1880 granules were scored in sections double stained for D3 and LF. Granules were only considered positive for LF if they contained at least 2 gold grains. In sections double stained for MPO and D3 a total 678 granules were scored. All scoring was performed by the same person.

Figure 3.1. D3 is present in human neutrophils. (A) Western Blot for D3 (antibody #676) and Beta Actin in whole

cell lysate of unstimulated human neutrophils. (B) Confocal microscopy images of freshly isolated human neutrophils using the differential interference contrast (DIC) setting. (C-D) Human neutrophils show intracellular D3 staining (shown in red; antibody #718 with Alexa Fluor 568-conjugated secondary antibody) in both the cytoplasm and small vesicle-like structures distributed throughout the cytoplasm and in close proximity to the nucleus. Cell nuclei are stained with DAPI (shown in blue). Scale bars are 25 µm (B; C) or 3 µm (D) in length. Stainings were repeated in at least 3 separate donors, representative images are shown.

Flow Cytometry

Neutrophils were incubated in ice cold FACS buffer (PBS with 5% heat-inactivated fetal calf serum (FCS) and 0,5% [wt/vol] sodium azide) with human FC block (eBioscience) for 20 minutes at 4°C. Cells were then either stained directly or fixed and permeabilized first using the eBioscience Intracellular Fixation & Permeabilization Buffer Set. Staining with primary antibodies was performed for 30 minutes at 4°C, followed by staining with secondary antibody for 30 minutes at 4°C. A total of 100,000 events was acquired using a BD FACS Canto II flow cytometer and data was analyzed using FlowJo software (v.10). Viable CD16+CD49- cells were selected for analysis of D3 staining.

516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek Processed on: 16-5-2018 Processed on: 16-5-2018 Processed on: 16-5-2018

Processed on: 16-5-2018 PDF page: 41PDF page: 41PDF page: 41PDF page: 41

41

3

Antibodies used were anti human APC-eFluor780-conjugated CD16 (1:100), anti-human PE-conjugated CD49d (1:100) (both eBioscience) and anti-D3 (#718; 1:250) with Alexa Fluor 647- conjugated donkey anti rabbit (1:250, Biolegend). Cell viability was determined using propidium iodide or fixable viability dye (eFluor 520-conjugated), both from eBioscience.

RNA isolation, cDNA synthesis and qualitative PCR

Neutrophils were lysed in Trizol-Reagent (Life Technologies) at 5x106 cells/ml. RNA was isolated using the Nucleospin RNA extraction kit (Machery Nagel). RNA yield was determined using the Nanodrop 2000 and quality was checked on the Bioanalyzer 2100 (Agilent Technologies). cDNA was synthesized with equal RNA input (150 ng) using AMV Reverse Transcriptase enzyme with oligo d(T) primers (Roche). As a control for genomic DNA contamination, a cDNA synthesis reaction without reverse transcriptase was included. Qualitative PCR was carried out using the Lightcycler 480 (Roche) and SensiFAST SYBR No-ROX (Bioline). Following amplification, PCR products were analyzed on DNA agarose gel and visualized on the ImageQuant LAS4000 (GE Healthcare). Published primer sequences were used for all genes (Bakker, 2001, Chan et al., 2006, Kwakkel et al., 2014, Kwakkel et al., 2006, Silva et al., 2002) except SLC16A10 for which primers were obtained from the Harvard Primer Bank (no. 221139821c1). Primer sequences and amplicon lengths are listed in Table 3.1.

Gene Protein Forward primer Reverse primer Amplicon length (bp) Source

Dio1 D3 AGCCACGACAACTGGATACC ACTCCCAAATGTTGCACCTC 160 Kwakkel et al; 200627

Dio2 D2 CCTCCTCGATGCCTACAAAC TCCTTCTGTACTGGAGACATGC

82 (trans. var. 1&2); 190 (trans. var. 3); 324 (trans. var. 4) 216 (trans. var.5) Kwakkel et al; 201426

Dio3 D1 AACTCCGAGGTGGTTCTGC TTGCGCGTAGTCGAGGAT 60 Kwakkel et al; 201426

SL-C16A2 MCT8 CAACGCACTTACCGCATCTG GTAGCCCCAATACACACCAAGAG 146 Chan et al; 200625

SL-C16A10 MCT10 ATGCTGGAAACCTTCGGCTC TGAAGACGCTGACTATTGGGC 115

Harvard Primer Bank no. 221139821c1

THRA1 TRα1 CATCTTTGAACTGGGCAAGT CTGAGGCTTTAGACTTCCTGATC 348 Bakker; 200124

THRA2 TRα2 CATCTTTGAACTGGGCAAGT GACCCTGAACAACATGCATT 337 Bakker; 200124

THRB1 TRβ1 AAGTGCCCAGACCTTCCAAA AAAGAAACCCTTGCAGCCTTC 151 Silva et al;

200223

(8)

516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek Processed on: 16-5-2018 Processed on: 16-5-2018 Processed on: 16-5-2018

Processed on: 16-5-2018 PDF page: 40PDF page: 40PDF page: 40PDF page: 40

40

their immunogold labelling. Granules were scored as either negative, MPO or LF positive, D3 positive or double positive for D3 and either MPO or LF. A total of 1880 granules were scored in sections double stained for D3 and LF. Granules were only considered positive for LF if they contained at least 2 gold grains. In sections double stained for MPO and D3 a total 678 granules were scored. All scoring was performed by the same person.

Figure 3.1. D3 is present in human neutrophils. (A) Western Blot for D3 (antibody #676) and Beta Actin in whole

cell lysate of unstimulated human neutrophils. (B) Confocal microscopy images of freshly isolated human neutrophils using the differential interference contrast (DIC) setting. (C-D) Human neutrophils show intracellular D3 staining (shown in red; antibody #718 with Alexa Fluor 568-conjugated secondary antibody) in both the cytoplasm and small vesicle-like structures distributed throughout the cytoplasm and in close proximity to the nucleus. Cell nuclei are stained with DAPI (shown in blue). Scale bars are 25 µm (B; C) or 3 µm (D) in length. Stainings were repeated in at least 3 separate donors, representative images are shown.

Flow Cytometry

Neutrophils were incubated in ice cold FACS buffer (PBS with 5% heat-inactivated fetal calf serum (FCS) and 0,5% [wt/vol] sodium azide) with human FC block (eBioscience) for 20 minutes at 4°C. Cells were then either stained directly or fixed and permeabilized first using the eBioscience Intracellular Fixation & Permeabilization Buffer Set. Staining with primary antibodies was performed for 30 minutes at 4°C, followed by staining with secondary antibody for 30 minutes at 4°C. A total of 100,000 events was acquired using a BD FACS Canto II flow cytometer and data was analyzed using FlowJo software (v.10). Viable CD16+CD49- cells were selected for analysis of D3 staining.

516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek Processed on: 16-5-2018 Processed on: 16-5-2018 Processed on: 16-5-2018

Processed on: 16-5-2018 PDF page: 41PDF page: 41PDF page: 41PDF page: 41

41

3

Antibodies used were anti human APC-eFluor780-conjugated CD16 (1:100), anti-human PE-conjugated CD49d (1:100) (both eBioscience) and anti-D3 (#718; 1:250) with Alexa Fluor 647- conjugated donkey anti rabbit (1:250, Biolegend). Cell viability was determined using propidium iodide or fixable viability dye (eFluor 520-conjugated), both from eBioscience.

RNA isolation, cDNA synthesis and qualitative PCR

Neutrophils were lysed in Trizol-Reagent (Life Technologies) at 5x106 cells/ml. RNA was isolated using the Nucleospin RNA extraction kit (Machery Nagel). RNA yield was determined using the Nanodrop 2000 and quality was checked on the Bioanalyzer 2100 (Agilent Technologies). cDNA was synthesized with equal RNA input (150 ng) using AMV Reverse Transcriptase enzyme with oligo d(T) primers (Roche). As a control for genomic DNA contamination, a cDNA synthesis reaction without reverse transcriptase was included. Qualitative PCR was carried out using the Lightcycler 480 (Roche) and SensiFAST SYBR No-ROX (Bioline). Following amplification, PCR products were analyzed on DNA agarose gel and visualized on the ImageQuant LAS4000 (GE Healthcare). Published primer sequences were used for all genes (Bakker, 2001, Chan et al., 2006, Kwakkel et al., 2014, Kwakkel et al., 2006, Silva et al., 2002) except SLC16A10 for which primers were obtained from the Harvard Primer Bank (no. 221139821c1). Primer sequences and amplicon lengths are listed in Table 3.1.

Gene Protein Forward primer Reverse primer Amplicon length (bp) Source

Dio1 D3 AGCCACGACAACTGGATACC ACTCCCAAATGTTGCACCTC 160 Kwakkel et al; 200627

Dio2 D2 CCTCCTCGATGCCTACAAAC TCCTTCTGTACTGGAGACATGC

82 (trans. var. 1&2); 190 (trans. var. 3); 324 (trans. var. 4) 216 (trans. var.5) Kwakkel et al; 201426

Dio3 D1 AACTCCGAGGTGGTTCTGC TTGCGCGTAGTCGAGGAT 60 Kwakkel et al; 201426

SL-C16A2 MCT8 CAACGCACTTACCGCATCTG GTAGCCCCAATACACACCAAGAG 146 Chan et al; 200625

SL-C16A10 MCT10 ATGCTGGAAACCTTCGGCTC TGAAGACGCTGACTATTGGGC 115

Harvard Primer Bank no. 221139821c1

THRA1 TRα1 CATCTTTGAACTGGGCAAGT CTGAGGCTTTAGACTTCCTGATC 348 Bakker; 200124

THRA2 TRα2 CATCTTTGAACTGGGCAAGT GACCCTGAACAACATGCATT 337 Bakker; 200124

THRB1 TRβ1 AAGTGCCCAGACCTTCCAAA AAAGAAACCCTTGCAGCCTTC 151 Silva et al;

200223

(9)

516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek Processed on: 16-5-2018 Processed on: 16-5-2018 Processed on: 16-5-2018

Processed on: 16-5-2018 PDF page: 42PDF page: 42PDF page: 42PDF page: 42

42

Results

Type 3 deiodinase is present in the cytosol and in granules of human neutrophils

Whole cell lysates of unstimulated freshly isolated human neutrophils expressed D3 protein as shown by a clear band at 37kD (Figure 3.1A). Confocal microscopy was used to elucidate the subcellular location of D3 in unstimulated human neutrophils. Both diffuse cytoplasmic D3 staining and D3 staining in small intracellular vesicles was seen. D3 did not appear to be present on the plasma membrane (Figure 3.1

B-D). In unstimulated human neutrophils, flow cytometric analysis of D3 expression in unpermeabilized

cells showed fluorescence levels similar to control cells incubated without primary antibody, indicating no D3 surface expression (Figure 3.2A). Permeabilized neutrophils showed strong D3 expression (Figure

3.2A-B), confirming the intracellular location of D3 in these cells.

Figure 3.2. D3 expression measured by flow cytometry. D3 expression following cell surface

staining and intracellular staining in freshly isolated unstimulated human neutrophils measured by flow cytometry. (A) Representative histograms are shown. The dotted line represents the degree of background staining in unpermeabilized cells stained with only fluorescently labelled secondary antibody in the total cell population. The black line represents D3 expression in unpermeabilized live CD16+CD49- human neutrophils (antibody #718 with AlexaFluor 568-conjugated secondary antibody). The filled gray histogram represents D3 expression in fixed and permeabilized live CD16+CD49- human neutrophils. (B) Median fluorescence intensity for D3 staining in live CD16+CD49- human neutrophils after cell surface staining (Surface) and following fixation, permeabilization and intracellular staining (Intracellular). Total data is shown from two separate experiments with different donors.

D3 colocalizes with lactoferrin and myeloperoxidase in intracellular granules, but not with early endosomes

In order to determine the type of intracellular vesicles that stained positive for D3 we performed a series of double-stainings using confocal microscopy. D3 is known to be present in early endosomes in certain cell types (Baqui et al., 2003). Therefore we performed a double-staining for D3 and Early Endosome Antigen 1 (EEA1). The granules in which D3 is located were clearly different in size and amount compared to early endosomes and there was no colocalization between the two markers

(Figure 3.3). 516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek Processed on: 16-5-2018 Processed on: 16-5-2018 Processed on: 16-5-2018

Processed on: 16-5-2018 PDF page: 43PDF page: 43PDF page: 43PDF page: 43

43

3

Figure 3.3. Colocalization of D3 with granule markers. Confocal microscopy images of freshly isolated human

neutrophils show double immunostaining for D3 (shown in red; antibody #718 with Alexa Fluor 568-conjugated secondary antibody) with early endosome antigen 1 (EEA1) and markers for neutrophil granule subsets: myeloperoxidase (MPO; FITC-labelled), lactoferrin (LF), matrix metallopeptidase 9 (MMP9) and human serum albumin (HSA), all shown in green. All antibodies besides D3 and MPO were stained with an Alexa Fluor 647-conjugated secondary antibody. Enlarged images of areas indicated with a white box are shown in the right-hand column. Colocalization between D3 and other markers is visible as yellow and indicated with white arrows in the enlarged images. Red arrows indicate granules without colocalization. Scale bars are 3 µm in length. Stainings were repeated in at least 3 separate donors with similar results, representative images from 1 donor are shown.

Human neutrophils contain several subsets of granules with proteins that are critical for their bactericidal function. Upon activation of the cell these granules are mobilized sequentially, and fuse

(10)

516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek Processed on: 16-5-2018 Processed on: 16-5-2018 Processed on: 16-5-2018

Processed on: 16-5-2018 PDF page: 42PDF page: 42PDF page: 42PDF page: 42

42

Results

Type 3 deiodinase is present in the cytosol and in granules of human neutrophils

Whole cell lysates of unstimulated freshly isolated human neutrophils expressed D3 protein as shown by a clear band at 37kD (Figure 3.1A). Confocal microscopy was used to elucidate the subcellular location of D3 in unstimulated human neutrophils. Both diffuse cytoplasmic D3 staining and D3 staining in small intracellular vesicles was seen. D3 did not appear to be present on the plasma membrane (Figure 3.1

B-D). In unstimulated human neutrophils, flow cytometric analysis of D3 expression in unpermeabilized

cells showed fluorescence levels similar to control cells incubated without primary antibody, indicating no D3 surface expression (Figure 3.2A). Permeabilized neutrophils showed strong D3 expression (Figure

3.2A-B), confirming the intracellular location of D3 in these cells.

Figure 3.2. D3 expression measured by flow cytometry. D3 expression following cell surface

staining and intracellular staining in freshly isolated unstimulated human neutrophils measured by flow cytometry. (A) Representative histograms are shown. The dotted line represents the degree of background staining in unpermeabilized cells stained with only fluorescently labelled secondary antibody in the total cell population. The black line represents D3 expression in unpermeabilized live CD16+CD49- human neutrophils (antibody #718 with AlexaFluor 568-conjugated secondary antibody). The filled gray histogram represents D3 expression in fixed and permeabilized live CD16+CD49- human neutrophils. (B) Median fluorescence intensity for D3 staining in live CD16+CD49- human neutrophils after cell surface staining (Surface) and following fixation, permeabilization and intracellular staining (Intracellular). Total data is shown from two separate experiments with different donors.

D3 colocalizes with lactoferrin and myeloperoxidase in intracellular granules, but not with early endosomes

In order to determine the type of intracellular vesicles that stained positive for D3 we performed a series of double-stainings using confocal microscopy. D3 is known to be present in early endosomes in certain cell types (Baqui et al., 2003). Therefore we performed a double-staining for D3 and Early Endosome Antigen 1 (EEA1). The granules in which D3 is located were clearly different in size and amount compared to early endosomes and there was no colocalization between the two markers

(Figure 3.3). 516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek Processed on: 16-5-2018 Processed on: 16-5-2018 Processed on: 16-5-2018

Processed on: 16-5-2018 PDF page: 43PDF page: 43PDF page: 43PDF page: 43

43

3

Figure 3.3. Colocalization of D3 with granule markers. Confocal microscopy images of freshly isolated human

neutrophils show double immunostaining for D3 (shown in red; antibody #718 with Alexa Fluor 568-conjugated secondary antibody) with early endosome antigen 1 (EEA1) and markers for neutrophil granule subsets: myeloperoxidase (MPO; FITC-labelled), lactoferrin (LF), matrix metallopeptidase 9 (MMP9) and human serum albumin (HSA), all shown in green. All antibodies besides D3 and MPO were stained with an Alexa Fluor 647-conjugated secondary antibody. Enlarged images of areas indicated with a white box are shown in the right-hand column. Colocalization between D3 and other markers is visible as yellow and indicated with white arrows in the enlarged images. Red arrows indicate granules without colocalization. Scale bars are 3 µm in length. Stainings were repeated in at least 3 separate donors with similar results, representative images from 1 donor are shown.

Human neutrophils contain several subsets of granules with proteins that are critical for their bactericidal function. Upon activation of the cell these granules are mobilized sequentially, and fuse

(11)

516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek Processed on: 16-5-2018 Processed on: 16-5-2018 Processed on: 16-5-2018

Processed on: 16-5-2018 PDF page: 44PDF page: 44PDF page: 44PDF page: 44

44

with the phagosome or the exterior of the cell in a process known as degranulation (Borregaard and Cowland, 1997, Faurschou and Borregaard, 2003). We performed double-stainings for D3 and specific markers of the four neutrophil granule subsets (Figure 3.3) (Borregaard and Cowland, 1997, Faurschou and Borregaard, 2003). Myeloperoxidase (MPO), a marker of azurophilic granules (Cramer et al., 1985), colocalized with D3 in some but not all granules within the cell. D3 also colocalized with a subset of lactoferrin (LF) containing specific granules (Cramer et al., 1985), but not with all LF-positive granules. No colocalization was seen between D3 and matrix metallopeptidase 9 (MMP9; previously known as 92 kD gelatinase B/type IV collagenase), a marker for gelatinase granules (Kjeldsen et al., 1993). Finally, no colocalization was observed between D3 and human serum albumin (HSA), a marker for secretory vesicles (Borregaard et al., 1992).

Figure 3.4. Partial colocalization of D3 with lactoferrin and myelo-peroxidase containing granules.

(A) Electron microscopy images of unstimulated human neutrophils with immunogold labeling for D3 (antibody #718; 15 nm gold particles) and lactoferrin (LF; 10 nm gold particles). Gold particles are visible as black dots. LF is located in intracellular granules. D3 is located in the cytoplasm (black circle) or at the limiting membrane of intracellular granules (white circle). Granules containing only D3 (white *), only LF (black *), or both D3 and LF (black encircled *) are indicated. No D3 staining was observed at the plasma membrane (white arrowheads). The scale bar is 200 nm in length. (B) Electron microscopy images of unstimulated human neutrophils stained for D3 (10 nm gold particles) and myeloperoxidase (MPO; 15 nm gold particles). D3 staining was observed in the cytoplasm (black circles) and associated with granules (white circles). Granules containing only D3 (white *), only MPO (white †) or both D3 and MPO (white encircled *) are indicated. Very sporadic D3 staining (dashed black circles) was observed at the plasma membrane (white arrowheads). The nucleus (N) is also indicated. The scale bar is 200 nm in length. (C-D) Colocalization of D3 with MPO and LF was quantified by scoring granules based on their immunogold labelling in electron microscopy sections. A total of 1880 granules were scored in sections double stained for D3 and LF (C). Granules were only considered positive for LF if they contained at least 2 gold grains. In sections double stained for MPO and D3 a total 678 granules were scored (D). The results are expressed as a percentage of the total amount of granules.

516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek Processed on: 16-5-2018 Processed on: 16-5-2018 Processed on: 16-5-2018

Processed on: 16-5-2018 PDF page: 45PDF page: 45PDF page: 45PDF page: 45

45

3

To confirm the colocalization of D3 with MPO and LF and to assess in further detail the subcellular location of D3 in human neutrophils we performed electron microscopy and immunogold labelling for these markers (Figure 3.4). Electron microscopy confirmed the presence of D3 in both the cytoplasm of human neutrophils and intracellular granules. Within these intracellular granules D3 was mainly found at the outer limiting membrane. Only very limited D3 staining was observed at the plasma membrane. In accordance with the confocal microscopy results, D3 was observed in both a subset of LF containing granules and a subset of MPO containing granules (Figure 3.4 A-B). Colocalization was quantified by scoring granules based on the presence of D3 and MPO and D3 and LF in double stained sections (Figure 3.4 C-D). This revealed that approximately 15-20% of all granules scored contained D3. For both double-stainings a minority of D3 positive granules also contained the other marker, either LF or MPO.

Subcellular location of D3 does not appear to change immediately following neutrophil activation

After determining the subcellular location of D3 in resting human neutrophils we assessed whether this localization changed after activation of the cells. Neutrophils were incubated with PMA, a pharmacological agent that induces neutrophil activation, or zymosan, yeast particles that induce phagocytosis. Subcellular D3 location was assessed using electron and confocal microscopy. Neutrophils stimulated with PMA showed a pattern of D3 staining similar to that seen in resting cells, following both short term (15 mins) and prolonged (1 hour) stimulation (Figure 3.5 A; Figure 3.6 A). Short term PMA stimulation already resulted in strong activation of the cells (Supplemental Figure 3.2). D3 was observed throughout the cytoplasm and at the limiting membrane of intracellular granules (Figure

3.5 Aii-iii; Figure 3.6 A). Electron microscopy revealed D3 in close proximity to the plasma membrane,

however it did not appear to be located at the plasma membrane itself (Figure 3.5 A). Incubation of human neutrophils with zymosan resulted in phagocytosis of these particles which can be clearly seen in phagosomes within the cell (Figure 3.5 B). D3 was again present throughout the cytoplasm and in intracellular granules. The percentage of D3 positive granules was quantified but did not change after PMA or zymosan stimulation (data not shown). No D3 was observed in the phagosomes or phagosomal membrane. Activation of neutrophils using either zymosan or PMA did not clearly alter the intracellular location of D3.

D3 remains present in intracellular granules during prolonged neutrophil stimulation and early NETosis

Upon prolonged stimulation neutrophils can form neutrophil extracellular traps (NETs) that consist of a web-like structure of chromatin and bactericidal proteins normally present in granules (Brinkmann et al., 2004). These NETs can then trap and kill extracellular pathogens. NETosis is highly regulated and eventually results in the death of the neutrophil. We stimulated neutrophils with high doses of PMA which resulted in the formation of NETs (Figure 3.6 B-C). In early stage NETosis, characterized by the flattening of the nucleus while still preserving the integrity of the nuclear envelope and the granular membranes (Brinkmann and Zychlinsky, 2007), D3 is still observed in intracellular granules (Figure

(12)

516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek Processed on: 16-5-2018 Processed on: 16-5-2018 Processed on: 16-5-2018

Processed on: 16-5-2018 PDF page: 44PDF page: 44PDF page: 44PDF page: 44

44

with the phagosome or the exterior of the cell in a process known as degranulation (Borregaard and Cowland, 1997, Faurschou and Borregaard, 2003). We performed double-stainings for D3 and specific markers of the four neutrophil granule subsets (Figure 3.3) (Borregaard and Cowland, 1997, Faurschou and Borregaard, 2003). Myeloperoxidase (MPO), a marker of azurophilic granules (Cramer et al., 1985), colocalized with D3 in some but not all granules within the cell. D3 also colocalized with a subset of lactoferrin (LF) containing specific granules (Cramer et al., 1985), but not with all LF-positive granules. No colocalization was seen between D3 and matrix metallopeptidase 9 (MMP9; previously known as 92 kD gelatinase B/type IV collagenase), a marker for gelatinase granules (Kjeldsen et al., 1993). Finally, no colocalization was observed between D3 and human serum albumin (HSA), a marker for secretory vesicles (Borregaard et al., 1992).

Figure 3.4. Partial colocalization of D3 with lactoferrin and myelo-peroxidase containing granules.

(A) Electron microscopy images of unstimulated human neutrophils with immunogold labeling for D3 (antibody #718; 15 nm gold particles) and lactoferrin (LF; 10 nm gold particles). Gold particles are visible as black dots. LF is located in intracellular granules. D3 is located in the cytoplasm (black circle) or at the limiting membrane of intracellular granules (white circle). Granules containing only D3 (white *), only LF (black *), or both D3 and LF (black encircled *) are indicated. No D3 staining was observed at the plasma membrane (white arrowheads). The scale bar is 200 nm in length. (B) Electron microscopy images of unstimulated human neutrophils stained for D3 (10 nm gold particles) and myeloperoxidase (MPO; 15 nm gold particles). D3 staining was observed in the cytoplasm (black circles) and associated with granules (white circles). Granules containing only D3 (white *), only MPO (white †) or both D3 and MPO (white encircled *) are indicated. Very sporadic D3 staining (dashed black circles) was observed at the plasma membrane (white arrowheads). The nucleus (N) is also indicated. The scale bar is 200 nm in length. (C-D) Colocalization of D3 with MPO and LF was quantified by scoring granules based on their immunogold labelling in electron microscopy sections. A total of 1880 granules were scored in sections double stained for D3 and LF (C). Granules were only considered positive for LF if they contained at least 2 gold grains. In sections double stained for MPO and D3 a total 678 granules were scored (D). The results are expressed as a percentage of the total amount of granules.

516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek Processed on: 16-5-2018 Processed on: 16-5-2018 Processed on: 16-5-2018

Processed on: 16-5-2018 PDF page: 45PDF page: 45PDF page: 45PDF page: 45

45

3

To confirm the colocalization of D3 with MPO and LF and to assess in further detail the subcellular location of D3 in human neutrophils we performed electron microscopy and immunogold labelling for these markers (Figure 3.4). Electron microscopy confirmed the presence of D3 in both the cytoplasm of human neutrophils and intracellular granules. Within these intracellular granules D3 was mainly found at the outer limiting membrane. Only very limited D3 staining was observed at the plasma membrane. In accordance with the confocal microscopy results, D3 was observed in both a subset of LF containing granules and a subset of MPO containing granules (Figure 3.4 A-B). Colocalization was quantified by scoring granules based on the presence of D3 and MPO and D3 and LF in double stained sections (Figure 3.4 C-D). This revealed that approximately 15-20% of all granules scored contained D3. For both double-stainings a minority of D3 positive granules also contained the other marker, either LF or MPO.

Subcellular location of D3 does not appear to change immediately following neutrophil activation

After determining the subcellular location of D3 in resting human neutrophils we assessed whether this localization changed after activation of the cells. Neutrophils were incubated with PMA, a pharmacological agent that induces neutrophil activation, or zymosan, yeast particles that induce phagocytosis. Subcellular D3 location was assessed using electron and confocal microscopy. Neutrophils stimulated with PMA showed a pattern of D3 staining similar to that seen in resting cells, following both short term (15 mins) and prolonged (1 hour) stimulation (Figure 3.5 A; Figure 3.6 A). Short term PMA stimulation already resulted in strong activation of the cells (Supplemental Figure 3.2). D3 was observed throughout the cytoplasm and at the limiting membrane of intracellular granules (Figure

3.5 Aii-iii; Figure 3.6 A). Electron microscopy revealed D3 in close proximity to the plasma membrane,

however it did not appear to be located at the plasma membrane itself (Figure 3.5 A). Incubation of human neutrophils with zymosan resulted in phagocytosis of these particles which can be clearly seen in phagosomes within the cell (Figure 3.5 B). D3 was again present throughout the cytoplasm and in intracellular granules. The percentage of D3 positive granules was quantified but did not change after PMA or zymosan stimulation (data not shown). No D3 was observed in the phagosomes or phagosomal membrane. Activation of neutrophils using either zymosan or PMA did not clearly alter the intracellular location of D3.

D3 remains present in intracellular granules during prolonged neutrophil stimulation and early NETosis

Upon prolonged stimulation neutrophils can form neutrophil extracellular traps (NETs) that consist of a web-like structure of chromatin and bactericidal proteins normally present in granules (Brinkmann et al., 2004). These NETs can then trap and kill extracellular pathogens. NETosis is highly regulated and eventually results in the death of the neutrophil. We stimulated neutrophils with high doses of PMA which resulted in the formation of NETs (Figure 3.6 B-C). In early stage NETosis, characterized by the flattening of the nucleus while still preserving the integrity of the nuclear envelope and the granular membranes (Brinkmann and Zychlinsky, 2007), D3 is still observed in intracellular granules (Figure

(13)

516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek Processed on: 16-5-2018 Processed on: 16-5-2018 Processed on: 16-5-2018

Processed on: 16-5-2018 PDF page: 46PDF page: 46PDF page: 46PDF page: 46

46

3.6 Cii). D3 is also still observed intracellularly in a later stage of NETosis, where membrane integrity

is compromised and the nuclear contents mix with granule proteins (Figure 3.6 Ci) (Brinkmann and Zychlinsky, 2007). In the final stage of NETosis in which the cells release their NETs, D3 can occasionally be found on the NETs themselves but in general less D3 is observed, suggesting that it has been released into the extracellular space (Figure 3.6 Ciii). During the formation of NETs, the location of D3 follows a similar pattern to that of other granule proteins, remaining present in the cell during early stage NETosis before finally being released into the extracellular space.

Figure 3.5. The subcellular location of D3 following neutrophil stimulation. (A) Electron

microscopy images of human neutrophils stimulated with PMA (20ng/ml) for 15 minutes. Sections were stained for D3 (antibody #718; 10 nm gold particles). The scale bar is 500 nm in length. In the enlarged images (Aii-iii) granules containing D3 are indicated (white *). D3 staining was also observed in the cytoplasm, as intermittently indicated (black arrowheads). The nucleus is also indicated (N). (B) Electron microscopy images of human neutrophils stimulated with serum opsonized zymosan for 15 minutes and stained for D3 (antibody #718; 15 nm gold particles). Phagocytosed zymosan particles are indicated by a Z. The nucleus is also indicated (N). The scale bar is 1 µm in length. The enlarged areas (Bii-iii) show granules that contain D3 (white *) and granules with no D3 staining. Cytoplasmic D3 staining is intermittently indicated with black arrowheads. 516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek Processed on: 16-5-2018 Processed on: 16-5-2018 Processed on: 16-5-2018

Processed on: 16-5-2018 PDF page: 47PDF page: 47PDF page: 47PDF page: 47

47

3

Figure 3.6. D3 is observed intracellularly during the formation of neutrophil extracellular traps. Confocal microscopy images of PMA-stimulated and unstimulated human neutrophils. (A) Neutrophils stimulated with PMA (100ng/ml) for 1 hour were stained for D3 (Aii-iii). Unstimulated control samples were cultured and stained in parallel (Ai). D3 is shown in red; antibody #718 with Alexa Fluor 568-conjugated secondary antibody. No significant changes in the pattern of D3 staining were observed. (B-C) Neutrophils were stimulated with 600 nM PMA until the formation of neutrophil extracellular traps (NETs), 2-3 hours. (B) After 3 hours unstimulated control samples show no signs of NETosis (Bi; Biii). In contrast almost all PMA-stimulated cells have undergone some degree of NETosis (Bii; Biv), characterized by loss of membrane integrity (Bii) and release of DNA, stained here by DAPI, into the extracellular space (Biv). (C) Neutrophils forming NETs were stained for D3 (red), MPO (green) and DAPI (blue). D3 is present intracellularly in early stage NETosis (white arrowheads) which is characterized by loss of nuclear lobular shape but preserved membrane integrity. After disintegration of the nuclear membrane, a later stage of NETosis (Ci and red arrowhead), D3 is still observed within the cell. A projected z-stack of a fully formed NET (Ciii; white arrows) shows that once released into the extracellular space, only intermittent D3 staining is observed. Stainings were repeated in two separate donors, representative images are shown. All scale bars are 15 µm in length except image Aii (20 µm) and Ci (5 µm).

(14)

516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek Processed on: 16-5-2018 Processed on: 16-5-2018 Processed on: 16-5-2018

Processed on: 16-5-2018 PDF page: 46PDF page: 46PDF page: 46PDF page: 46

46

3.6 Cii). D3 is also still observed intracellularly in a later stage of NETosis, where membrane integrity

is compromised and the nuclear contents mix with granule proteins (Figure 3.6 Ci) (Brinkmann and Zychlinsky, 2007). In the final stage of NETosis in which the cells release their NETs, D3 can occasionally be found on the NETs themselves but in general less D3 is observed, suggesting that it has been released into the extracellular space (Figure 3.6 Ciii). During the formation of NETs, the location of D3 follows a similar pattern to that of other granule proteins, remaining present in the cell during early stage NETosis before finally being released into the extracellular space.

Figure 3.5. The subcellular location of D3 following neutrophil stimulation. (A) Electron

microscopy images of human neutrophils stimulated with PMA (20ng/ml) for 15 minutes. Sections were stained for D3 (antibody #718; 10 nm gold particles). The scale bar is 500 nm in length. In the enlarged images (Aii-iii) granules containing D3 are indicated (white *). D3 staining was also observed in the cytoplasm, as intermittently indicated (black arrowheads). The nucleus is also indicated (N). (B) Electron microscopy images of human neutrophils stimulated with serum opsonized zymosan for 15 minutes and stained for D3 (antibody #718; 15 nm gold particles). Phagocytosed zymosan particles are indicated by a Z. The nucleus is also indicated (N). The scale bar is 1 µm in length. The enlarged areas (Bii-iii) show granules that contain D3 (white *) and granules with no D3 staining. Cytoplasmic D3 staining is intermittently indicated with black arrowheads. 516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek Processed on: 16-5-2018 Processed on: 16-5-2018 Processed on: 16-5-2018

Processed on: 16-5-2018 PDF page: 47PDF page: 47PDF page: 47PDF page: 47

47

3

Figure 3.6. D3 is observed intracellularly during the formation of neutrophil extracellular traps. Confocal microscopy images of PMA-stimulated and unstimulated human neutrophils. (A) Neutrophils stimulated with PMA (100ng/ml) for 1 hour were stained for D3 (Aii-iii). Unstimulated control samples were cultured and stained in parallel (Ai). D3 is shown in red; antibody #718 with Alexa Fluor 568-conjugated secondary antibody. No significant changes in the pattern of D3 staining were observed. (B-C) Neutrophils were stimulated with 600 nM PMA until the formation of neutrophil extracellular traps (NETs), 2-3 hours. (B) After 3 hours unstimulated control samples show no signs of NETosis (Bi; Biii). In contrast almost all PMA-stimulated cells have undergone some degree of NETosis (Bii; Biv), characterized by loss of membrane integrity (Bii) and release of DNA, stained here by DAPI, into the extracellular space (Biv). (C) Neutrophils forming NETs were stained for D3 (red), MPO (green) and DAPI (blue). D3 is present intracellularly in early stage NETosis (white arrowheads) which is characterized by loss of nuclear lobular shape but preserved membrane integrity. After disintegration of the nuclear membrane, a later stage of NETosis (Ci and red arrowhead), D3 is still observed within the cell. A projected z-stack of a fully formed NET (Ciii; white arrows) shows that once released into the extracellular space, only intermittent D3 staining is observed. Stainings were repeated in two separate donors, representative images are shown. All scale bars are 15 µm in length except image Aii (20 µm) and Ci (5 µm).

(15)

516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek Processed on: 16-5-2018 Processed on: 16-5-2018 Processed on: 16-5-2018

Processed on: 16-5-2018 PDF page: 48PDF page: 48PDF page: 48PDF page: 48

48

Human neutrophils express other elements of thyroid hormone metabolism

Human neutrophils expressed type 1 deiodinase (Dio1), type 3 deiodinase (Dio3), thyroid hormone (TH) monocarboxylate transporter 10 (SLC16A10) and TH receptor α1 (THRA1) mRNA. (Figure 3.7). By contrast, deiodinase type 2 (Dio2), TH monocarboxylate transporter 8 (SLC16A2), TH receptor α2 (THRA2) or TH receptor β1 (THRB1) mRNA expression was not observed (data not shown). This indicates that, besides D3, human neutrophils express a number of important genes that are essential for intracellular TH metabolism and action.

Figure 3.7. Expression of genes related to thyroid hormone metabolism. qPCR was performed on

cDNA synthesized with total RNA extracted from unstimulated human neutrophils for a number of genes related to intracellular thyroid hormone metabolism. PCR products run on agarose gel are shown. Human neutrophils were shown to express deiodinase type 1 (D1), D3, thyroid hormone transporter MCT10 and thyroid hormone receptor α1 at the transcriptional level (see Table 3.1 for primer sequences and amplicon length). Results are shown for four different donors for each gene.

Discussion

Thyroid hormones are essential for key biological processes such as growth, development and the regulation of energy metabolism (Mullur et al., 2014, Tata, 1968, Bianco and Kim, 2006). The amount of biologically active TH in tissues and cells is tightly regulated by the deiodinase enzyme family. These enzymes can activate or inactivate T3 and T4, thereby affecting the availability of intracellular TH (Bianco and Kim, 2006). Deiodinating enzymes are thought to play an important role in the function of various innate immune cells. Type 2 deiodinase is expressed in macrophages and involved in the inflammatory response of these cells (Kwakkel et al., 2014). D3, the TH inactivating enzyme is strongly expressed in infiltrating murine neutrophils during both bacterial infection and sterile inflammation (Boelen et al., 2008, Boelen et al., 2005). Pronounced D3 activity is found in a turpentine induced abscess containing large amounts of neutrophils (Boelen et al., 2005). Furthermore, mice that lack D3 have impaired bacterial clearance upon infection (Boelen et al., 2009). Therefore D3 is thought to play an important role in the bacterial killing capacity of neutrophils (Boelen et al., 2009, Fliers et al., 2015, Fliers et al., 2014). 516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek 516644-L-bw-spek Processed on: 16-5-2018 Processed on: 16-5-2018 Processed on: 16-5-2018

Processed on: 16-5-2018 PDF page: 49PDF page: 49PDF page: 49PDF page: 49

49

3

Here we demonstrate the presence of D3 in human neutrophils for the first time (Figure 3.1-3.2). D3 is located in both the cytoplasm and in the limiting membrane of a subset of both azurophilic and specific granules, which contain myeloperoxidase (MPO) and lactoferrin (LF) respectively (Figure 3.3-3.4). In contrast to what has been reported in other cell types, we observed very limited D3 staining at the plasma membrane and no D3 in the nucleus or early endosomes of human neutrophils (Baqui et al., 2003, Olvera et al., 2015). However, more recent papers demonstrate that the subcellular localization of D3 could be cell type specific as the location of D3 in neurons also differs from that described in other cell types (Jo et al., 2012, Kallo et al., 2012, Alkemade et al., 2005).

D3 has previously been described to translocate to the nucleus following hypoxia in neurons (Jo et al., 2012). We did not observe an acute change in the subcellular location of D3 following activation of neutrophils using either a chemical or a particulate stimulus (Figure 3.5; Figure 3.6A). This could be due to the fact that this translocation is time dependent and was only observed in the neuronal model after at least 16 hours of hypoxia (Jo et al., 2012). The duration of stimulation used here is much shorter because neutrophils respond extremely quickly to stimuli (Supplemental Figure 3.2) and are also significantly shorter lived cells. Longer stimulation of neutrophils results in the formation of neutrophil extracellular traps (NETs). The subcellular location of D3 during NETosis behaves in a very similar manner to that described for other granule proteins, with D3 remaining present in intracellular granular structures until membrane integrity is dissolved and the NETs are released at which point D3 staining reduces significantly indicating its release into the extracellular space (Figure 3.6 B-C) (Brinkmann et al., 2010, Brinkmann et al., 2004, Brinkmann and Zychlinsky, 2007).

Type 3 deiodinase is a type 1 transmembrane protein meaning that its C-terminus, containing the active center of the enzyme, is located in the cytosol (Chou and Elrod, 1999, Jo et al., 2012, Kallo et al., 2012, Schweizer et al., 2014). The D3 antibody used for confocal and electron microscopy was raised against a peptide located in the C-terminus of the enzyme (Huang et al., 2003). Thus the staining of D3 in the limiting membrane of intracellular granules would suggest that the active site of the enzyme is on the cytosolic side of the granules, giving it easy access to its substrate. Previous studies have also described a cytosolic location for D3 in neurons (Alkemade et al., 2005, Jo et al., 2012).

The subcellular location of D3 in human neutrophils could provide important clues about its function in these cells. D3 was hypothesized to play a role in the bacterial killing capacity of neutrophils but the mechanism behind this effect has not yet been clarified (Boelen et al., 2011). Neutrophil bacterial killing can be mediated by a number of different mechanisms.

The 3 main killing mechanisms are 1) antibacterial proteins that can be released into the phagosome or extracellular space, 2) generation of reactive oxygen species (ROS) used to kill phagocytosed microorganisms and 3) generation of neutrophil extracellular traps (NETs) that immobilize and kill extracellular pathogens (Kolaczkowska and Kubes, 2013). Based on the subcellular location there are several potential models for the role of D3 in these cells.

Referenties

GERELATEERDE DOCUMENTEN

Maar daarnaast ben je er altijd voor mij en onze twee kleintjes, heb je er altijd het volste vertrouwen in gehad dat ik kon wat ik kan en heb je me over heel wat dalen

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

The sublime as it has been theorized since the early seventeenth century appears here as a most fruitful concept for addressing the unsettling or overwhelming effect

Staurastrum brachiatum Ralfs and Staurodesmus cuspidatus (Ralfs) Teil. West) Teil., species confined to acidic, soft water habitats, showed remarkably different behavior in the pH

Moreover, a subsequently performed region of interest (ROI) analysis confirms our prediction that context modulates the more basic decision-making processes: If participants decided

Activation of the NF- κ B/Rel transcription family, by nuclear translocation of cytoplasmic complexes, plays a central role in inflammation through its ability to induce

We show that the atom-molecule mixture formed in a degenerate atomic Fermi gas with interspecies repul- sion near a Feshbach resonance constitutes a peculiar system where the

ARP-modified electrodes were used for the detection of not only hydrogen peroxide but also organic peroxides, Le., cumol hydroperoxide, 2-butanone peroxide,