SUPPLEMENTARY INFORMATION

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

Resident cardiac stem cells: how to find and use them

Nature Genetics: doi: /ng Supplementary Figure 1. Parameters and consequences of mononuclear cardiomyocyte frequency.

Moore-Morris et al. Supplemental Table 1.

Supplemental Information. Myocardial Polyploidization Creates a Barrier. to Heart Regeneration in Zebrafish

Supplemental Figure 1. Intracranial transduction of a modified ptomo lentiviral vector in the mouse

Supplementary Figure 1. Nature Neuroscience: doi: /nn.4547

Supplementary Figure 1. Baf60c and baf180 are induced during cardiac regeneration in zebrafish. RNA in situ hybridization was performed on paraffin

Tcf21 MCM ; R26 mtmg Sham GFP Col 1/3 TAC 8W TAC 2W. Postn MCM ; R26 mtmg Sham GFP Col 1/3 TAC 8W TAC 2W

Supplementary Materials for

SUPPLEMENTARY INFORMATION

Extending the time window of mammalian heart regeneration by thymosin beta 4

SUPPLEMENTARY INFORMATION

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

Chronic variable stress activates hematopoietic stem cells

Supplementary Information

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

Supplementary material page 1/10

Heart Development. Robert G. Kelly Developmental Biology Institute of Marseilles - Luminy

(a-r) Whole mount X-gal staining on a developmental time-course of hearts from

Probe. Hind III Q,!&#12?R'!! /0!!!!D1"?R'! vector. Homologous recombination

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

Supplementary Figure 1. Spatial distribution of LRP5 and β-catenin in intact cardiomyocytes. (a) and (b) Immunofluorescence staining of endogenous

Fig. S1. Upregulation of K18 and K14 mrna levels during ectoderm specification of hescs. Quantitative real-time PCR analysis of mrna levels of OCT4

SUPPLEMENTARY INFORMATION

Alternatively Activated Macrophages Determine the Repair of the Infarcted

BNP mrna expression in DR and DS rat left ventricles (n = 5). (C) Plasma norepinephrine

SUPPLEMENTARY INFORMATION

Cardiomyocyte proliferation and progenitor cell recruitment underlie therapeutic regeneration after myocardial infarction in the adult mouse heart

Heart Development. Origins of congenital heart defects Properties of cardiac progenitor cells. Robert G. Kelly

SUPPLEMENTARY INFORMATION

1. Cardiomyocytes and nonmyocyte. 2. Extracellular Matrix 3. Vessels שאלה 1. Pathobiology of Heart Failure Molecular and Cellular Mechanism

SUPPLEMENTARY INFORMATION

Supplementary Materials for

Supplementary Figure 1

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

SUPPLEMENTARY INFORMATION

Supplementary Materials for

International Graduate Research Programme in Cardiovascular Science

SUPPLEMENTARY FIGURES

Generation of ST2-GFP reporter mice and characterization of ILC1 cells following infection

a 0,8 Figure S1 8 h 12 h y = 0,036x + 0,2115 y = 0,0366x + 0,206 Labeling index Labeling index ctrl shrna Time (h) Time (h) ctrl shrna S G2 M G1

TISSUE-SPECIFIC STEM CELLS

SUPPLEMENTARY INFORMATION

20 2 Stomach Fig. 2.1 An illustration showing different patterns of the myenteric plexus peculiar to the regions in the guinea-pig stomach stained wit

SUPPLEMENTARY INFORMATION

Supplementary Figure 1. c Human

Impaired Regional Myocardial Function Detection Using the Standard Inter-Segmental Integration SINE Wave Curve On Magnetic Resonance Imaging

SUPPLEMENTARY INFORMATION

Imaging of Coronary Artery Disease: II

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

Supplemental Figures Supplemental Figure 1:

Fog2 is critical for cardiac function and maintenance of coronary vasculature in the adult mouse heart

SUPPLEMENTARY FIGURES

Supplementary Figure 1.

Supplementary Figure 1. Confocal immunofluorescence showing mitochondrial translocation of Drp1. Cardiomyocytes treated with H 2 O 2 were prestained

SUPPLEMENTARY INFORMATION

Santulli G. et al. A microrna-based strategy to suppress restenosis while preserving endothelial function

Supplementary Figure 1: Signaling centers contain few proliferating cells, express p21, and

E10.5 E18.5 P2 10w 83w NF1 HF1. Sham ISO. Bmi1. H3K9me3. Lung weight (g)

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

Myocardial infarction

doi: /nature14508 Rappsilber et al.

Nature Neuroscience: doi: /nn Supplementary Figure 1. MADM labeling of thalamic clones.

Supplementary Figure 1

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

SUPPLEMENTARY INFORMATION

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

Supplementary Information

SUPPLEMENTARY INFORMATION

Supplementary Figure S1 Enlarged coronary artery branches in Edn1-knockout mice. a-d, Coronary angiography by ink injection in wild-type (a, b) and

Mechanisms of heart failure with normal EF Arterial stiffness and ventricular-arterial coupling. What is the pathophysiology at presentation?

PHENOTYPIC DYNAMICS OF MICROGLIAL AND MONOCYTE-DERIVED CELLS IN GLIOBLASTOMA-BEARING MICE.

SUPPLEMENTARY LEGENDS...

Supplementary Figure 1. EC-specific Deletion of Snail1 Does Not Affect EC Apoptosis. (a,b) Cryo-sections of WT (a) and Snail1 LOF (b) embryos at

Automatic cardiac contour propagation in short axis cardiac MR images

Gallic acid prevents isoproterenol-induced cardiac hypertrophy and fibrosis through regulation of JNK2 signaling and Smad3 binding activity

Cardiac Care in pa+ents with Duchenne muscular dystrophy

Supporting Information

SUPPLEMENTARY INFORMATION

Supplementary Figure S1: Tanycytes are restricted to the central/posterior hypothalamus

שינויים מולקולאריים ומבניים באי ספיקת לב אפשרויות לטיפול עתידני

Mass Histology Service

Prospect Cardiac Packages. S-Sharp

T H E J O U R N A L O F C E L L B I O L O G Y

SUPPLEMENTARY RESULTS

Stimulation of endogenous cardioblasts by exogenous cell therapy after myocardial infarction

Supplementary Figure 1 The ability to regenerate an ear hole is discontinuous with wound healing. Ear-hole closure at D85 for each sex within each

9/23/2017. Prof. Steven S. Saliterman. Department of Biomedical Engineering, University of Minnesota

Vevo 2100 System Cardio Measurements. Dieter Fuchs, PhD FUJIFILM VisualSonics, Inc.

Cell Combination Therapy. Disclosures

SUPPLEMENTARY INFORMATION

The Jay Cohn Lecture 2012: Therapeutic regeneration of the injured heart

Supplementary fig. 1. Crystals induce necroptosis does not involve caspases, TNF receptor or NLRP3. A. Mouse tubular epithelial cells were pretreated

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

SUPPLEMENT Supplementary Figure 1: (A) (B)

Supplementary Figure 1: Expression of NFAT proteins in Nfat2-deleted B cells (a+b) Protein expression of NFAT2 (a) and NFAT1 (b) in isolated splenic

Nature Neuroscience: doi: /nn Supplementary Figure 1. Splenic atrophy and leucopenia caused by T3 SCI.

Age-related changes in cardiovascular system. Dr. Rehab Gwada

Mitral valve repair for functional regurgitation caused by Chagas disease

Transcription:

doi:10.1038/nature10188 Supplementary Figure 1. Embryonic epicardial genes are down-regulated from midgestation stages and barely detectable post-natally. Real time qrt-pcr revealed a significant down-regulation (p* 0.05; p** 0.01) of the epicardial genes Wt1, Tbx18, Raldh2 and EndoB in intact hearts from embryonic E11.5 through to post-natal day 7 (P7) and 4 months of age. Wt1 is only expressed either transiently or at low levels in rare, isolated cells within the postnatal epicardium (data not shown). Bars represent mean + s.e.m., N values are numbers of hearts analysed for each group: N=4 (E11.5 and P7), N=3 (4 months). All statistics Student s t-test. WWW.NATURE.COM/NATURE 1

Supplementary Figure 2. Tβ4 primed Wt1+ cells give rise to cardiac progenitors ex vivo. Primed GFP+ progenitors (white arrowheads) migrated from epicardial explants (explant highlighted by white asterisk; a). Clusters of migrating GFP+ cells (b) at higher magnification (white asterisk in c, d). GFP+ cells co-stained for Isl-1 (e, f; white arrowhead in f indicates an Isl-1 negative EPDC surrounded by an Isl-1+ cluster) and 2 WWW.NATURE.COM/NATURE

Nkx2.5 (g, h; white arrowheads indicate Nkx2.5+ cells) provided evidence of cardiac progenitor formation. At day 6 of culture (d6) explant-derived GFP+ cells were observed which co-stained for ctnt (i, j), SαA (l, m) and MYBPC (o, p) as evidence of maturation of progenitors. Importantly, vehicle-treated cells failed to adopt a myocardial fate and only isolated fibroblast-derivatives were observed in control cultures (not shown). At day 14 (d14) derivatives were observed with more punctate GFP expression and evidence of ctnt+ (k), SαA+ (n) MYBPC+ (q) sarcomeric structure along with an elongated rod-shaped phenotype, indicative of mature cardiomyocytes. Cell quantification analysis revealed a significant % of GFP+ cells positive for Isl-1 with a relatively smaller % positive for Nkx2.5 at d6. At d14 a significant % of GFP+ cells also positive for the cardiomyocyte differentiation markers ctnt, SαA and MYBPC was detectable (r). Scale bars in a, m, n, q represent 100µm; in c represents 500µm and in c, d, e, g, i, l and o represent 50µm. Bars represent mean + s.e.m., N values are numbers of cultures analysed for each group: N=4 (d6 and d14). WWW.NATURE.COM/NATURE 3

Supplementary Figure 3. Injury and Tβ4 priming reactivate embryonic epicardial gene expression in the adult heart. After MI alone Wt1 and Tbx18 were significantly increased by day 7 post-injury relative to sham operated control hearts (p*** 0.001) with the up-regulation in expression dependent upon severity of injury: histogram key illustrates short axis sections through representative injured hearts/per injury group (d2: purple=mild injury; cream=severe injury; d7: green=mild injury; yellow=severe injury) with extent of injury highlighted in red (n=4 hearts per injury category per day post-mi) (b). Tβ4 priming resulted in a precocious (relative to injury alone) up-regulation in Wt1 (p*** 0.001) and Tbx18 (p* 0.05) as early as day 2 post-mi, dependent upon severity of injury (categories: purple=mild injury; cream=severe injury; n=4 hearts per MI group. Representative sections shown in b), with levels of both genes returned to sham-operated control levels at day 4 through to day 7 post-mi (c). Bars represent mean + s.e.m., N values are numbers of hearts analysed for each group: N=2 (a, b). All statistics Student s t-test. 4 WWW.NATURE.COM/NATURE

Supplementary Figure 4. Up-regulation of embryonic epicardial gene expression in Tβ4/injury primed FACS isolated GFP+ progenitors and activated Wt1 in situ post- MI. qpcr analyses on progenitors isolated by FACS from the hearts of Wt1-GFP animals at day 4 post-tβ4 treatment and MI revealed significant up-regulation in Wt1, Tbx18 and Raldh2 relative to the GFP- fraction (a). In situ hybridisation for Wt1 on sections through Tβ4-treated hearts at day 4 post MI (b), revealed a significant increase in Wt1 expressing cells residing in the epicardium and sub epicardial region proximal to the scar whereas areas of non-wt1 expressing cells could be distinguished in the underlying myocardium (highlighted by black asterices; c). In remote myocardium, Wt1 expression was confined to the epicardium post-tβ4/injury (highlighted by black arrowheads; d). Black boxes in b, are shown at high power in c and d. Scale bar in b, represents 1mm; c and d represents 500µm. ep, epicardium, sc, scar. Bars represent mean + s.e.m., N values are numbers of hearts analysed for each group: N=6 (a); p** 0.01, p*** 0.001; all statistics Student s t-test. WWW.NATURE.COM/NATURE 5

Supplementary Figure 5. Elevated numbers of GFP+ and YFP+ Progenitors in whole hearts following Tβ4 priming and injury relative to Tβ4-primed without tamoxifen controls. 6 WWW.NATURE.COM/NATURE

FACS analyses scatter plots of GFP+ (a, b) and YFP+ (c, d) at day 7 post-mi following priming of Wt1-GFP and Wt1-ERT2YFP mice, respectively, with PBS vehicle (co; a, c) or Tβ4 (b, d). Note injury alone resulted in a modest detection of both GFP+ and YFP+ cells (a, c) and the numbers of both GFP+ and YFP+ cells were significantly augmented by Tβ4 priming (GFP+ and YFP+ p 0.001; n=3 hearts per PBS versus Tβ4 treatment). P-values calculated by Student s t-test. Analyses of GFP+ and YFP+ cell counts, detected by α-gfp immunostaining at day 7, revealed a reduced number of YFP+ cells relative to GFP+ cells in situ (** p 0.01), however, the % of Wt1+ cells within each labelled population was equivalent (e). A reduced frequency of pulse-labelled YFP+ cells, compared to constitutively labelled GFP+ cells, is consistent with that observed in the developing heart 4 and likely due to inefficient CreERT2 activation by tamoxifen when administered at non-toxic concentrations. An important further control with respect to the Wt1-ERT2YFP model was PBS-treatment or Tβ4-priming in the absence of tamoxifen. This resulted in an expected failure to flow sort YFP+ cells (data not shown) and confirmed Tβ4 priming did not result in a non-specific cellular recombination. In Wt1-ERT2YFP hearts primed with Tβ4, but without tamoxifen treatment, we were unable to flow sort YFP+ cells but observed rare, isolated YFP+ cells in the epicardium in response to injury, at day 2 post-mi as detected by α-gfp immunostaining (f; YFP+ cells highlighted throughout by white arrowheads). The YFP+ cells remained in the epicardium through to days 4 (g) and 7 post-mi (h, i); without migrating into the underlying subepicardial space or myocardium and failed to differentiate into cardiovascular derivatives (i). ep, epicardium; my, myocardium; ses, subepicardial space. Scale bars f-i represent 100 µm. Bars represent mean + s.e.m., N values are numbers of hearts analysed for each group: N=9 (GFP+) and N=6 (YFP+) (e). All statistics Student s t-test. WWW.NATURE.COM/NATURE 7

Supplementary Figure 6. Tβ4/injury responsive progenitors are c-kit negative but contain a significant Sca-1 positive sub-fraction. Single cell suspensions from hearts of Wt1 GFPCre/+ mice treated with Tβ4 were prepared 8 WWW.NATURE.COM/NATURE

by enzymatic digestion 4 days post-mi and incubated with antibodies against the stem cell markers, c-kit (a-e) or Sca-1 (f-j) and analyzed by flow cytometry. Scatter plots revealed GFP+ cells (a, c, f, h) which were negative for c-kit relative to secondary antibody alone (b, d) but revealed a significant sub-fraction (~80%) which were positive for Sca-1 relative to background (g, i). The c-kit negative and Sca-1 positive GFP+ populations were also plotted in the representative histograms showing relative number of cells (% Max) expressing the relevant level of signal. Cells incubated with secondary antibody alone, as a negative control, are shown in black and cells incubated with antibodies against c-kit and Sca-1 are shown in green (e, j). WWW.NATURE.COM/NATURE 9

Supplementary Figure 7. Proliferation and expansion of GFP+ and YFP+ cells within the epicardium and sub-epicardial regions following Tβ4 priming/injury. Ki67+ cells, at low magnification (a) and higher magnification of white box inset (b), proliferated within the epicardium and subepicardial regions (highlighted by white arrowheads in b) after 2 days post-mi when primed by Tβ4. YFP+ cells (c) co-stained for 10 WWW.NATURE.COM/NATURE

PHH3 (d, e) proliferated within the epicardium (white arrowheads in e) after Tβ4/injury priming (c-f). Cell quantification reveled an increase in total numbers of GFP+ and YFP+ cells with injury compared to sham operated control hearts from days 2, 4, 7 (p** 0.01) to day 14 (p*** 0.001 post-mi; n=4 hearts per sham and MI group; g). Total cell number increased over time as verified by spatiotemporal immunostaining with α- GFP. Rarely were YFP+ cells detectable in the absence of injury and were spatially restricted to the epicardium (h). Progressive increases in YFP+ cells were observed across days 2, 4 and 7, with evidence of migration into the underlying myocardium (i-k). YFP+ cells were detected throughout the myocardium in regions proximal to the site of injury and scar (l). Multi-photon-derived 3D reconstructions revealed the extent of migration of YFP+ progenitors from the epicardial surface to underlying myocardium in response to injury and Tβ4 (m). Scale bar in a represents 1mm; b represents 50µm c, represents 50µm applies to c-f; l represents 50 µm applies to h-l. ep, epicardium, my, myocardium; sc, scar region. Bars represent mean + s.e.m., N values are numbers of hearts analysed for each group: N=4 (d2 and d4) and N=7 (d7 and d14) (g). All statistics Student s t-test. WWW.NATURE.COM/NATURE 11

Supplementary Figure 8. Evidence of Nkx2.5+/YFP+ cardiac progenitors. Immunostaining with α-gfp (a) and α-nkx2.5 (b) revealed double positive cells (c; highlighted by white arrowheads) residing in the epicardium 2 days post-mi, indicative of Wt1+-derived cardiac progenitors in response to Tβ4/injury priming. Note the cells 12 WWW.NATURE.COM/NATURE

residing in the epicardium have large nuclei relative to their cytoplasm as is common to progenitor populations (see merge in c highlighting GFP+ cytoplasm against GFP+/Nkx2.5+ nuclei). Real time qrt-pcr analyses failed to reveal increases in Nkx2.5 or the additional early cardiac progenitor marker Gata4 across days 2, 4 and 7 post-mi (categories: blue=sham operated controls; purple=mild injury; cream=severe injury; n=4 hearts per group, representative sections as for Supplementary Fig. 1b) despite the evident increase in progenitor cell numbers (see Fig. 2m) (e). Similarly qrt-pcr did not reveal any change in markers of mature cardiomyocytes, ctnt or MyBPC at day 7 post- MI despite evidence of the addition of de novo YFP+ cells staining positive for ctnt (Fig. 3f, g) (f). The lack of increase in expression of early and late cardiac markers when measured across the entire heart reflects the pre-existing high levels of these genes in resident myocardium, masking the effect of a contribution via de novo progenitor/myocytes. Scale bar in a represents 50µm and applies to a-d. ep, epicardium; my, myocardium. Bars represent mean + s.e.m., N values are numbers of hearts analysed for each group: N=2 (g, f). WWW.NATURE.COM/NATURE 13

Supplementary Figure 9. BrdU pulse chase identifies mitotically active GFP+ progenitors at day 4 traced to BrdU+/GFP+ cardiomyocytes at day 14 post-mi. Sections through hearts of Wt1 GFPCre/+ animals injected with BrdU on days 0, 2 and 4 post-mi revealed BrdU+ progenitor-like cells residing in the epicardium at day 4 as determined by α-brdu immunostaining (a) which were also GFP+ as determined by α- GFP immunostaining (b). White arrowheads in (a) highlight BrdU+ cells located in the epicardium, which were positive for GFP as indicated in (d); white asterisk in (a) indicates a cell positive for BrdU located in underlying myocardium which was GFP-. At day 14 post-mi BrdU+ cardiomyocytes, by virtue of staining for SαA, were detected within the myocardium, residing in the border zone and scar region, which were also GFP+ (e; as highlighted by white arrowheads). Higher magnification view of two BrdU+/GFP+ cells in the scar region with evident sarcomeric banding as an indicator of a mature cardiomyocyte phenotype (f; as highlighted by white arrowheads). bz, border zone; ep, epicardium; my, myocardium; sc, scar. Scale bars in d represents 20µm, in e represents 50 µm and in f represents 20µm.. 14 WWW.NATURE.COM/NATURE

Supplementary Figure 10. YFP+ EPDC-derivatives residing in the border zone of injury and associated with coronary vessels. Multi-photon imaging and rhod-2 loading at day 14 post-mi (a) revealed Tβ4-primed maturing YFP+ rod-shaped cells, which did not support calcium transients, residing in the border zone of injury (white arrowheads highlight YFP+ cells; b). Merge of YFP+/rhod-2 illustrates disrupted fibres (green) at site of injury (c). YFP+ cells were observed in close proximity to and/or contributing to coronary vessels (d-e) with putative pericyte-like (d, e; highlighted by white arrowheads), endothelial-like (white arrowhead; f) or fibroblast-like appearances (based on cell morphology and localization; white asterisk highlights cell cluster; f). Scale bars in a and f represent 20 µm; scale bars in d and e represent 10 µm. bv, blood vessel; bz, border zone; mi, myocardial infarction. WWW.NATURE.COM/NATURE 15

Supplementary Figure 11. Wt1+ progenitor-derived cardiomyocytes contribute to improved cardiac output and myocardial regeneration. MRI analyses with representative short axis images (mid ventricular slice and one slice 1.5 mm offset towards the apex) of infarcted hearts 28 days after surgery with late gadolinium enhancement following treatment with either Tβ4 (a) or PBS (b). Infarcted myocardium (scar tissue) appears bright on late gadolinium enhancement images due to gadolinium accumulation. Scatter plots for end diastolic volume (EDV; c), end systolic volume (ESV; d) and infarct volume (e) for PBS (blue, n=5) and TB4 (red, n=4) treated animals 28 days after surgery. The horizontal bars indicate the means for each group. All three parameters were significantly lower in TB4-treated versus PBS-treated groups indicative of reduced scarring, remodeling and increased cardiac function. At the early time point, 7 days post-mi, we observed an increase in ejection fraction (EF) in Tβ4 treated animals (36 + 4%) compared to the control PBS treated group (27 + 6%). This was consistent with an early role for Tβ4 in maintaining cardiomyocyte survival as 16 WWW.NATURE.COM/NATURE

previously reported (Supplementary Table 1). Measurements at 28 days post-mi revealed end diastolic volumes (EDV) and end systolic volumes (ESV) for PBS treated animals (EDV, 173.9 ± 28.4µl and ESV, 138.1 ± 28.6 µl; n=5) which were significantly higher compared to Tβ4 treated animals (EDV, 89.8 ± 5.1µl; n=4, p 0.05 and ESV,52.8 ± 5.0µl; n=4, p 0.05; c, d)). These data revealed significantly reduced pre- and afterloading, respectively, on the injured heart with Tβ4 treatment, which was progressively lowered over time (Supplementary Table 1). Reduction in loading represented a prelude to decreased ventricular distension and remodelling (a, b) and a significant improvement in ejection fraction (EF) in the TB4 treated animals (41 ± 3%; n=4) compared to the PBS treated controls (23 ± 3%; n=5; p 0.01; repeated measures one way ANOVA; Supplementary Table 1). Scar volume, as assessed via late gadolinium enhancement, was significantly higher in PBS treated mice (43.3 ± 4.9µl; n=5) compared to those that received TΒ4 (22.0 ± 5.3µl; n=4; p 0.05), suggesting that the Tβ4 priming reduced infarct volume (e). In addition, % infarct relative to LVM was significantly reduced in Tβ4 treated (18 + 4%; n=4) as compared to PBS treated (27 + 2; n=5; p 0.05) animals (Supplementary Table 1) indicating that Tβ4-priming not only reduced scarring but also increased ventricular myocardial mass. LVM can be interpreted as an index of hypertrophy, with respect to normal pathology post-mi, but when viewed in the context of reduced infarct volume and % scarring may indicate increased myocardial cell number. Trichrome stained transverse sections (cut at the level of the ventricular papillary muscles) from PBS- (f) and Tβ4- (g) treated hearts, 28 days post MI; red represents viable myocardium and blue represents collagen deposition indicative of scarring and fibrosis. Note the increased proportion of healthy myocardium highlighted by the black box in (g) relative to the comparative region in (f). * p 0.05; all statistics by repeat measures one way ANOVA. Scale bar in g represents 1mm applies to f, g. WWW.NATURE.COM/NATURE 17

Supplementary Table 1. Functional cardiac parameters from MRI analyses of TB4 and PBS primed animals following MI 7 days post surgery 14 days post surgery 28 days post surgery TB4 (n=4) PBS (n=5) TB4 (n=4) PBS (n=5) TB4 (n=4) PBS (n=5) body weight [g] 27.0 ± 3.3 26.3 ± 0.9 28.1 ± 3.7 27.3 ± 0.8 27.4 ± 2.7 27.5 ± 0.9 heart rate [bmp] 552 ± 29 561 ± 11 558 ± 19 570 ± 27 547 ± 24 559 ± 13 left ventricle EDV [µl] 93.4 ± 11.6 135.2 ± 18.2 101.1 ± 15.0 153.3 ± 23.3 89.0 ± 5.1* 173.9 ± 28.4 ESV [µl] 61.2 ± 11.9 102.2 ± 21.5 67.9 ± 13.1 114.7 ± 25.6 52.8 ± 5.0* 138.1 ± 28.6 SV [µl] 32.2 ± 1.2 33.0 ± 3.7 33.2 ± 2.0 38.6 ± 2.7 36.2 ± 2.1 35.8 ± 0.4 EF [%] 36 ± 4 27 ± 6 34 ± 3 28 ± 5 41 ± 3** 23 ± 3 CO [ml/min] 17.8 ± 1.3 18.7 ± 2.4 18.5 ± 0.9 22.0 ± 2.0 19.9 ± 1.7 20.0 ± 0.5 LVM [mg] 129 ± 7 171 ± 10 132 ± 9 152 ± 10 128 ± 6 166 ± 7 infarct Vol. [µl] 29.2 ± 5.8 53.2 ± 10.0 28.1 ± 3.7 42.6 ± 7.2 22.0 ± 5.3* 43.3 ± 4.9 infarct [% LVM] 24 ± 5 32 ± 5 23 ± 4 29 ± 4 18 ± 4* 27 ± 2 #data presented as mean ± standard error of mean. Asterices indicate significant differences between TB4 and PBS treated:* p 0.05; **p 0.01; repeated measures one way ANOVA. 18 WWW.NATURE.COM/NATURE