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

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

SUPPLEMENTARY INFORMATION

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

Supplementary Figures

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

Supplementary Figure 1. Genotyping strategies for Mcm3 +/+, Mcm3 +/Lox and Mcm3 +/- mice and luciferase activity in Mcm3 +/Lox mice. A.

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

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

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

Zhu et al, page 1. Supplementary Figures

Supplementary Figure 1. AdipoR1 silencing and overexpression controls. (a) Representative blots (upper and lower panels) showing the AdipoR1 protein

Supplementary Information

Supplementary Materials for

GFP/Iba1/GFAP. Brain. Liver. Kidney. Lung. Hoechst/Iba1/TLR9!

SUPPLEMENTARY INFORMATION

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

Diabetic pdx1-mutant zebrafish show conserved responses to nutrient overload and anti-glycemic treatment

Supplemental Information. Ciliary Beating Compartmentalizes. Cerebrospinal Fluid Flow in the Brain. and Regulates Ventricular Development

Supplemental Information. Otic Mesenchyme Cells Regulate. Spiral Ganglion Axon Fasciculation. through a Pou3f4/EphA4 Signaling Pathway

Lack of cadherins Celsr2 and Celsr3 impairs ependymal ciliogenesis, leading to fatal

SUPPLEMENTARY INFORMATION

Supplementary Table 1. List of primers used in this study

Supplementary Figure 1 Madm is not required in GSCs and hub cells. (a,b) Act-Gal4-UAS-GFP (a), Act-Gal4-UAS- GFP.nls (b,c) is ubiquitously expressed

Ahtiainen et al., http :// /cgi /content /full /jcb /DC1

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

Inhibition of DYRK1A stimulates human beta-cell proliferation

Supporting Online Material for

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

Genesis of cerebellar interneurons and the prevention of neural DNA damage require XRCC1.

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

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

SUPPLEMENTARY INFORMATION

Vikas Gupta, Matthew Gemberling, Ravi Karra, Gabriel E. Rosenfeld, Todd Evans, and Kenneth D. Poss

Figure S1. (A) Schematic diagram of dnrar transgene allele. (B) X-Gal staining of testis from

Title: Epigenetic mechanisms underlying maternal diabetes-associated risk of congenital heart disease

Supplemental Figure 1. Egr1 expression in adult Achilles tendons. (A,B) Achilles tendons were isolated from 2 month-old Egr1 +/- mice and stained for

Supplemental Information. Figures. Figure S1

Supplementary Figure 1. Properties of various IZUMO1 monoclonal antibodies and behavior of SPACA6. (a) (b) (c) (d) (e) (f) (g) .

Supplementary Figure 1: Hsp60 / IEC mice are embryonically lethal (A) Light microscopic pictures show mouse embryos at developmental stage E12.

SUPPLEMENTARY INFORMATION

SUPPLEMENTARY INFORMATION

Genome-editing via Oviductal Nucleic Acids Delivery (GONAD) system: a novel microinjection-independent genome engineering method in mice

Macrophages form functional vascular mimicry channels in vivo. SI Figures and Legend

Supplemental Figure 1: Leydig cells are reduced at multiple stages in both male sterile mutants

(A) PCR primers (arrows) designed to distinguish wild type (P1+P2), targeted (P1+P2) and excised (P1+P3)14-

Supplementary Materials for

Supplemental Figure 1. Western blot analysis indicated that MIF was detected in the fractions of

a) Primary cultures derived from the pancreas of an 11-week-old Pdx1-Cre; K-MADM-p53

Supplementary Information

SUPPLEMENTARY LEGENDS...

Nature Medicine doi: /nm.2860

Supplementary Information. Supplementary Figure 1

SUPPLEMENTARY INFORMATION

Supporting Information Table of Contents

McWilliams et al., http :// /cgi /content /full /jcb /DC1

Supplemental Figure S1. Expression of Cirbp mrna in mouse tissues and NIH3T3 cells.

Supplementary Figure 1. Chimeric analysis of inner ears. (A-H) Chimeric inner ears with fluorescent ES cells and (I,J) Rainbow inner ears.

Supplementary Figure 1

Retinoid Signaling in Progenitors Controls Specification and Regeneration of the Urothelium

Specimen. Humeral Head. Femoral Head. Objective. Femoral Condyle (medial) Supplementary Figure 1

Supplemental Information. Metabolic Maturation during Muscle Stem Cell. Differentiation Is Achieved by mir-1/133a-mediated

Nature Neuroscience: doi: /nn Supplementary Figure 1

Supplementary Information

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

Supplementary Information. Cofilin Regulates Nuclear Architecture through a Myosin-II Dependent Mechanotransduction Module

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

(a) Significant biological processes (upper panel) and disease biomarkers (lower panel)

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

hexahistidine tagged GRP78 devoid of the KDEL motif (GRP78-His) on SDS-PAGE. This

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

Supplementary Figure 1 Expression of Crb3 in mouse sciatic nerve: biochemical analysis (a) Schematic of Crb3 isoforms, ERLI and CLPI, indicating the

Nature Medicine: doi: /nm.4322

Supplementary Figure 1

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

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 INFORMATION

Soft Agar Assay. For each cell pool, 100,000 cells were resuspended in 0.35% (w/v)

(Stratagene, La Jolla, CA) (Supplemental Fig. 1A). A 5.4-kb EcoRI fragment

SUPPLEMENTARY FIGURES AND TABLE

Supporting Information

EPIGENETIC RE-EXPRESSION OF HIF-2α SUPPRESSES SOFT TISSUE SARCOMA GROWTH

Prss56, a novel marker of adult neurogenesis in the mouse brain. - Supplemental Figures 1 to 5- Brain Structure and Function

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

SUPPLEMENTARY INFORMATION. Rett Syndrome Mutation MeCP2 T158A Disrupts DNA Binding, Protein Stability and ERP Responses

Downregulation of the small GTPase SAR1A: a key event underlying alcohol-induced Golgi fragmentation in hepatocytes

Nature Biotechnology: doi: /nbt Supplementary Figure 1

Supplementary Table 1. The primers used for quantitative RT-PCR. Gene name Forward (5 > 3 ) Reverse (5 > 3 )

CD3 coated cover slips indicating stimulatory contact site, F-actin polymerization and

Nature Immunology: doi: /ni Supplementary Figure 1. Gene expression profile of CD4 + T cells and CTL responses in Bcl6-deficient mice.

Type of file: PDF Size of file: 0 KB Title of file for HTML: Supplementary Information Description: Supplementary Figures

Supplemental Figure 1. Quantification of proliferation in thyroid of WT, Ctns -/- and grafted

Nature Neuroscience: doi: /nn Supplementary Figure 1. Neuron class-specific arrangements of Khc::nod::lacZ label in dendrites.

Supplementary Figure 1

SUPPLEMENTARY INFORMATION

Supplementary Figure 1. Expression of phospho-sik3 in normal and osteoarthritic articular cartilage in the knee. (a) Semiserial histological sections

Supplementary Figure 1. Generation of knockin mice expressing L-selectinN138G. (a) Schematics of the Sellg allele (top), the targeting vector, the

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

SUPPLEMENTARY INFORMATION

Nature Immunology: doi: /ni eee Supplementary Figure 1

Supplementary Figure 1 Transcription assay of nine ABA-responsive PP2C. Transcription assay of nine ABA-responsive PP2C genes. Total RNA was isolated

Transcription:

Developmental Cell, Volume 44 Supplemental Information Myocardial Polyploidization Creates a Barrier to Heart Regeneration in Zebrafish Juan Manuel González-Rosa, Michka Sharpe, Dorothy Field, Mark H. Soonpaa, Loren J. Field, Caroline E. Burns, and C. Geoffrey Burns

Figure S1. Quantitative strategies to measure cardiomyocyte nucleation and relative DNA content, Related to Figure 1 and STAR Methods. (A-H) Double transgenic reporters allow myocardial cell

aggregates to be distinguished from binucleated cardiomyocytes. Fluorescent images of cardiomyocytes dissociated from double transgenic Tg(cmlc2:nucGFP; cmlc2:mkate-caax) hearts. Myocardial cell aggregates (A-D) can be clearly distinguished from binucleated cardiomyocytes (E-H). (A,E) Fluorescent images showing overlapping nuclear GFP and DAPI signals. (B,F) Flourescent images showing membrane localized mkate signal. (C,G) Merged images of (A,B) and (E,F), respectively. (D,H) Pseudocolored masks based on mkate-caax signal highlight cell contours revealing the presence of either three mononucleated cardiomyocytes (D) or a binucleated primordial cardiomyocyte (H). (I-M) Strategy used to quantify DNA content in cardiomyocytes from homeostatic hearts using non-myocardial cells as the diploid reference population. (I) Representative image of cells from dissociated Tg(cmlc2:nucGFP) ventricles (as shown in Figure 1B), showing GFP and DAPI signals. (I ) Magnification of boxed area in (I). Cardiomyocytes are GFP+ rod-shaped cells with bright nuclei (white arrow). Non-myocardial cells (i.e. fibroblasts, epicardial and endocardial cells) are GFP- (white arrowhead). (J,K) Individual fluorescent channels showing GFP and DAPI signals. (L) Nuclear masks were generated based on DAPI signal. Cardiomyocyte and non-cardiomyocyte nuclear masks appear in green and white, respectively. (M) For each nucleus, the integrated density (ID) of the DAPI signal was obtained. The average ID value of noncardiomyocytes (reference population, ID rp, red) was determined for every picture. This value represents the average DNA content of the diploid population (2c). Then, the ID value of each cell from both populations was divided by the ID rp and multiplied by 2. The resulting value represents the DNA content of a given cell relative to the reference population. See STAR Methods section for further details. (N-S) Strategy used to quantify DNA content in cardiomyocytes from regenerating hearts using homeostatic cardiomyocytes as the reference diploid population. Double transgenic Tg(cmlc2:nucGFP; cmlc2:mkate-caax) animals were subjected to apical resection. Because polyploidization of non-myocardial cells from amputated hearts cannot be ruled out, injured ventricles were dissociated in combination with uninjured ventricles from Tg(ubb:Zebrabow) animals, that express mcherry in all cells. mcherry+ ubb:zebrabow cardiomyocytes were used as the reference diploid population. (N) Representative image of cells from dissociated injured Tg(cmlc2:nucGFP; cmlc2:mkate-caax) and uninjured Tg(ubb:Zebrabow) ventricles, showing mcherry, GFP and DAPI signals. (N ) Magnification of boxed area in (N). Cardiomyocytes from regenerating hearts are nucgfp+ (white arrows) and cardiomyocytes from homeostatic hearts are mcherry+ (pink arrows). (O- Q) Individual fluorescent channels showing mcherry, GFP, and DAPI signals. Asterisks in (O) indicate faint signal from mkate-caax in regenerating cardiomyocytes. (R) Nuclear masks were generated based on DAPI signal. Regenerating cardiomyocyte and homeostatic cardiomyocyte nuclear masks appear in green and red, respectively. (S) For each nucleus, the ID of the DAPI signal was obtained. The average ID value of the mcherry+ reference population (ID rp, red) was determined for every picture. This value represents the average DNA content of the diploid population (2c). Then, the ID value of each cell was divided by the ID rp and multiplied by 2. The resulting value represents the DNA content of a given cell relative to the reference diploid population. (T-Y) Validation of our quantification strategy using the MetaMorph Cell Cycle Module. (T)

Representative image of dissociated cells from Tg(cmlc2:nucGFP) ventricles at 7 days post-resection. Animals were injected with 10 µm EdU 4 hours before dissociation to label proliferating cells. (U) Individual fluorescent channels showing EdU signal. Boxed areas are shown at higher magnification. Yellow arrow indicates an EdU+ cardiomyocyte nucleus. (V-W) Ploidy measurement of image shown in (T) using MetaMorph software. (V) Color-coded masks automatically generated by the MetaMorph Cell Cycle Module based on DAPI integrated density. (W) DNA content histogram generated by MetaMorph. The software identified a cell with double the DNA content compared to the rest of the population (black arrow). (X,Y) Ploidy measurement of image shown in (T) using the quantification strategy described above. (X) Nuclear masks generated manually based on DAPI signal. Cardiomyocyte and non-cardiomyocyte nuclear masks appear in green and white, respectively. (Y) DNA content histogram generated by the quantification strategy described above. This method identified a cell with double the DNA content compared to the rest of the population (black arrow). Scale bars: 50 µm.

Figure S2. Ect2 is downregulated during mouse cardiomyocyte maturation, Related to Figure 1. Data extracted from publically available sources (O'Meara et al., 2015). Ect2 declines at the onset of cardiomyocyte polyploidization (P7) while other cell cycle-related transcripts (mki67, pcna) remain unchanged. Samples were obtained from whole ventricles. *, p<0.05, unpaired t-test. A, adult; P, postnatal day.

Figure S3. The retroviral insertion hi3820atg results in disrupted splicing of ect2 mrna, Related to Figure 2. (A) Schematic representation of the Danio rerio ect2 genomic locus and transcript variants. The hi3820atg (nlacz-gt) retroviral integration site is located in the first intron. The retroviral sequence includes splice acceptor sequence and the nlacz cdna. Boxed area shows at higher magnification the first three exons and the location of real-time PCR primers used to detect correct splicing of ect2 mrna. (B) Bright-field images of control (ect2 +/+ ) and ect2 hi3820atg/ hi3820atg (ect2 -/- ) embryos at 72 hpf. Mutant embryos exhibit smaller heads and curly tails. (C,D) Real-time PCR amplification (C) and dissociation (D) curves of rps11 (housekeeping control) and ect2 in ect2 +/+ and ect2 -/- embryos, revealing splicing disruption in ect2 mutant embryos. Scale bar: 200 µm.

Figure S4. Ect2 is required for cardiomyocyte cytokinesis, but dispensable for entry into S-phase, and constitutive myocardial dnect2 expression induces cardiomyocyte hypertrophy, Related to Figure 2. (A,B) Confocal projections of embryonic hearts of ect2+/+ (A) and ect2-/- (B) animals carrying the Tg(cmlc2:nucDsRed) transgene at 30 hpf. Single confocal planes of boxed regions are shown at higher magnification with Alcama immunostaining to highlight plasma membranes. White and yellow arrows point to diploid and polyploid cardiomyocytes, respectively. (C) Quantification of indicated cardiomyocyte populations from ect2+/+ (n=4) and ect2-/- (n=4) hearts at 30 hpf (mean±s.d; ** P<0.01 by two-tailed unpaired t-test). (D,E) Single confocal planes of 72 hpf Tg(cmlc2:nucGFP) (D) and Tg(cmlc2:GdnEct2) (E) animals exposed to BrdU between 48 and 72 hpf, immunostained for GFP and BrdU and counterstained with DAPI. Yellow arrowheads indicate BrdU+ cardiomyocyte nuclei. The BrdU signals from (D) and (E) are shown below. (F) Quantification of the Tg(cmlc2:nucGFP) (n=4) and Tg(cmlc2:GdnEct2) (n=4) cardiomyocyte BrdU labeling index at 72 hpf (mean ± s.d, non-significant by two-tailed unpaired t-test). (G,H) Ventricular sections from 30 dpf non-tg (G) or Tg(cmlc2:GdnEct2) animals (H) carrying the Tg(cmlc2:mKate-CAAX) transgene to label cardiomyocyte cell membranes. Hearts were immunostained to detect mkate and GFP and

counterstained with DAPI. Boxed regions are shown at higher magnifications (right and insets). Individual fluorescent channels showing mkate and DAPI signals (bottom) as indicated. Pseudocolored cells depicting the cross-sectional area of individual cardiomyocytes. (I) Quantification of cardiomyocyte cross-sectional area in non-tg (n=1996 total cardiomyocytes from 3 animals) and cmlc2:gdnect2 (n=688 total cardiomyocytes from 5 animals). Mean values and standard deviations are shown in red. ****, P<1 10-15, Mann-Whitney test. Scale bars: 25 µm (A-B, D-E) 100 µm (G-H), 50 µm (magnifications from G and H).

Figure S5. Transgenic strategy used to create mosaic zebrafish hearts composed of GFP- diploid and GFP+ polyploid-enriched cardiomyocyte populations, Related to Figure 3, Figure 4 and STAR Methods. (A) In untreated hearts, the cmlc2:(if-s) lox -mg transgene (top) directs the expression of a loxp-

flanked cassette that contains a nuclear-localized morange2 fluorescent protein and a tamoxifen-inducible Flipase (FlpOER T2 ). The hsp:(p*-s) FRT -Cre-dnEct2 transgene (bottom), which only becomes activated in response to heat-shock, directs expression of a FRT-flanked cassette that contains a mutated, nonfluorescent nuclear-localized PhiYFP protein (nucphiyfp*, abbreviated P*). The presence of the loxp- and FRT-flanked cassettes prevents the expression of downstream genes. Therefore, untreated animals carrying both transgenes express nuc-morange2 and FlpOER T2 specifically in cardiomyocytes. Heat-shock treatment induces the expression of nucphiyfp*. (B,C) Exposure to 4-HT during development induces FlpOER T2 -mediated recombination of the FRT-flanked cassette (in the hsp:(p*-s) FRT -Cre-dnEct2 transgene) randomly in some cardiomyocytes. (B) Cardiomyocytes that undergo FlpOER T2 -mediated recombination and are exposed to heat-shock express Cre and dnect2. As a consequence, Cre catalyzes the recombination of the loxp-flanked cassette in the cmlc2:(if-s) lox -mg transgene resulting in cardiomyocytes that are permanently labeled by GFP. Heat-shock treatment induces expression of Cre and dnect2. (C) Cardiomyocytes that do not undergo FlpOER T2 -mediated recombination and are exposed to heat-shock express nucphiyfp* but do not express Cre and dnect2. In the absence of Cre, the cmlc2:(if-s) lox -mg transgene remains unrecombined and cardiomyocytes are not labeled by GFP. GFP- cardiomyocytes do not express Cre or dnect2 in response to heat-shock. This binary system creates mosaic hearts containing GFP- and GFP+ populations, the latter being susceptible to polyploidization through dnect2-mediated cytokinesis inhibition. (D) Embryos containing mosaic hearts are initially grown in the absence of heat-shock to allow the expansion of the GFP+ and GFP- populations. When animals are 2-3 weeks old, they are exposed to daily heat-shocks for the following 3-4 months. Heat-shock induces the expression of dnect2 in the GFP+ population, inhibiting cytokinesis. Consequently, the GFP+ population becomes enriched in polyploid cells while the GFP- population remains diploid.

Figure S6. Myocardial dnect2 expression does not induce DNA damage or apoptosis, and exposure to dnect2 during adulthood does not induce cardiomyocyte polyploidization, Related to Figure 3

and Figure 4. (A-D) Sections from hearts from the indicated cohorts, immunostained for γh2ax (to detect DNA damage), GFP and counterstained with DAPI. γh2ax signals are shown in bottom panels. Boxed regions are shown at higher magnifications (insets). Irradiated animals (B) serve as a positive control for the DNA damage response. Yellow arrowheads in insets indicate cardiomyocyte nuclei. (E-G) TUNEL staining in sections from hearts from the indicated cohorts, immunostained for GFP and counterstained with DAPI. TUNEL signal is shown in bottom panels. Section in (E) was incubated in DNAse I as a positive control for apoptosis. (H) External appearance of double-transgenic adult zebrafish, subjected to early recombination during embryogenesis, grown in the absence of polyploid-inducing heat-shock treatments and exposed to one week of daily heat-shock treatments during adulthood. (I,J) DNA content (I) and quantification of indicated cardiomyocyte populations (J) from (H). n=2839 total cardiomyocytes from 3 replicates, 3 ventricles per replicate. Scale bars: 50 µm (A-G, K), 5 mm (H).

Figure S7. Ect2 is re-expressed after injury in polyploid-enriched hearts, related to Figure 5 and Figure 6. (A,C) RNAScope in situ hybridization showing ect2 transcripts (yellow arrowheads) in a section

from the indicated cohorts at 7 dpr, immunostained for GFP and counterstained with DAPI. Asterisk indicates the wound area. (B,D) Magnified regions from (A) and (D). Boxed region is shown to the right with individual fluorescent signals for DAPI, GFP and ect2 mrna. n=4 hearts per group with 3 sections per heart. Scale bars: 50 µm (A,C), 10 µm (B,D).

Figure S8. Constitutive Ect2 expression in the myocardium of mouse hearts is insufficient to increase cardiomyocyte proliferation after injury, Related to Figure 5 and 6. (A) Transgene used to overexpress murine Ect2 in mouse cardiomyocytes. (B) Sections from ventricles from non-tg and MHCEct2 transgenic siblings, immunostained for Ect2, Tropomyosin and counterstained with Hoechst (top). Ect2 signal is shown in bottom panels. Yellow arrowheads indicate Ect2+ cardiomyocyte nuclei. (C) Experimental strategy used to evaluate cardiomyocyte proliferation after myocardial infarction (MI) in mouse. All mice carried the MHC-nLacZ transgene to identify cardiomyocyte nuclei. (D) Representative section of a MHCnLacZ+, MHC-Ect2+ animal at 14 days post-infarction, immunostained for β-galactosidase, BrdU and counterstained with DAPI. White circles highlight BrdU+ ventricular cardiomyocyte nuclei. (E) Ventricular cardiomyocyte BrdU labeling indices in non-tg (n=116,457 cardiomyocytes scored from 5 ventricles, 2 sections per ventricle) and MHC-Ect2+ (n=122,571 cardiomyocytes scored from 4 ventricles, 2 sections per ventricle) animals. Scale bar: 50 µm.