Zurich Open Repository and Archive University of Zurich Main Library Strickhofstrasse 39 CH-8057 Zurich

Similar documents
Supporting Information

Blocking antibodies and peptides. Rat anti-mouse PD-1 (29F.1A12, rat IgG2a, k), PD-

Alternatively Activated Macrophages Determine the Repair of the Infarcted

The toll-like receptor 4 ligands Mrp8 and Mrp14 play a critical role in the development of autoreactive CD8 + T cells

Interferon γ regulates idiopathic pneumonia syndrome, a. Th17 + CD4 + T-cell-mediated GvH disease

MATERIALS AND METHODS. Neutralizing antibodies specific to mouse Dll1, Dll4, J1 and J2 were prepared as described. 1,2 All

Supplemental Table 1. Primer sequences for transcript analysis

Supplementary Figures

Eosinophils are required. for the maintenance of plasma cells in the bone marrow

c Ischemia (30 min) Reperfusion (8 w) Supplementary Figure bp 300 bp Ischemia (30 min) Reperfusion (4 h) Dox 20 mg/kg i.p.

SUPPLEMENTARY METHODS

Supplementary Figure 1. Characterization of basophils after reconstitution of SCID mice

Suppl Video: Tumor cells (green) and monocytes (white) are seeded on a confluent endothelial

Supplementary information. The proton-sensing G protein-coupled receptor T-cell death-associated gene 8

Postn MCM Smad2 fl/fl Postn MCM Smad3 fl/fl Postn MCM Smad2/3 fl/fl. Postn MCM. Tgfbr1/2 fl/fl TAC

Role of Tyk-2 in Th9 and Th17 cells in allergic asthma

Supplemental Figure 1. Signature gene expression in in vitro differentiated Th0, Th1, Th2, Th17 and Treg cells. (A) Naïve CD4 + T cells were cultured

Targeting tumour associated macrophages in anti-cancer therapies. Annamaria Gal Seminar Series on Drug Discovery Budapest 5 January 2018

Pretargeting and Bioorthogonal Click Chemistry-Mediated Endogenous Stem Cell Homing for Heart Repair

SUPPLEMENTARY INFORMATION

Supplemental figure 1. PDGFRα is expressed dominantly by stromal cells surrounding mammary ducts and alveoli. A) IHC staining of PDGFRα in

Supplementary Figure 1

Cardiac myosin-th17 responses promote heart failure in human myocarditis

Supplemental Information. T Cells Enhance Autoimmunity by Restraining Regulatory T Cell Responses via an Interleukin-23-Dependent Mechanism

Supplemental Materials for. Effects of sphingosine-1-phosphate receptor 1 phosphorylation in response to. FTY720 during neuroinflammation

Supporting Information

Supplemental Figure 1

Supplementary Materials for

Combined Rho-kinase inhibition and immunogenic cell death triggers and propagates immunity against cancer

Cell isolation. Spleen and lymph nodes (axillary, inguinal) were removed from mice

SUPPLEMENTARY INFORMATION

Supplementary Fig. 1 p38 MAPK negatively regulates DC differentiation. (a) Western blot analysis of p38 isoform expression in BM cells, immature DCs

sequences of a styx mutant reveals a T to A transversion in the donor splice site of intron 5

Supplementary Information. Tissue-wide immunity against Leishmania. through collective production of nitric oxide

Cardiac myosin-th17 responses promote heart failure in human myocarditis

Intracellular MHC class II molecules promote TLR-triggered innate. immune responses by maintaining Btk activation

hemodynamic stress. A. Echocardiographic quantification of cardiac dimensions and function in

B6/COLODR/SPL/11C/83/LAP/#2.006 B6/COLODR/SPL/11C/86/LAP/#2.016 CD11C B6/COLODR/SPL/11C/80/LAP/#2.011 CD11C

effect on the upregulation of these cell surface markers. The mean peak fluorescence intensity

Supporting Information. Calculation of the relative contributions of myocyte proliferation, stem cell. Supporting Information Fig 1 (page 9)

Supporting Online Material for

Supplementary Figure 1. Efficient DC depletion in CD11c.DOG transgenic mice

% of live splenocytes. STAT5 deletion. (open shapes) % ROSA + % floxed

D CD8 T cell number (x10 6 )

Trim29 gene-targeting strategy. (a) Genotyping of wildtype mice (+/+), Trim29 heterozygous mice (+/ ) and homozygous mice ( / ).

SUPPLEMENTARY INFORMATION

SUPPLEMENTARY MATERIALS. IL-4 as a Repurposed Biological Drug for Myocardial Infarction through. Augmentation of Reparative Cardiac Macrophages:

Supplementary Figure 1. H-PGDS deficiency does not affect GI tract functions and anaphylactic reaction. (a) Representative pictures of H&E-stained

Supplementary Materials for

International Graduate Research Programme in Cardiovascular Science

Supplementary Figure S1. Flow cytometric analysis of the expression of Thy1 in NH cells. Flow cytometric analysis of the expression of T1/ST2 and

Cover Page. The handle holds various files of this Leiden University dissertation.

Nature Immunology: doi: /ni Supplementary Figure 1. Huwe1 has high expression in HSCs and is necessary for quiescence.

Serum cytokine levels in control and tumor-bearing male and female mice at day 15.

Role of Inflammatory and Progenitor Cells in Pulmonary Vascular Remodeling: Potential Role for Targeted Therapies. Traditional Hypothesis Stress

Inflammatory dilated cardiomyopathy refers to an end-stage. Heart Failure

The encephalitogenicity of TH17 cells is dependent on IL-1- and IL-23- induced production of the cytokine GM-CSF

well for 2 h at rt. Each dot represents an individual mouse and bar is the mean ±

VEGFR2-Mediated Vascular Dilation as a Mechanism of VEGF-Induced Anemia and Bone Marrow Cell Mobilization

Hua Tang, Weiping Cao, Sudhir Pai Kasturi, Rajesh Ravindran, Helder I Nakaya, Kousik

NK cell flow cytometric assay In vivo DC viability and migration assay

Supplementary Figure 1.

Online supplement. Phenotypic, functional and plasticity features of classical and alternatively activated

Supplemental Table I.

CD14 + S100A9 + Monocytic Myeloid-Derived Suppressor Cells and Their Clinical Relevance in Non-Small Cell Lung Cancer

Supplemental Information. Tissue Myeloid Progenitors Differentiate. into Pericytes through TGF-b Signaling. in Developing Skin Vasculature

Nature Medicine: doi: /nm.3922

Canberra, Australia). CD11c-DTR-OVA-GFP (B6.CD11c-OVA), B6.luc + and. Cancer Research Center, Germany). B6 or BALB/c.FoxP3-DTR-GFP mice were

FOR OPTIMAL GUT HEALTH KEMIN.COM/GUTHEALTH

Fig 1 CD163. CD11b S100A9. Sirius Red. 100μm ** ** CD163. CD11b S100A9 ** Sirius Red (PL) Sirius Red SUM Mo.

Chronic variable stress activates hematopoietic stem cells

Supplementary Figure S1. PTPN2 levels are not altered in proliferating CD8+ T cells. Lymph node (LN) CD8+ T cells from C57BL/6 mice were stained with

Supplementary Data Table of Contents:

Supplementary Figure 1. mrna expression of chitinase and chitinase-like protein in splenic immune cells. Each splenic immune cell population was

SUPPLEMENTARY INFORMATION

Supplementary Figure 1 Chemokine and chemokine receptor expression during muscle regeneration (a) Analysis of CR3CR1 mrna expression by real time-pcr

Nature Medicine: doi: /nm.2109

Uncovering the mechanisms of wound healing and fibrosis

Gene expression was analyzed by real time PCR (SYBR GREEN PCR Master. Mix, Roche Applied Science) using specific oligonucleotides.

BMDCs were generated in vitro from bone marrow cells cultured in 10 % RPMI supplemented

An epithelial-to-mesenchymal transition-inducing potential of. granulocyte macrophage colony-stimulating factor in colon. cancer

Evaluation of directed and random motility in microslides Assessment of leukocyte adhesion in flow chambers

Detailed step-by-step operating procedures for NK cell and CTL degranulation assays

Supplementary Figure 1. HOPX is hypermethylated in NPC. (a) Methylation levels of HOPX in Normal (n = 24) and NPC (n = 24) tissues from the

Role of Inflammation in Pulmonary Hypertension

C57BL/6 Mice are More Appropriate. than BALB/C Mice in Inducing Dilated Cardiomyopathy with Short-Term Doxorubicin Treatment

Supplementary Figure 1. Expression of CUGBP1 in non-parenchymal liver cells treated with TGF-β

Liver-Resident Macrophage Necroptosis Orchestrates Type 1 Microbicidal Inflammation and Type-2- Mediated Tissue Repair during Bacterial Infection

Supplementary Information:

Supplemental Information. CD4 + CD25 + Foxp3 + Regulatory T Cells Promote. Th17 Cells In Vitro and Enhance Host Resistance

CD25-PE (BD Biosciences) and labeled with anti-pe-microbeads (Miltenyi Biotec) for depletion of CD25 +

CYTOKINES. Based on: Cellular and Molecular Immunology, 4 th ed.,abbas A.K., Lichtman A.H. and Pober J.S. Sounders company; Philadelphia, 2010.

SUPPLEMENTARY FIGURES

As outlined under External contributions (see appendix 7.1), the group of Prof. Gröne at the

Supporting Information

Figure S1. PMVs from THP-1 cells expose phosphatidylserine and carry actin. A) Flow

Pearson r = P (one-tailed) = n = 9

Journal Club Semmler Lorenz

Optimizing Intracellular Flow Cytometry:

SUPPLEMENTARY INFORMATION

Transcription:

Zurich Open Repository and Archive University of Zurich Main Library Strickhofstrasse 39 CH-8057 Zurich www.zora.uzh.ch Year: 2013 Nitric oxide synthase 2 is required for conversion of pro-fibrogenic inflammatory CD133+ progenitors into F4/80+ macrophages in experimental autoimmune myocarditis Blyszczuk, Przemyslaw; Berthonneche, Corrine; Behnke, Silvia; Glönkler, Marcel; Moch, Holger; Pedrazzini, Thierry; Lüscher, Thomas F; Eriksson, Urs; Kania, Gabriela Abstract: AIMS: Experimental Autoimmune Myocarditis (EAM) model mirrors important mechanisms of inflammatory dilated cardiomyopathy (idcm). In EAM, inflammatory CD133(+) progenitors are a major cellular source of cardiac myofibroblasts in post-inflammatory myocardium. We hypothesized that exogenous delivery of Macrophage-Colony Stimulating Factor (M-CSF) can stimulate macrophage lineage differentiation of inflammatory progenitors and therefore prevent their naturally occurring myofibroblast fate in EAM.Methods and ResultsEAM was induced in wild-type (BALB/c) and Nitric oxide synthase 2 deficient (Nos2(-/-)) mice and CD133(+) progenitors were isolated from inflamed hearts. In vitro, M-CSF converted inflammatory CD133(+) progenitors into nitric oxide-producing F4/80(+) macrophages and prevented TGF--mediated myofibroblast differentiation. Importantly, only a subset of heart-infiltrating CD133(+) progenitors expresses macrophage-specific antigen F4/80 in EAM. These CD133(+)/F4/80(hi) cells show impaired myofibrogenic potential compared to CD133(+)/F4/80(-) cells. M-CSF treatment of wild-type mice with EAM at the peak of disease markedly increased CD133(+)/F4/80(hi) cells in the myocardium, and CD133(+) progenitors isolated from M-CSF-treated mice failed to differentiate into myofibroblasts. In contrast, M-CSF was not effective in converting CD133(+) progenitors from inflamed hearts of Nos2(-/-) mice into macrophages, and M-CSF treatment did not result in increased CD133(+)/F4/80(hi) cell population in hearts of Nos2(-/-) mice. Accordingly, M-CSF prevented postinflammatory fibrosis and left ventricular dysfunction in wild-type but not in Nos2(-/-) mice. CON- CLUSIONS: Active and NOS2-dependent induction of macrophage lineage differentiation abrogates the myofibrogenic potential of heart-infiltrating CD133(+) progenitors. Modulating the in vivo differentiation fate of specific progenitors might become a novel approach for the treatment of inflammatory heart diseases. DOI: https://doi.org/10.1093/cvr/cvs317 Posted at the Zurich Open Repository and Archive, University of Zurich ZORA URL: https://doi.org/10.5167/uzh-66174 Accepted Version Originally published at: Blyszczuk, Przemyslaw; Berthonneche, Corrine; Behnke, Silvia; Glönkler, Marcel; Moch, Holger; Pedrazzini, Thierry; Lüscher, Thomas F; Eriksson, Urs; Kania, Gabriela (2013). Nitric oxide synthase 2 is required for conversion of pro-fibrogenic inflammatory CD133+ progenitors into F4/80+ macrophages in experimental autoimmune myocarditis. Cardiovascular Research, 97(2):219-229. DOI: https://doi.org/10.1093/cvr/cvs317

Cardiovascular Research Advance Access published October 22, 2012 1 Nitric oxide synthase 2 is required for conversion of pro-fibrogenic inflammatory CD133 + progenitors into F4/80 + macrophages in experimental autoimmune myocarditis Przemyslaw Blyszczuk 1,2, Corrine Berthonneche 3, Silvia Behnke,4, Marcel Glönkler 5, Holger Moch 5, Thierry Pedrazzini 3, Thomas F. Lüscher 1,6, Urs Eriksson 1,2, Gabriela Kania 1,2 1 Cardiovascular Research and Zürich Center for Integrative Human Physiology; Institute of Physiology, University of Zürich, Winterthurerstr. 190, CH-8057 Zürich, Switzerland; 2 Department of Medicine, GZO - Zürich Regional Health Center, Spitalstr. 66, CH-8620 Wetzikon, Switzerland; 3 Experimental Cardiology Unit, Department of Medicine, University of Lausanne Medical School, CH-1011 Lausanne, Switzerland 4 Sophistolab AG, Bauelenzelgstrasse 20, 8193 Eglisau, Switzerland, 5 Institute for Surgical Pathology, Department of Pathology; 6 Department of Cardiology; University of Zurich, Switzerland; Winterthurerstr. 190, CH-8057 Zürich, and University Hospital Zürich, Rämistr. 100, CH-8001 Zürich, Switzerland Corresponding author: Gabriela Kania, PhD Cardioimmunology, Cardiovascular Research, Institute of Physiology, University of Zürich Winterthurerstrasse 190, CH-8057 Zürich, Switzerland phone: ++41-44-6354995; fax: ++41-44-2558701 E-mail: gabriela.kania@uzh.ch Shared last authorship Published on behalf of the European Society of Cardiology. All rights reserved. The Author 2012. For permissions please email: journals.permissions@oup.com

2 Abstract Aims: Experimental Autoimmune Myocarditis (EAM) model mirrors important mechanisms of inflammatory dilated cardiomyopathy (idcm). In EAM, inflammatory CD133 + progenitors are a major cellular source of cardiac myofibroblasts in post-inflammatory myocardium. We hypothesized that exogenous delivery of Macrophage-Colony Stimulating Factor (M-CSF) can stimulate macrophage lineage differentiation of inflammatory progenitors and therefore prevent their naturally occurring myofibroblast fate in EAM. Methods and Results: EAM was induced in wild-type (BALB/c) and Nitric oxide synthase 2 deficient (Nos2 / ) mice and CD133 + progenitors were isolated from inflamed hearts. In vitro, M-CSF converted inflammatory CD133 + progenitors into nitric oxide-producing F4/80 + macrophages and prevented TGF-βmediated myofibroblast differentiation. Importantly, only a subset of heart-infiltrating CD133 + progenitors expresses macrophage-specific antigen F4/80 in EAM. These CD133 + /F4/80 hi cells show impaired myofibrogenic potential compared to CD133 + /F4/80 cells. M-CSF treatment of wild-type mice with EAM at the peak of disease markedly increased CD133 + /F4/80 hi cells in the myocardium, and CD133 + progenitors isolated from M-CSF-treated mice failed to differentiate into myofibroblasts. In contrast, M-CSF was not effective in converting CD133 + progenitors from inflamed hearts of Nos2 / mice into macrophages, and M- CSF treatment did not result in increased CD133 + /F4/80 hi cell population in hearts of Nos2 / mice. Accordingly, M-CSF prevented post-inflammatory fibrosis and left ventricular dysfunction in wild-type but not in Nos2 / mice. Conclusions: Active and NOS2-dependent induction of macrophage lineage differentiation abrogates the myofibrogenic potential of heart-infiltrating CD133 + progenitors. Modulating the in vivo differentiation fate of specific progenitors might become a novel approach for the treatment of inflammatory heart diseases. Key words: experimental autoimmune myocarditis, CD133 progenitor, M-CSF, myofibroblast, macrophage, nitric oxide synthase 2

Introduction Inflammatory dilated cardiomyopathy (idcm) is an important cause of heart failure and sudden death in children and young patients 1. Progressive cardiac dilation and fibrosis are hallmarks of idcm in humans. idcm refers to an end stage heart failure phenotype which often results from virus-triggered myocarditis 2. Both, clinical observations and animal experiments suggest that infection-triggered autoimmunity plays an important role in idcm 3. Heart-specific autoimmune responses are a consequence of the lack of T cell tolerance to heart-specific alpha-myosin heavy chain (αmyhc) in mice and in humans 4. Experimental autoimmune myocarditis (EAM) is a CD4 + T cell-mediated mouse model of idcm. In susceptible mouse strains, EAM is commonly induced after immunization with αmyhc peptide together with a strong adjuvant 4. In BALB/c mice, the extent of cardiac infiltrates peaks three weeks after immunization. Inflammatory infiltrates largely resolve thereafter, but the process of pathological remodelling continues and many mice develop an end stage heart failure phenotype, including ventricular dilation, myocardial fibrosis and heart failure on follow-up 5. At the peak of EAM, inflammatory cells, including granulocytes, monocytes, macrophages, T cells, B cells, and CD133 + progenitors infiltrate the myocardium 4, 6. CD133 (prominin-1) and its human orthologue, AC133, are often expressed on primitive cell populations including hematopoietic stem cells. In EAM, bone marrow-derived CD133 + progenitors represent the major cellular source of collagen-producing fibroblasts involved in the progressive Transforming Growth Factor-beta (TGF-β)-mediated cardiac remodelling 6, which reflects the transition from acute inflammation into the typical end stage heart failure phenotype. Macrophages represent another important heart-infiltrating cell subset in EAM 7. In mice, macrophages commonly express the F4/80 antigen. Their role in disease pathogenesis, however, remains controversial. So far, the nitric oxide synthase 2 (NOS2)-expressing M1 macrophage subset had been considered to contribute to disease progression 7, 8, On the other hand, nitric oxide also acts as a strong immunosuppressive molecule preventing EAM development via induction of T cell apoptosis 6. Alternatively activated M2 macrophages, are considered to promote resolution of inflammation and reduce disease severity 7-9.

4 Macrophage-colony stimulating factor (M-CSF) is one of the key cytokines regulating immune responses and reparative processes in many inflammatory and cardiovascular disorders 10, 11. M-CSF plays a pivotal role in differentiation and maturation of the macrophage myeloid lineage 12. Mice lacking functional M-CSF show macrophage deficiency, reduced phagocytosis and strongly impaired nitric oxide production 13. In animal models of ischemic heart failure and viral myocarditis, M-CSF treatment improves cardiac function 14, 15,16, but the mechanism is still unclear. We hypothesize that altering the in vivo fate of inflammatory progenitors by selectively promoting macrophage lineage differentiation, may protect from myofibroblast accumulation and cardiac fibrosis in EAM. In fact, M-CSF induced conversion of heart-infiltrating CD133 + progenitors into F4/80 + macrophages and prevented their myofibroblast differentiation in the post-inflammatory phase of EAM. Moreover, our data points to critical role of NOS2 and nitric oxide in M-CSF-dependent macrophage lineage differentiation of inflammatory CD133 + progenitors. Thus, we demonstrate how exogenous cytokine treatments may change the default differentiation fate of inflammatory progenitors and can prevent pathological tissue remodelling in idcm.

Methods Mice BALB/c (n=250) and Nos2 / (n=50) mice on BALB/c background were used in this study. Animal experiments were performed in accordance with the Swiss federal law and with the Guide for the Care and Use of Laboratory Animals published by the US National Institutes of Health (NIH Publication, 8th Edition, 2011). All animal experiments were approved by the Cantonal Veterinary Office in Zürich. EAM induction and cytokine treatments Mice were injected subcutaneously with 150 µg of αmyhc (Ac-RSLKLMATLFSTYASADR-OH; Caslo) peptide emulsified 1:1 with Complete Freund's Adjuvant (CFA, Difco) on days 0 and 7. 200 µg/kg M-CSF (Peprotech) were intravenously injected 5x every second day between days 21-29 or days 40-48 of EAM. Control mice received solvent only. Histopathology and Immunocytochemistry Hearts were formalin-fixed and paraffin-embedded. Heart sections were stained with rat anti-mouse CD45 (BD Bioscience), rabbit anti-mouse CD3 (Neomarkers), rat anti-mouse F4/80 (BMA biomedicals) and rabbit anti-rat IgG (Abcam) antibodies, and with Bond Polymer Refine Detection kit using BOND-MAX system (both Leica). Masson s Trichrom staining was used to detect fibrosis. Immunopositive cells and fibrotic areas were quantified using analysis FIVE software (Olympus). Echocardiography Mice were lightly anesthetized with 1-1.5% isoflurane, maintaining heart rate at 400-500 beats per minute. Transthoracic echocardiography was performed using a 30 MHz probe and the Vevo 770 Ultrasound system (VisualSonics) as described 17 and in Supplementary Material. Cell cultures Myocarditis-positive hearts were perfused, dissected and digested with Liberase Blendzyme (Roche) for 45

6 min at 37 C and tissue suspensions were passed sequentially through 70 µm and 40 µm cell strainers. Cardiomyocytes were separated by low speed centrifugation (50 g, 2 min). CD133 + cells were isolated by positive selection using anti-cd133-pe antibody (ebioscience) and magnetic anti-pe-microbeads (Miltenyi) or using FACS. Enriched CD133 + cells were plated onto gelatine-coated cell culture plates and cultured in the Iscove's Modified Dulbecco's Medium supplemented with 20% foetal bovine serum, 1:100 penicillin/streptomycin, 100 mm non-essential amino acids, 100 mm sodium pyruvate and 50mM β- mercaptoethanol (all Invitrogen). Macrophage differentiation was induced with 10 ng/ml M-CSF, and fibroblast differentiation with 10 ng/ml TGF-β (both PeproTech). Flow cytometry and cell sorting Single cell suspensions were prepared from digested hearts and cultured cells. Cells were incubated 30 min on ice with the appropriate combination of fluorochrome- or biotin-conjugated antibodies. The following antibodies were used: anti-cd45-fitc, anti-cd45-pe, anti-nos2-fitc (all BD Bioscience), anti-cd133- PE, anti-f4/80-pe (both ebioscience), biotin-conjugated anti-f4/80 (Cedarlane), anti-cd16/32 (Miltenyi), anti-cd206 (Biolegend), anti-cd301 (Serotec), anti-cd133 (ebioscience). Streptavidin-APC (BD Bioscience) was used to detect biotin-conjugated antibodies. Cells were analysed with the FACSCanto analyser (BD Bioscience) and FlowJo software (Tree Star). In some experiments, cells were sorted with FACSAria III (BD Bioscience). Quantitative RT-PCR Total RNA was isolated using RNeasy micro kit (Qiagen). cdnas were amplified using the Power SYBR Green PCR Master Mix (Applied Biosystems) and oligonucleotides complementary to transcripts of the analyzed genes using the 7500 Fast Real-Time PCR System (Applied Biosystems). The following oligonucleotides were used in this study: αsma (Acta2): 5 -cgctgtcaggaaccctgaga-3, 5 - cgaagccggccttacaga-3 ; collagen I (Col1a1): 5 -gatgacgtgcaatgcaatgaa-3, 5 -ccctcgactcctacatcttctga- 3 ; fibronectin (Fn1): 5 -taccaaggtcaatccacacccc-3, 5 -cagatggcaaaagaaagcagagg-3 ; gapdh (Gapdh): 5 - ctgcaccaccaactgcttagc-3, 5 -ggcatggactgtggtcatgag-3. Transcript levels of gapdh were used as endogenous reference, and relative gene expression was analyzed using the 2 - Ct method.

Immunohistochemistry and phagocytosis assays Cells were cultured on gelatine-coated cover slips and fixed with 4% paraformaldehyde or methanol:acetone (7:3). 1% bovine serum albumin was used as blocking solution. The following antibodies were used: mouse IgG anti-αsma (Sigma), rabbit IgG anti-fibronectin (Milipore), AlexaFluor488 anti-mouse IgG and AlexaFluor546 anti-rabbit IgG 1:600 (both Invitrogen). DAPI was used to label nuclei. Alexa Fluor 488- conjugated Escherichia coli BioParticles (Invitrogen) were used according to manufacturer s recommendations. Immunofluorescence was analysed using the Olympus BX51 fluorescence microscope. Statistics Normally distributed data were analysed by unpaired, two-tailed Student s t-test and by one-way ANOVA followed by Bonferroni post-hoc test. Severity scores were analysed by the one-way Kruskal-Wallis analysis. For correlation analysis, Pearson's correlation coefficient was calculated. All analyses were computed using GraphPad Prism 5 software. Differences were considered as statistically significant for p<0.05.

8 Results M-CSF converts heart-infiltrating CD133 + progenitors into functional F4/80 + macrophages, and prevents TGF-β-induced myofibroblast differentiation in vitro. BALB/c mice receiving two subcutaneous injections of αmyhc/cfa at days 0 and 7 develop severe myocarditis at day 21. At this stage, CD133 + progenitors represent about 30% of all CD45 + inflammatory cells in the heart 6. To assess the differentiation capacity of these cells at the peak of disease, CD133 + cells were isolated from diseased hearts at day 21 of EAM, plated and expanded for two weeks in vitro. Expanded cells were cultured in the presence or absence of M-CSF or TGF-β for additional 14 days. In the presence of TGF-β, CD133 + cells differentiated into myofibroblasts, positive for αsma and fibronectin (Fig. 1A-B). In contrast, addition of M-CSF failed to trigger myofibroblast differentiation (Fig. 1A-B) and pre-treatment of CD133 + cells with M-CSF for 3 days prevented TGF-β-triggered myofibroblast formation (Fig. 1C-D). In cytokine-free cultures, most expanded cells remained positive for CD133 and were mainly negative for macrophage marker F4/80 (Supplementary Figure 1A-B). In the presence of M-CSF, however, CD133 expression was reduced (Supplementary Figure 1A) and cells up-regulated F4/80, CD11b and CD16/32, suggesting macrophage lineage differentiation (Supplementary Figure 1B-D). Cells cultured with M-CSF developed into fully active macrophages producing NOS2, secreting nitric oxide and phagocyting E.coli bacteria (Supplementary Figure 1E,F,J). Of note, M-CSF treatment did not affect expression of markers characteristic for alternatively activated M2 macrophages (CD206, CD301, Supplementary Figure 1G-H) or markers characteristic for granulocytes or dendritic cells, i.e. Gr-1 (Ly6C, Supplementary Figure 1I), MHC class II and CD11c (not shown). To address the immunomodulating potential of CD133 + progenitors and CD133 + -derived M-CSFdifferentiated macrophages we co-cultured them with αmyhc-reactive Th17 cell lines in the presence of αmyhc-pulsed irradiated splenocytes as antigen presenting cells. On irradiated splenocytes, αmyhcreactive CD4 + T cells proliferated rapidly. In the presence of CD133 + progenitors or M-CSF-differentiated macrophages, however, proliferation was completely abolished (Supplementary Figure 2A). Further, we induced EAM in BALB/c mice and additionally administrated intravenously 2x10 6 of either in vitro

expanded CD133 + progenitors or CD133 + -derived mature macrophages at day 7 after EAM induction and analysed myocarditis severity at day 21. In animals treated with both, CD133 + progenitors and M-CSFdifferentiated macrophages practically no myocarditis was observed compared to sham treated mice (Supplementary Figure 2B). In contrast, administration of M-CSF-differentiated macrophages expanded from Nos2 / mice failed to protect from myocarditis (Supplementary Figure 2C). These latter findings confirm the protective and NOS2-dependent role of CD133 + -derived M-CSF-differentiated macrophages in EAM. M-CSF treatment promotes accumulation of F4/80 + macrophages in the post-inflammatory heart At day 21, inflamed hearts contained a substantial pool of CD133 + progenitors, but fibrosis was not evident at this time point. As illustrated above, M-CSF effectively directs CD133 + progenitors into functional macrophages and prevents their myofibroblast differentiation. We therefore analysed how systemic M-CSF treatment of αmyhc/cfa-immunized mice affects the pattern of heart-infiltrating cell subsets. Accordingly, αmyhc/cfa-immunized mice received M-CSF injections between days 21-29 of EAM (Supplementary figure 3A). Three days after the first M-CSF injection, we observed significantly increased number of F4/80 + and CD133 + cells in the myocardium of M-CSF treated mice (day 24, Figure 2A-B). Immunopositive F4/80 + macrophages in the myocardium decreased over time and remained at low numbers at day 40 (Figure 2A; Supplementary Figure 4A-B). M-CSF treatments, however, did not affect the expression of CD16/32, CD206 and CD301 in inflammatory macrophages (Figure 2E-G). Instead, we observed significantly increased NOS2 on F4/80 + cells in the heart after M-CSF treatment of wild-type mice (Figure 2D). M-CSF inhibits formation of myofibroblasts from inflammatory CD133 + progenitors in EAM Heart-infiltrating CD133 + progenitors represent the major cellular source of myofibroblasts in postinflammatory EAM 6. At day 24 of EAM, we sorted inflammatory (CD45-positive) CD133 + /F4/80 and CD133 + /F4/80 hi cells from the inflamed myocardium of BALB/c mice and found elevated levels of myofibroblast-specific genes in CD133 + cells negative for F4/80 (Figure 3A-B). These findings suggest that CD133 + /F4/80 rather than CD133 + /F4/80 hi cells contribute to fibrogenesis in post-inflammatory EAM.

10 M-CSF-stimulated CD133 + progenitors acquired a functional macrophage phenotype within 3 days in vitro (Figure 2H). We therefore analysed F4/80 expression on heart-infiltrating CD133 + progenitors in M- CSF-treated mice. We found that heart-infiltrating CD133 + /CD45 + cells of M-CSF-treated mice had marked higher F4/80 at day 24 (i.e. 3 days after the cytokine treatment, Figure 3C-D). Next, we sorted heartinfiltrating CD133 + /CD45 + cells from PBS- and M-CSF-treated mice and found reduced mrna levels of myofibroblast-specific genes in the M-CSF-treated group (Figure 3E). Furthermore, sorted heart-infiltrating CD133 + /CD45 + cells from both groups were plated and cultured in the presence of TGF-β for 10 days. CD133 + /CD45 + cells isolated from the myocardium of M-CSF-treated mice failed to differentiate into αsma- and fibronectin-expressing myofibroblasts (Figure 3F). NOS2 controls M-CSF-dependent macrophage differentiation of heart-infiltrating CD133 + progenitors F4/80 hi macrophages derived from CD133 + progenitors up-regulate NOS2 and produce nitric oxide. We therefore addressed the role of NOS2 in the macrophage differentiation processes. Accordingly, CD133 + progenitors were isolated at day 21 after immunization from inflamed hearts of wild-type or Nos2 / mice and cultivated in the presence of M-CSF. Nos2 / CD133 + progenitors showed reduced F4/80 and CD14 expression and were functionally impaired as indicated by reduced E.coli phagocytosis (Figure 4A-E). To determine whether macrophage differentiation was mediated by NOS activity, CD133 + progenitors isolated from inflamed hearts of BALB/c mice were cultured for 3 days in the presence of M-CSF with or without L- NAME, a non-specific NOS inhibitor. Cells cultured with L-NAME showed significantly reduced E.coli phagocytosis (Figure 4F) and reduced nitric oxide levels (Figure 4G). M-CSF treatment prevents cardiac fibrosis and left ventricular dysfunction in EAM M-CSF prevents TGF-β-mediated myofibroblast differentiation and promotes formation of macrophages from inflammatory CD133 + progenitors. Given that CD133 + progenitors represent the major source for myofibroblasts in EAM, we addressed whether M-CSF treatment reduces fibrogenesis in the postinflammatory heart. Accordingly, we treated αmyhc/cfa-immunized mice with M-CSF between days 21-29 or 40-48 of EAM, and analysed the hearts at day 40 and 60 respectively (Supplementary figure 3A). As

illustrated in the Figure 5, M-CSF treatment completely prevented accumulation of fibroblasts in the postinflammatory heart (Figure 5A-C; Supplementary figure 4B). Of note, M-CSF treatment failed to attenuate fibrosis when delivered between days 40-48 (Figure 5A). These findings suggest that M-CSF treatment prevented the fibrotic process, but did not revert already established cardiac fibrosis. Relapses of inflammatory cells are quite common in autoimmune diseases. In BALB/c mice, we observed spontaneous inflammatory relapses 40 days after the first immunization (Figure 5D-E). During relapses, CD45 + inflammatory cells were mainly detected in the pericardium and largely represented CD3 + T lymphocytes (Figure 5D-E, Supplementary figure 3C). Interestingly, M-CSF treatment between days 21-29 inhibited relapses in αmyhc/cfa-immunized mice as reflected by reduced numbers of CD45 + and CD3 + T cells in the myocardium at day 40 (Figure 5D-E; Supplementary figure 3C). Cardiac fibrosis parallels both, diastolic dysfunction, and reduced cardiac contractility. Not all immunized mice develop severe myocarditis. Consequently the extent of post-inflammatory fibrosis varies in the EAM model. In our series, 11 out of 20 αmyhc/cfa-immunized BALB/c mice showed substantial fibrosis (>2% of total heart area) on day 40. At this time point, echocardiography revealed reduced ejection fraction and fractional shortening in mice with significant fibrosis (Figure 5F). In addition, the extent of fibrosis correlated with increased mass, systolic and diastolic volume and isovolumetric contraction time of the left ventricle (Supplementary Figure 5A). Echocardiography of αmyhc/cfa-immunized and M-CSFtreated mice showed unaffected cardiac function at day 40 (Figure 5F, Supplementary Figure 5E). Next, we immunized Nos2 / mice with αmyhc/cfa and treated with M-CSF or PBS between days 21-29. αmyhc/cfa immunization resulted in myocarditis in Nos2 / mice (not shown), but M-CSF treatment failed to affect fibrosis and heart functions during the post-inflammatory phase of EAM in Nos2 / mice (Figure 6A, Supplementary figure 6). Similar results were observed if mice were M-CSF treated between days 14-22 (not shown). M-CSF treatment of Nos2 / mice failed to increase the numbers of F4/80 + macrophages and CD133 + cells in the myocardium at day 24 and 40 of EAM (Figure 6A-C). We also observed no differences in F4/80 expression on inflammatory CD133 + /CD45 + progenitors (not shown) and in the expression of M1/M2 markers on heart infiltrating F4/80 + /CD45 + macrophages between M-CSF- and PBS-treated Nos2 / mice (Figure 6D).

12 Discussion We previously reported that in the EAM model post-inflammatory pathogenic myofibroblasts mainly originate from heart-infiltrating CD133 + progenitor cells 6. In line with our previous findings, herein we demonstrate that M-CSF controls the formation of anti-inflammatory macrophages from inflammatory CD133 + progenitors and prevents TGF-β-mediated differentiation into pathogenic myofibroblasts. Thus, our data suggest that in myocarditis, a significant pool of cells infiltrating the myocardium at the peak of disease represent non-committed progenitors. From a clinical perspective it is important that the differentiation fate of multipotent, noncommitted precursor cells infiltrating the myocardium, can be modulated via specific cytokine signalling in a controlled fashion. M-CSF is a key cytokine guiding macrophage differentiation in vitro 8 and in vivo 18. In EAM, M-CSF treatment up-regulated F4/80 expression on CD133 + inflammatory progenitors promoting macrophage differentiation. In untreated mice, however, heart-infiltrating CD133 + progenitors spontaneously differentiate into pathogenic myofibroblasts. To our knowledge, this is the first report describing the change of in vivo fate of inflammatory progenitors naturally present in the inflamed organ. So far, it has been described that M-CSF treatment improved cardiac function in a viral model of myocarditis 16, after myocardial infarction 14, 15, 19 and in the ischemia-reperfusion model 20. Similar to our findings, M-CSF treatment promoted monocyte/macrophage accumulation in the heart after myocardial infarction 14, 15 or virus inoculation 16. Myocardial fibrosis plays a dual role in remodelling after cardiac injury. On one hand, it is a prerequisite for wound healing, as in case of ischemic injuries, for example. On the other hand, it contributes to ventricular stiffening and typical pathological remodelling in heart failure. Our previous and current data clearly show that excessive cardiac fibrosis parallels impaired cardiac function in EAM 17. Thus, preserved left ventricular function in M-CSF-treated mice results primarily from reduced myofibroblast differentiation, rather than protective macrophage activity. We propose that the conversion of inflammatory progenitors into functional macrophages, instead of myofibroblasts, represent a key mechanism explaining reduced fibrogenesis upon M-CSF treatment. However, we cannot rule out that M-CSF also activates other pathways inhibiting myocardial fibrogenesis. For example, M-CSF may protect cardiomyocytes from H 2 O 2 -induced death 20. Moreover, M-CSF could affect extracellular matrix composition and levels of matrix

metalloproteinases and cytokines/chemokines, which drastically alter the myocardial microenvironment. Nevertheless, we can exclude direct M-CSF-mediated effects on cardiac fibroblasts, because upon M-CSF treatment during the late post-inflammatory phase of the EAM (days 40-48), the fibrosis was not reverted. It has been proposed that differential macrophage activation defines protective and pathogenic functions. Accordingly, adoptive transfer of M1 macrophages promoted histological disease scores, whereas injection of M2 macrophages was largely protective in Coxackievirus-induced myocarditis 9. Also most heart-infiltrating macrophages were classified as M2 macrophages expressing mannose receptor and Gr-1 in αmyhc/cfa- and Coxackievirus-induced myocarditis 7, 21. However, macrophages expressing M1 marker NOS2 have been identified in myocarditis as well 22. We critically analysed macrophages in the myocardium after the peak of disease, during the post-inflammatory phase of EAM, and detected both, M1- and M2- specific markers. We were, however, unable to clearly delineate two distinct activation states. We assume that during EAM, heart-infiltrating macrophages were activated classically (M1) and alternatively (M2) at the same time. In fact, it is well known that T cells and other heart-infiltrating cells massively produce both, M1-activating IFN-γ as well as M2-activating IL-4 and IL-13 7. Importantly, M-CSF treatment did not affect the expression of M1 and M2 markers except of NOS2. M-CSF-induced macrophage accumulation in the post-inflammatory heart was not pathogenic in our model, because resolution of inflammation, defined by the extent of inflammatory cells was clearly M-CSF independent. Moreover, M-CSF-induced macrophages showed strong immunosuppressive activity in vitro and in vivo. We believe that accumulation of the immunosuppressive macrophages in M-CSF-treated mice protected from spontaneous T cell relapse. Nevertheless, we cannot, entirely rule out that M-CSF also mediates protective effects by acting on other target cells. We previously found that M-CSF-induced CD11b + monocytes prevented EAM development and suppressed CD4 + T cell proliferation in a nitric oxide-dependent manner 23. M-CSF treatment promoted inflammatory macrophages expressing NOS2 in EAM, We therefore hypothesized that macrophageproduced nitric oxide might control both; T cell mediated early inflammation as well as post-inflammatory fibrosis in EAM. Indeed, mice lacking NOS2 or treated with nitric oxide inhibitors showed enhanced cardiac inflammation and fibrotic lesions in a model of Coxackievirus-induced myocarditis 24, 25. Similarly, Nos2 / mice also develop more severe myocarditis in the EAM model (Kania, Blyszczuk & Eriksson- unpublished

14 observations). Nevertheless, we observed spontaneous resolution of inflammation in Nos2 / mice, suggesting that NOS2 is not critically involved in the resolution of inflammation in the EAM model. Surprisingly, treatment of Nos2 / mice with M-CSF failed to turn CD133 + progenitors into macrophages and to prevent fibrosis development. Furthermore, we showed that Nos2 / CD133 + progenitors stimulated with M-CSF failed to generate functional macrophages. These results suggested that NOS2 was required for M-CSF-induced macrophage differentiation and maturation. Accordingly, NOS2 has been reported to control differentiation of human monoblasts 26. Regulation of gene expressions by nitric oxide can explain requirement of NOS2 in this differentiation processes 27. At this stage, we cannot, however, exclude that in Nos2 / mice, heart-infiltrating CD133 + progenitors are committed toward fibroblast phenotype due to alerted cytokine expression profile in the myocardium. Based on our findings, we can assume that M-CSF stimulates nitric oxide production that, in turn, promotes macrophage differentiation of inflammatory progenitors. In summary, our data demonstrate that a single hematopoietic cytokine, such as M-CSF can modulate the in vivo fate of inflammatory CD133 + progenitors in a way to promote their differentiation into macrophages instead of myofibroblasts in the post-inflammatory phase of the EAM. Furthermore, our data suggest that mature inflammatory macrophages, in contrast to progenitor-derived fibroblasts, do not promote heart failure after acute myocarditis. These findings are in line with the clinical observation that fulminant myocarditis implies an excellent long-term prognosis whereas subacute myocarditis usually progresses to end stage heart failure 28. Acknowledgment We thank Marta Bachman for excellent technical assistance, Flow Cytometry Facility at University Zurich for excellent sorting service and Dr. Pavani Mocharla for critical reading. Funding U.E. acknowledges support from the Swiss Life Foundation, P.B. from the Swiss Heart Foundation and G.K. from the Olga Mayenfisch Foundation, Hartmann Müller Foundation, and Holcim Foundation. The study was supported by the Swiss National Science Foundation (U.E. and G.K.: Grant 32003B_130771) and Swiss

National Science Foundation MHV subsidy (G.K.: Grant PMPDP3_129013). Conflict of Interest None

16 References: 1. Aretz HT, Billingham ME, Edwards WD, Factor SM, Fallon JT, Fenoglio JJ, Jr. et al. Myocarditis. A histopathologic definition and classification. Am J Cardiovasc Pathol. 1987;1:3-14. 2. Kishimoto C, Abelmann WH. In vivo significance of T cells in the development of Coxsackievirus B3 myocarditis in mice. Immature but antigen-specific T cells aggravate cardiac injury. Circ Res. 1990;67:589-598. 3. Leuschner F, Katus HA, Kaya Z. Autoimmune myocarditis: past, present and future. J Autoimmun. 2009;33:282-289. 4. Rose NR. Myocarditis: infection versus autoimmunity. J Clin Immunol. 2009;29:730-737. 5. Kania G, Blyszczuk P, Eriksson U. Mechanisms of cardiac fibrosis in inflammatory heart disease. Trends Cardiovasc Med. 2009;19:247-252. 6. Kania G, Blyszczuk P, Stein S, Valaperti A, Germano D, Dirnhofer S et al. Heart-infiltrating prominin-1+/cd133+ progenitor cells represent the cellular source of transforming growth factor beta-mediated cardiac fibrosis in experimental autoimmune myocarditis. Circ Res. 2009;105:462-470. 7. Fairweather D, Cihakova D. Alternatively activated macrophages in infection and autoimmunity. J Autoimmun. 2009;33:222-230. 8. Gordon S, Martinez FO. Alternative activation of macrophages: mechanism and functions. Immunity. 2010;32:593-604. 9. Li K, Xu W, Guo Q, Jiang Z, Wang P, Yue Y et al. Differential macrophage polarization in male and female BALB/c mice infected with coxsackievirus B3 defines susceptibility to viral myocarditis. Circ Res. 2009;105:353-364. 10. Parissis J, Filippatos G, Adamopoulos S, Li X, Kremastinos DT, Uhal BD. Hematopoietic colony stimulating factors in cardiovascular and pulmonary remodeling: promoters or inhibitors? Curr Pharm Des. 2006;12:2689-2699. 11. Hamilton JA. Colony-stimulating factors in inflammation and autoimmunity. Nat Rev Immunol. 2008;8:533-544. 12. Wiktor-Jedrzejczak W, Gordon S. Cytokine regulation of the macrophage (M phi) system studied

using the colony stimulating factor-1-deficient op/op mouse. Physiol Rev. 1996;76:927-947. 13. Naito M, Hayashi S, Yoshida H, Nishikawa S, Shultz LD, Takahashi K. Abnormal differentiation of tissue macrophage populations in 'osteopetrosis' (op) mice defective in the production of macrophage colony-stimulating factor. Am J Pathol. 1991;139:657-667. 14. Yano T, Miura T, Whittaker P, Miki T, Sakamoto J, Nakamura Y et al. Macrophage colonystimulating factor treatment after myocardial infarction attenuates left ventricular dysfunction by accelerating infarct repair. J Am Coll Cardiol. 2006;47:626-634. 15. Morimoto H, Takahashi M, Shiba Y, Izawa A, Ise H, Hongo M et al. Bone marrow-derived CXCR4+ cells mobilized by macrophage colony-stimulating factor participate in the reduction of infarct area and improvement of cardiac remodeling after myocardial infarction in mice. Am J Pathol. 2007;171:755-766. 16. Hiraoka Y, Kishimoto C, Takada H, Suzaki N, Shiraki K. Colony-stimulating factors and coxackievirus B3 myocarditis in mice: macrophage colony-stimulating factor suppresses acute myocarditis with increasing interferon-alpha. Am Heart J. 1995;130:1259-1264. 17. Blyszczuk P, Kania G, Dieterle T, Marty RR, Valaperti A, Berthonneche C et al. Myeloid differentiation factor-88/interleukin-1 signaling controls cardiac fibrosis and heart failure progression in inflammatory dilated cardiomyopathy. Circ Res. 2009;105:912-920. 18. Wiktor-Jedrzejczak W, Ratajczak MZ, Ptasznik A, Sell KW, Ahmed-Ansari A, Ostertag W. CSF-1 deficiency in the op/op mouse has differential effects on macrophage populations and differentiation stages. Exp Hematol. 1992;20:1004-1010. 19. Miki T, Miura T, Nishino Y, Yano T, Sakamoto J, Nakamura Y et al. Granulocyte colony stimulating factor/macrophage colony stimulating factor improves postinfarct ventricular function by suppression of border zone remodelling in rats. Clin Exp Pharmacol Physiol. 2004;31:873-882. 20. Okazaki T, Ebihara S, Asada M, Yamanda S, Saijo Y, Shiraishi Y et al. Macrophage colonystimulating factor improves cardiac function after ischemic injury by inducing vascular endothelial growth factor production and survival of cardiomyocytes. Am J Pathol. 2007;171:1093-1103. 21. Cihakova D, Barin JG, Afanasyeva M, Kimura M, Fairweather D, Berg M et al. Interleukin-13 protects against experimental autoimmune myocarditis by regulating macrophage differentiation.

18 Am J Pathol. 2008;172:1195-1208. 22. Lowenstein CJ, Hill SL, Lafond-Walker A, Wu J, Allen G, Landavere M et al. Nitric oxide inhibits viral replication in murine myocarditis. J Clin Invest. 1996;97:1837-1843. 23. Valaperti A, Marty RR, Kania G, Germano D, Mauermann N, Dirnhofer S et al. CD11b+ monocytes abrogate Th17 CD4+ T cell-mediated experimental autoimmune myocarditis. J Immunol. 2008;180:2686-2695. 24. Zaragoza C, Ocampo C, Saura M, Leppo M, Wei XQ, Quick R et al. The role of inducible nitric oxide synthase in the host response to Coxsackievirus myocarditis. Proc Natl Acad Sci U S A. 1998;95:2469-2474. 25. Hiraoka Y, Kishimoto C, Takada H, Nakamura M, Kurokawa M, Ochiai H et al. Nitric oxide and murine coxsackievirus B3 myocarditis: aggravation of myocarditis by inhibition of nitric oxide synthase. J Am Coll Cardiol. 1996;28:1610-1615. 26. Kim SJ, Bang OS, Lee YS, Kang SS. Production of inducible nitric oxide is required for monocytic differentiation of U937 cells induced by vitamin E-succinate. J Cell Sci. 1998;111 ( Pt 4):435-441. 27. Wink DA, Hines HB, Cheng RY, Switzer CH, Flores-Santana W, Vitek MP et al. Nitric oxide and redox mechanisms in the immune response. J Leukoc Biol. 2011;89:873-891. 28. McCarthy RE, 3rd, Boehmer JP, Hruban RH, Hutchins GM, Kasper EK, Hare JM et al. Long-term outcome of fulminant myocarditis as compared with acute (nonfulminant) myocarditis. N Engl J Med. 2000;342:690-695.

Figure legends Figure 1. M-CSF prevents differentiation of heart-infiltrating CD133 + progenitors into myofibroblasts. (A,B) Heart-infiltrating CD133 + cells were expanded from myocarditis-positive hearts at day 21 of EAM and stimulated without (control, white) or with 10 ng/ml M-CSF (gray), or 10 ng/ml TGF-β (black) for 14 days. Relative mrna expression of myofibroblast-specific fibronectin (Fn1), collagen I (Col1a1) and αsma (Acta2) (A, n=5). Representative immunofluorescence and quantification analysis of αsma (top) and fibronectin (bottom) in cultured cells (B, n=5). (C,D) Expanded heart-infiltrating CD133 + cells (EAM d21) pre-treated for 3 days without (control, black) or with 10 ng/ml M-CSF (+M-CSF d-3, hatch), washed and stimulated with 10 ng/ml TGF-β for 14 days. Relative mrna levels of myofibroblast-specific genes (C, n=5) and representative immunofluorescence and quantification analysis of αsma (D, top) and fibronectin (D, bottom) are shown (n=5). *** p<0.001 (two-tailed Student s t-test versus control), bar = 20µm. Figure 2. M-CSF treatment turns inflammatory CD133 + progenitors into F4/80 + macrophages in EAM. BALB/c mice were immunized with αmyhc/cfa at day 0 and 7, and treated with PBS or M-CSF between days 21-29. (A) Quantification of F4/80 + macrophages in the myocardium of PBS- (black) and M-CSF-treated (white) mice at day 24 (n=6), 28 (n=5) and 40 (n=20). Immunopositive cells were quantified per mm 2 of heart tissue. (B) Flow cytometry analysis of CD133 and F4/80 on heart infiltrating CD45 + -gated cells (left) in EAM. Quantification of CD133 + (left graph) and F4/80 + (right graph) cells gated on CD45 + infiltrates of PBS- (black) and M-CSF-treated (white) mice at day 24 (n=5) and 28 (n=5) of EAM. (C-G) Flow cytometry analysis of M1 and M2 activation markers on heart infiltrating CD45 + /F4/80 + -gated macrophages (left) in EAM. Mean fluorescence intensity (MFI) of membrane markers (CD16/32, CD206, CD301) and intracellular NOS2 expression gated on inflammatory macrophages of PBS- (black) and M- CSF-treated (white) mice at day 24 (n=10) of EAM. ( ) isotype controls, (+) antigen-specific antibodies.

20 (H) Overlay of phase contrast (left and middle) or F4/80 expression (right, red) with E.coli bacteria (green) taken up in the phagocytosis assay of expanded heart-infiltrating CD133 + cells (isolated from heart at d21) stimulated with M-CSF for 1 (left) and 3 days (middle and right). Bar = 20µm. ns p>0.05, * p<0.05, ** p<0.01, *** p<0.001 (two-tailed Student s t-test). Figure 3. M-CSF inhibits myofibroblast lineage differentiation of inflammatory CD133 + in EAM. (A) Representative flow cytometry analysis of CD133 + and F4/80 (right) gated on heart infiltrating CD45 + cells (left) at day 24 of EAM. Numbers indicate percentage of positive cells in the adjacent gates. (B) Heart-infiltrating CD45 + /CD133 + /F4/80 (white) and CD45 + /CD133 + /F4/80 hi (black) cells were FACS sorted from myocarditis-positive hearts at day 24 of EAM. Relative mrna levels of myofibroblast-specific genes are shown for 1 out of 2 independent experiments (n=5). (C) Representative flow cytometry analysis of CD133 and F4/80 gated on heart infiltrating CD45 + /CD133 + progenitor cells (left) of PBS- (+PBS) and M-CSF-treated (+M-CSF) mice at day 24 of EAM. Staining with anti-cd45 and anit-cd133 and IgG control to anti-f4/80 antibodies (IgG control) was used to set the gates. Numbers indicate percentage of positive cells in the adjacent gates. (D) Quantification of F4/80 hi (left) and F4/80 (right) cells gated of CD45 + /CD133 + progenitor cells (gated as shown in C) of PBS- (black) and M-CSF-treated (white) mice at day 24 of EAM. (E,F) Heart-infiltrating CD45 + /CD133 + cells were FACSorted from myocarditis-positive hearts at day 24 of EAM from control PBS-treated (black, +PBS) or M-CSF-treated (white, +M-CSF). A relative mrna level of myofibroblast-specific genes of sorted cells is shown (E, n=5). In addition, sorted cells were plated and stimulated with 10 ng/ml TGF-β for 10 days. Representative immunofluorescence and quantification analysis of αsma (F, top) and fibronectin (F, bottom) are shown (n=5). Bar = 20µm, ns p>0.05, * p<0.05, ** p<0.01, *** p<0.001 (two-tailed Student s t-test). Figure 4. NOS2 is required for M-CSF-dependent macrophage lineage differentiation from CD133 + progenitors. (A-E) Expanded heart-infiltrating CD133 + cells (EAM d21) from wild-type BALB/c and Nos2 / mice were stimulated with 10 ng/ml M-CSF for 3 days. (A) Representative flow cytometry analysis of expanded cells

using anti-cd133 (right) and IgG control (left) antibodies. Numbers indicate percentage of positive cells in the adjacent gates. Representative histogram analysis of macrophage specific antigens F4/80 (B), CD11b (C), CD14 (D), and E.coli bacteria taken up in the phagocytosis assay (E) on CD133 + cells (gated as shown in A). Isotype controls are displayed in grey. Numbers in histograms indicate mean fluorescence intensity (MFI). Bar graphs show quantification of the respective analyses for wild-type (black) and Nos2 / (hatch) cells. n=5, ( ) isotype controls, (+) antigen-specific antibodies or E.coli. (F) Expanded heart-infiltrating CD133 + cells (EAM d21) from BALB/c (wild-type) mice were stimulated with 10 ng/ml M-CSF for 3 days in the absence (control) or presence of 2mM L-NAME (+L-NAME). Representative histogram analysis of E.coli bacteria uptake by CD133 + cells (gated as shown in A). Controls without E.coli are displayed in grey. Numbers in histograms indicate MFI. Bar graphs show quantification of the respective analyses in the absence (white) or presence of L-NAME (grey). n=5, ( ) control, (+) E.coli (G) Nitrite levels in supernatants of cultures stimulated with 0.1 µg/ml LPS for 24h. n = 4 ns p>0.05, * p<0.05, ** p<0.01, *** p<0.001 (two-tailed Student s t-test). Figure 5. M-CSF treatment attenuates myocardial fibrosis and prevents heart failure in EAM. BALB/c mice were immunized at day 0 and 7 with αmyhc/cfa and injected either with PBS (black) or M-CSF between days 21-29 (white) or days 40-48 (grey). (A) Fibrosis was evaluated in heart sections at day 24 (n=10), 28 (n=10), 40 (n=20) and 60 (n=6-8). The degree of fibrosis was analysed by Masson s trichrome staining and calculated as percentage of the fibrotic area in heart sections. (B,C) The representative microphotographs of the myocardial tissue (d60) following PBS (B) or M-CSF (C) treatment between days 21-29 stained with Masson s trichrome. Bar = 100 µm. p values were computed using two-tailed Student s t-test (A, d24, d28 and d40), one-way ANOVA (A, d60). (D-E) Quantification of CD45 + cells (defining all inflammatory infiltrates, D) and CD3 + cells (defining T lymphocytes, E) within the myocardium during EAM at day 24 (n=10), 28 (n=10), 40 (n=20) and 60 (n=6-8) after the first immunization. Immunopositive cells were quantified per mm 2 of heart tissue. (F) Echocardiographic parameters of healthy, untreated BALB/c mice (white) in comparison to

22 αmyhc/cfa-immunised mice with <2% myocardial fibrosis (hatch, n=9), >2% myocardial fibrosis (black, n=11) and αmyhc/cfa-immunised mice treated with M-CSF at days 21-29 (grey, n=20). p values were computed using correlation analysis (F) and the one-way ANOVA (A,D,E). * p<0.05, ** p<0.01, *** p<0.001 for the respective group versus healthy controls (white) calculated with Bonferroni post-hoc test. Figure 6. M-CSF treatment fails to reduce fibrosis in Nos2 / mice. Nos2 / mice were immunized at day 0 and 7 with αmyhc/cfa and treated with PBS or M-CSF between days 21-29. (A,B) Quantification of fibrosis and F4/80 + macrophages in the myocardium of PBS- (black) and M-CSFtreated (white) Nos2 / mice at day 24 (A, n=5) and 40 (B, n=10) of EAM. F4/80 + cells were quantified per mm 2 of heart tissue, and the degree of fibrosis was analysed by Masson s trichrome staining. (C) Representative histograms of CD133 expression gated on heart infiltrating CD45 + inflammatory cells (left) of PBS- (+PBS) and M-CSF-treated (+M-CSF) Nos2 / mice at day 24 of EAM. Isotype controls are displayed in grey. Numbers indicate percentage of positive cells in the presented histograms. Bar graphs show quantification of CD133 + and F4/80 + gated on CD45 + cells of PBS- (black) and M-CSF-treated (white) Nos2 / mice (n=5). (D) Flow cytometry analysis of M1 and M2 activation markers in EAM. Quantification of mean fluorescence intensity (MFI) of membrane macrophage activation markers gated on heart infiltrating CD45 + /F4/80 + -gated macrophages (left) of PBS- (grey) and M-CSF-treated (hatch) Nos2 / mice at day 24 of EAM (n=5). ( ) isotype controls, (+) antigen-specific antibodies, ns p>0.05 (two-tailed Student s t-test).

A! relative expression! 4! 2! Fn1! Col1a1! Acta2! ***! 4! ***! 4! ***! 2! 2! ***! control! +M-CSF! +TGF-β! B! control! +M-CSF! +TGF-β! αsma! Fibronectin! Fibronectin (%)! αsma (%)! 10 5 10 5 ***! ***! C! +TGF-β! relative expression! 1.2! 0.8! 0.4! Fn1! Col1a1! Acta2! ***! ***! 1.2! 1.2! ***! 0.8! 0.8! 0.4! 0.4! control! +M-CSF d-3! D! +TGF-β! αsma! Fibronectin! control! +M-CSF d-3! Fibronectin (%)! αsma (%)! 10 5 10 5 ***! ***! Blyszczuk et al., Figure 1!

A! F4/80 + cells/mm 2! 60 30 p=0.001! p=0.04! p=0.37! d24! d28! d40! +PBS! +M-CSF! C! CD45! B! CD45! F4/8 10000 1000 100 10 1 0 200 400 600 800 1000 FSC! H! + M-CSF! CD133 +! (% of CD45 + )! 6 3 **! d24! d28! d24! d28! MFI! 10 5 4 2 **! **! F4/80 +! (% of CD45 + )! D! E! cell count! control! +M-CSF! NOS2! 26.9! 8 NOS2!! +! cell count! F! G! cell count! control! +M-CSF! CD206! 35.7! 30.9! MFI! 6 4 2 control! +M-CSF! ***! 518! 442! CD16/32! +PBS!!+M-CSF! 30 5! +!! +! CD301! E.coli! E.coli! F4/80/E.coli! d1! d3! d3! MFI! CD16/32! 60! +! CD206! CD301 63.1!! 10 cell count! control! +M-CSF! 54.6! MFI! Blyszczuk et al., Figure 2!

A! CD45! 10000 1000 100 10 C! 1 0 200 400 600 800 1000 10 4 FSC! CD133! 10 4 10 3 10 2 10 1 10 0 Fn1! Col1a1! Acta2! 33! 9! ***! 8! 4! 40 ***! 10 0 10 1 10 2 10 3 10 4 F4/8 IgG control! B! relative expression! 10 4 10 4 10 4 - F4/80! - F4/80 hi! 4! 2! 20 +PBS! +M-CSF! CD45! 10 3 10 2 10 1 10 0 10 0 10 1 10 2 10 3 10 4 8 4 8 4 10 3 10 2 10 1 10 0 D! +M-CSF! +PBS! F4/80 hi (%)! CD133! ***! F4/80 (%)! CD133! 97! 10 0 10 1 10 2 10 3 10 4 **! E! relative expression! CD133! IgG control! 2! 1! 10 3 10 2 10 1 10 0 10 3 10 2 10 66! 1 1 7! 76! F4/8 10 0 10 1 10 2 10 3 10 4 CD133! 10 0 F4/8 10 0 10 1 10 2 10 3 10 4 Fn1! Col1a1! Acta2! 2! 2! **! **! 1! 1! +TGF-β! F! αsma! Fibronectin! +PBS! CD133 + cells! +M-CSF! CD133 + cells! Fibronectin (%)! αsma (%)! 10 5 10 5 ***! ***! Blyszczuk et al., Figure 3!

A! IgG! B! cell count! D! cell count! F! IgG! 10 4 10 3 10 2 10 1 10 0 10 4 10 3 10 2 10 1 10 0 0 200 400 600 800 1000 WT! Nos2 /! WT! Nos2 /! 0.4! 63.9! FSC! 362! F4/8 CD14! 10 4 10 0.2! 3 64.5! FSC! 0 200 400 600 800 1000 19 137! 92! MFI! CD133! MFI! CD133! 10 2 10 1 10 0 10 4 10 3 10 2 10 1 10 0 40 20 10 5 0 200 400 600 800 1000 FSC! FSC! F4/80! ***!! +! CD14! *!! +! 0 200 400 600 800 1000 C! E! cell count! cell count! cell count! control! +L-NAME! WT! Nos2 /! WT! Nos2 /! E.coli! - wild-type +M-CSF! - Nos2 / +M-CSF! CD11b! E.coli! 393! 292! 25.2! 149! 104! 32! MFI! MFI! MFI! 50 25 20 10 40 20 CD11b!! +! E.coli! E.coli!! +! ***!! +! G! nitrite (μm)! 3 ***! ***! 2 1 - M-CSF! - M-CSF +L-NAME! Blyszczuk et al., Figure 4!

A D! E! Fibrosis (%)! CD45 + cells/mm 2! CD3 + cells/mm 2! 15! 1 5! 70 35 40 20 p<0.0001! p=0.003! p=0.0005! p=0.005! p=0.0002! p=0.97! p=0.0004! p=0.79! p=0.27! d24! d28! d40! p<0.0001! p=0.38! p=0.89! d60! B! C! +PBS! +M-CSF, d21-29! +PBS! +M-CSF, d21-29! +M-CSF, d40-48! F! FS%! LV Vol ; d (µl)! 4 3 2 1 10 75! 5 25! -healthy! -MyHC/CFA, fibrosis-negative! -MyHC/CFA, fibrosis-positive! -MyHC/CFA,+M-CSF!! p=0.001! p=0.0001! 8 p=0.0002! 4! **! 6 **! 6 3! **! 4 2! 4 2 1! 2 EF%! p=0.03! p=0.004! p<0.0001! p=0.009! *! 16 *! 12! ***! 2 **! 12 8! 8 1 4 4! LV mass (mg)! LVID, s (mm)! HW/Tibial length! LV Vol ; s (µl)! IVCT (ms)! p=0.003! *! Blyszczuk et al., Figure 5!