• No results found

Analysis of Cre-mediated genetic deletion of Gdf11 in cardiomyocytes of young mice

N/A
N/A
Protected

Academic year: 2021

Share "Analysis of Cre-mediated genetic deletion of Gdf11 in cardiomyocytes of young mice"

Copied!
40
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

University of Groningen

Analysis of Cre-mediated genetic deletion of Gdf11 in cardiomyocytes of young mice Garbern, Jessica; Kristl, Amy C; Bassaneze, Vinicius; Vujic, Ana; Schoemaker, Henk; Sereda, Rebecca; Peng, Liming; Ricci-Blair, Elisabeth M; Goldstein, Jill M; Walker, Ryan G

Published in:

American Journal of Physiology - Heart and Circulatory Physiology DOI:

10.1152/ajpheart.00615.2018

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from it. Please check the document version below.

Document Version

Final author's version (accepted by publisher, after peer review)

Publication date: 2019

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

Garbern, J., Kristl, A. C., Bassaneze, V., Vujic, A., Schoemaker, H., Sereda, R., Peng, L., Ricci-Blair, E. M., Goldstein, J. M., Walker, R. G., Bhasin, S., Wagers, A. J., & Lee, R. T. (2019). Analysis of Cre-mediated genetic deletion of Gdf11 in cardiomyocytes of young mice. American Journal of Physiology - Heart and Circulatory Physiology. https://doi.org/10.1152/ajpheart.00615.2018

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

(2)

1 2

Analysis of Cre-mediated genetic deletion of Gdf11 in cardiomyocytes of young mice

3 4 5

Jessica C. Garberna,b, Amy C. Kristla, Vinicius Bassanezea,c, Ana Vujica, Henk Schoemakerc, 6

Rebecca Seredaa, Liming Pengd, Elisabeth Ricci-Blaira, Jill M. Goldsteina, Ryan G. Walkera, 7

Shalender Bhasind, Amy J. Wagersa,e,f, Richard T. Leea,c

8 9 10

Author contributions: 11

J.C.G. Designed and conducted experiments, wrote manuscript; A.C.K. Designed and conducted 12

experiments; V.B. Designed and conducted experiments, edited manuscript; A.V. Designed and 13

conducted experiments, edited manuscript; H.S. Designed and conducted experiments; R.S. 14

Conducted experiments; L.P. Conducted experiments; E.R-B. Conducted experiments; J.M.G. 15

Designed experiments, edited manuscript; R.G.W. Designed experiments, edited manuscript; 16

S.B. conducted experiments; A.J.W. Designed experiments, edited manuscript; R.T.L. Designed 17

experiments, edited manuscript 18

19 20

Author affiliations: 21

aDepartment of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute,

22

Harvard University, 7 Divinity Ave, Cambridge, MA 02138 23

bDepartment of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA

24

02115 25

cDivision of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital

26

and Harvard Medical School, 75 Francis St, Boston, MA 02115 27

dBrigham Research Assay Core, Brigham and Women’s Hospital, 221 Longwood Ave, Boston,

28

MA 02115 29

ePaul F. Glenn Center for the Biology of Aging, Harvard Medical School, 77 Louis Pasteur

30

Avenue, Boston, MA 02115 31

fSection on Islet Cell and Regenerative Biology, Joslin Diabetes Center, One Joslin Place,

32

Boston, MA 02215 33

34

Abbreviated title: Gdf11 deletion in cardiomyocytes 35 36 Corresponding author: 37 Dr. Richard T. Lee 38

Department of Stem Cell and Regenerative Biology 39 Harvard University 40 7 Divinity Ave 41 Cambridge, MA 02138 42 Phone: 617-496-5394 43 Fax: 617-496-8351 44 richard_lee@harvard.edu 45

(3)

Abstract

46

Administration of active growth differentiation factor 11 (GDF11) to aged mice can reduce 47

cardiac hypertrophy, and low serum levels of GDF11 measured together with the related protein, 48

myostatin (also known as GDF8), predict future morbidity and mortality in coronary heart 49

patients. Using mice with a loxP-flanked (“floxed”) allele of Gdf11 and Myh6-driven expression 50

of Cre recombinase to delete Gdf11 in cardiomyocytes, we tested the hypothesis that cardiac-51

specific Gdf11 deficiency might lead to cardiac hypertrophy in young adulthood. We observed 52

that targeted deletion of Gdf11 in cardiomyocytes does not cause cardiac hypertrophy but rather 53

leads to left ventricular dilation when compared to control mice carrying only the Myh6-cre or 54

Gdf11-floxed alleles, suggesting a possible etiology for dilated cardiomyopathy. However, the

55

mechanism underlying this finding remains unclear due to multiple confounding effects 56

associated with the selected model. First, whole heart Gdf11 expression did not decrease in 57

Myh6-cre;Gdf11-floxed mice, possibly due to upregulation of Gdf11 in non-cardiomyocytes in

58

the heart. Second, we observed Cre-associated toxicity, with lower body weights and increased 59

global fibrosis in Cre-only control male mice compared to flox-only controls, making it 60

challenging to infer which changes in Myh6-cre;Gdf11-floxed mice were due to Cre toxicity 61

versus deletion of Gdf11. Third, we observed differential expression of cre mRNA in Cre-only 62

controls compared to the cardiomyocyte-specific knockout mice, also making comparison 63

between these two groups difficult. Thus, targeted Gdf11 deletion in cardiomyocytes may lead to 64

left ventricular dilation without hypertrophy, but alternative animal models are necessary to 65

understand the mechanism for these findings. 66

67 68

(4)

New and Noteworthy

69

We observed that targeted deletion of Gdf11 in cardiomyocytes does not cause cardiac 70

hypertrophy but rather leads to left ventricular dilation when compared to control mice carrying 71

only the Myh6-cre or Gdf11-floxed alleles. However, the mechanism underlying this finding 72

remains unclear due to multiple confounding effects associated with the selected mouse model. 73

74

Keywords: Growth differentiation factor 11, myostatin, Myh6-cre, cardiomyocytes,

75

cardiomyopathy 76

(5)

Introduction

77

Growth differentiation factor 11 (GDF11) is a member of the transforming growth factor 78

β (TGF-β) superfamily, best known for its morphogenic roles during development (14). The role 79

of GDF11 in cardiac aging has been controversial (9, 21, 24, 25). We initially demonstrated that 80

GDF11 is a circulating factor that when administered to aged mice can decrease age-related 81

cardiac hypertrophy as indicated by a decrease in heart weight to tibia length ratios (12). Others 82

have shown that GDF11 administration may be beneficial following myocardial infarction (6). 83

However, following our initial report, contrasting work indicated that GDF11 administration to 84

aged mice does not affect heart weight to body weight ratios (24), or, that GDF11 85

supplementation impairs cardiac function in concert with reducing cardiomyocyte size and 86

cardiac mass (21). Furthermore, additional controversy has been raised due to a lack of 87

specificity of antibody and aptamer reagents used to detect and quantify GDF11 separate from 88

the closely related protein, myostatin (also known as GDF8 and encoded by the gene, Mstn). 89

Because GDF11 and GDF8 share approximately 90% identity in their active domains, it has been 90

challenging to quantify levels of each protein independently (7). 91

Human clinical data indicate that low serum levels of the GDF11 + myostatin pool at 92

study entry predict increased mortality in adult patients with heart disease over the subsequent 93

eight years (16). These human data point to low levels of the circulating pool of GDF11 + 94

myostatin as a potential pathogenic factor in human heart disease. However, conflicting human 95

data have also been published suggesting that higher levels of GDF11 in adults with severe aortic 96

stenosis are associated with an increased risk of adverse events following valve replacement 97

surgery (22). Thus, further studies are needed to understand how perturbations to the GDF11 and 98

myostatin system might contribute to cardiovascular risk. 99

(6)

The effect of GDF11 deficiency on the heart also remains unclear, but prior studies have 100

evaluated the effect of myostatin deficiency on the heart. Mstn-/- mice are viable and have 101

increased skeletal muscle weight as well as increased heart weight and heart weight to tail length, 102

but they show no increase in heart weight to body weight ratio (10). Mstn-/- mice also have 103

mildly increased left ventricular volumes and mildly reduced systolic function at baseline but 104

have an enhanced response to isoproterenol stress with a higher percent change in fractional 105

shortening compared to wild type mice (10). Inducible cardiomyocyte-specific deletion of Mstn 106

leads to transient chamber dilation and impairment of systolic function; however, upregulation of 107

Mstn in non-cardiomyocytes occurs leading to an unexpected overall increase in Mstn expression

108

in whole heart extracts of knockout mice (3). Conversely, overexpression of Mstn leads to 109

decreased heart weights in males but not females (20). Despite similarities in sequence between 110

mature myostatin and GDF11, there are structural differences between the two proteins that 111

could lead to distinct effects with deletion of Gdf11 in cardiomyocytes that have not previously 112

been described with cardiomyocyte-specific Mstn deletion (25). 113

We sought to test the effects of reduced levels of GDF11 in the heart during young 114

adulthood in mice. Because systemic germline deletion in the mouse (Gdf11-/-) results in a

115

number of developmental defects and is lethal by 1 day of age, likely due to renal agenesis (14), 116

we generated a mouse model to specifically delete Gdf11 only within cardiomyocytes. We chose 117

to delete Gdf11 from cardiomyocytes for our initial study as opposed to other cardiac cell types 118

given the demonstrated phenotype seen with cardiomyocyte deletion of Mstn and challenges of 119

deleting Gdf11 only in the heart if targeting other cell types. By crossing a cardiomyocyte-120

specific Myh6-cre allele with a Gdf11-floxed allele (13), we were able to induce genomic 121

excision of the regions encoding mature GDF11 protein exclusively in cardiomyocytes. Our 122

(7)

experiment was designed to test the hypothesis that postnatal Gdf11 deficiency in 123

cardiomyocytes would promote cardiac hypertrophy. We found instead that targeted 124

cardiomyocyte deletion of Gdf11 during young adulthood, using the Myh6-cre system, does not 125

result in cardiac hypertrophy and rather leads to progressive left ventricular dilation that is 126

apparent in both females and males by 6 months of age. However, because of multiple adverse 127

effects from Cre recombinase itself on the heart and potential differential expression of the cre 128

gene across genotypes, we are unable to define the molecular mechanism of the dilated 129

cardiomyopathy phenotype that develops when the Gdf11 gene is removed from cardiomyocytes 130

using this mouse model. 131

132

Materials and Methods

133 134

Animals – constitutively active Cre

135

Mice expressing constitutively active Cre recombinase driven by the Myh6 promoter 136

(B6.FVB-Tg(Myh6-cre)2182Mds/J) were obtained from The Jackson Laboratory. Mice 137

containing a floxed (flanking loxP) Gdf11 allele with loxP sites flanking exons 2 and 3 of Gdf11 138

were generously provided by Dr. Se-Jin Lee (Johns Hopkins University) (13). Mice were bred to 139

obtain three genotypes used in this study: Myh6cre/wt; Gdf11fl/fl (experimental genotype), 140

Myh6cre/wt; Gdf11wt/wt (Cre-only control genotype, referred to as Myh6cre/wt), and Myh6wt/wt;

141

Gdf11fl/fl (flox-only control genotype and littermate control, referred to as Gdf11fl/fl). All mice

142

were on a mixed C57Bl/6J and 6N background. 143

Young adult male and female mice of all three genotypes were weighed weekly starting 144

at 2 months of age. Echocardiograms were performed in all mice at 1-2 and 6 months of age in a 145

(8)

blinded manner. Additional echocardiogram time points at 3 or 4 months of age were also 146

performed in a subset of mice. Mice were observed for up to 6 months, and then animals were 147

euthanized and serum and tissues harvested for further analysis. We also determined tibia length 148

as a normalizing parameter because GDF11 administration has been shown to decrease body 149

weight (17). Tissues for downstream DNA, RNA and protein analysis were flash frozen in liquid 150

nitrogen and stored at -70˚C until further processing was performed. All experiments were 151

conducted according to the Guide for the Use and Care of Laboratory Animals and approved by 152

the Institutional Animal Care and Use Committee of Harvard University Faculty of Arts and 153

Sciences. 154

155

Animals – tamoxifen-inducible Cre

156 157

Mice expressing tamoxifen-inducible Cre recombinase (Mer-Cre-Mer, abbreviated 158

MCM) driven by the Myh6 promoter (B6.FVB(129)-A1cfTg(Myh6-cre/Esr1*)1Jmk/J) were obtained

159

from The Jackson Laboratory (backcrossed to a C57Bl/6J agouti background per vendor). Mice 160

were bred with Gdf11fl/fl mice to obtain two genotypes used in this experiment: Myh6MCM/wt; 161

Gdf11fl/fl (experimental genotype), and Myh6wt/wt; Gdf11fl/fl (flox control genotype). Male and

162

female mice were stratified based on gender then randomized at 5 months of age to receive 163

injections of either 4-hydroxytamoxifen (4OH-tamoxifen, 75 mg/kg) or vehicle (10% ethanol, 164

90% sunflower oil) via intraperitoneal (i.p.) injection for 5 days. Echocardiography was 165

performed at baseline (at 5 months of age) and 4 weeks later (at 6 months of age) prior to harvest 166

to evaluate left ventricular size and function. Tissues were harvested and processed for histology 167

or qPCR. 168

(9)

Echocardiography

170

Mice were sedated with 0.1-0.5% inhaled isoflurane for echocardiography, with the dose 171

titrated to maintain heart rates of >500 beats per minute for acquired images. Mice were placed 172

on a heating pad, and echocardiograms were obtained with the Vevo770 (Visualsonics, Toronto, 173

Ontario, Canada). M-mode was used to measure left ventricular (LV) interventricular septal 174

(IVS) wall thickness, LV posterior wall thickness (LVPW), and LV internal diameter (LVID) 175

during both systole and diastole. Fractional shortening (%), LV mass and LV volumes were 176

calculated with the Visualsonics software package. 177

178

Cardiomyocyte and non-cardiomyocyte isolation from adult hearts

179

Cardiomyocytes and non-cardiomyocytes were isolated from adult male and female mice 180

using a Langendorff-free method as previously described (1). We modified the original protocol 181

to include blebbistatin (5 µM, Sigma) instead of 2-3-butanedione monoxime in the culture 182

media. In addition, we used a peristaltic pump rather than hand injection to better control the 183

flow rate of the perfused solutions. Briefly, mice were anesthetized with isoflurane then the chest 184

cavity was opened. The descending aorta and inferior vena cava were cut followed by perfusion 185

of 7 mL of EDTA buffer into the apex of the right ventricle. The aorta was then clamped and cut 186

distal to the clamp, and the heart was removed. EDTA buffer (10 mL) was then perfused into the 187

apex of the left ventricle followed by injection of 3 ml of perfusion buffer then 30-40 mL of 188

collagenase buffer delivered into the left ventricular apex. The clamp was removed and the heart 189

was manually dissociated. Stop buffer (perfusion buffer + 5% fetal bovine serum) was added, 190

cells were passed through a 300 µm strainer then cardiomyocytes were allowed to gravity settle 191

for 20 minutes. The supernatant containing non-cardiomyocytes and debris was plated into an 192

(10)

uncoated tissue culture plate in DMEM:F12/10% FBS for 3 hours. The cardiomyocyte fraction in 193

the pellet then underwent sequential gravity settling with low speed centrifugation (12 g x 3 min) 194

with calcium re-introduction followed by plating into laminin-coated plates for 3 hours. After 3 195

hours, both cardiomyocytes and non-cardiomyocytes were washed and harvested in Trizol. 196

Samples were frozen at -70 deg C until RNA extraction by QIAcube. 197

198

PCR

199

DNA was extracted from harvested tissue using the REDExtract-N-Amp Tissue PCR Kit 200

(Sigma). Table 1 shows the primers that were used to amplify Myh6, Myh6-cre and Gdf11 PCR 201

products and their expected product sizes. PCR products were run on a 2% agarose gel in TAE 202

buffer at 100V for 45-60 minutes and gels were imaged in a Gel Doc EZ system (Bio-Rad). 203

204

Quantitative PCR

205

RiboZol reagent (VWR) and the E.Z.N.A. Total RNA I kit (Omega) were used to isolate 206

RNA from homogenized whole organ tissue, followed by the High Capacity cDNA Reverse 207

Transcription kit (Thermo Fisher Scientific) to reverse transcribe mRNA to cDNA according to 208

the manufacturers’ instructions. Quantitative PCR (qPCR) was performed using TaqMan probes 209

for Nppb (Mm01255770_g1), Gdf11 (Mm01159973_m1, spanning exons 1-2), Gdf15 210

(Mm00442228_m1), Inhba (activin A, Mm00434339_m1), Mstn (Mm01254559_m1), and 211

Tgfbr1 (Mm00436964_m1), with TATA-binding protein (Tbp) used as the housekeeping gene

212

(Mm01277042_m1) using a Bio-Rad CFX384 Real-Time System. Due to low yields of isolated 213

cardiomyocytes and non-cardiomyocytes from adult hearts, RNA was extracted using the Qiagen 214

QIACube from Trizol (Thermo) followed by reverse transcription to cDNA using the 215

(11)

SuperScript VILO cDNA synthesis kit (Thermo) per the manufacturer’s instructions. The 216

isolated cDNA (10-20 ng) was then pre-amplified using the Taqman PreAmp Master Mix 217

(Thermo) for 14 cycles. The pre-amplified cDNA was then diluted 1:20 for quantification of 218

Gdf11 expression in isolated cardiomyocytes and non-cardiomyocytes by qPCR on the

219

QuantStudio 6 Flex Real-Time PCR System (Applied Biosystems). 220

221

Flow cytometry

222

Flow cytometry was performed to quantify the percentage of cardiomyocytes in the cell 223

population obtained immediately after the final step fo the extraction procedure. In brief, the 224

cardiomyocyte fraction was fixed with 1 mL of 70% ethanol then stored at -20 deg C. The non-225

cardiomyocyte fraction was plated into a six-well plate and allowed to expand for seven days in 226

DMEM/F12 + 10% fetal bovine serum + 1% penicillin/streptomycin. Cells were dissociated with 227

trypsin then fixed in 70% ethanol. The fixative was removed via low speed centrifugation (20 g x 228

3 min) and the cells were washed then permeabilized in 0.1% Triton in PBS. Cells were labeled 229

overnight at 4 deg C using an antibody to cardiac troponin T (Abcam ab8295, diluted 1:250 in 230

phosphate buffered saline (PBS) supplemented with 10% goat serum) to detect cardiomyocytes, 231

and an antibody to vimentin (Abcam ab92547, diluted 1:200 in PBS supplemented woith 10% 232

goat serum), which is highly expressed in fibroblasts. After washing steps and incubation with 233

the corresponding AlexaFluor 568 conjugated secondary antibody for 2h at room temperature 234

(Thermo, Mouse IgG1 and Rabbit IgG, respectively), cells were analyzed by flow cytometry 235

(MoFlo Astrios, Beckman Coulter, using the nozzle of 200µm). Data were analyzed with FlowJo 236

software (version 10.0.8). 237

(12)

239

Histology

240

Harvested tissues were fixed in 4% paraformaldehyde for 24 hours then exchanged for 241

70% ethanol for 3 days prior to embedding in paraffin. Sections were stained with Masson’s 242

trichrome staining as previously described (8). Global fibrosis was quantified using a Python 243

script written to quantify the percentage of blue pixels out of the total pixels from a cross-244

sectional image of the heart (https://github.com/jgarbern/global-fibrosis). Average 245

cardiomyocyte cross-sectional area for each mouse was quantified in a blinded manner with a 246

total of 40 cardiomyocytes measured from multiple sections from each heart. 247

248

Quantitative mass spectrometry

249

Blood was obtained by retro-orbital collection at the time of harvest and transferred to 250

serum separator tubes (SST). Tubes were spun at 2000 g for 5 min and serum was transferred to 251

clean low binding microcentrifuge tubes and stored at -70 deg C until further processing. 252

Samples (100 µL) were submitted to the Brigham Research Assay Core (BRAC) at Brigham and 253

Women’s Hospital for quantitative mass spectrometry. Mouse serum was denatured, reduced and 254

alkylated, followed by pH based fractionation using cation ion exchange solid phase extraction 255

(SPE); appropriate elution fraction was digested with trypsin. After desalting and concentrating 256

of tryptic digest, the peptide mixture was separated and eluted by liquid chromatography 257

followed by mass spectrometric analysis operated in positive electrospray ionization mode. The 258

most intensive and unique proteotypic peptides from GDF11 and myostatin as surrogated 259

peptides along with heavy-labeled unique peptides as internal standards were used for 260

quantitative determination of GDF11 and myostatin. 261

(13)

262

Statistical analysis

263

Data are expressed as mean ± standard error of the mean (SEM) unless noted otherwise. 264

Data were evaluated with the D’Agostino and Pearson omnibus normality test; normally 265

distributed data were evaluated with Student’s t-test or two-way ANOVA with Tukey post-hoc 266

analysis, while data that were not normally distributed were analyzed with Mann-Whitney or 267

Kruskal-Wallis with Dunn’s multiple comparisons test as indicated. Kaplan-Meier survival 268

curves were evaluated the Mantel-Cox log-tank test. Body weight trends with time were 269

analyzed with a two-way repeated measures ANOVA with Tukey multiple comparisons test. A 270

p-value < 0.05 was considered statistically significant. 271

272

Results

273 274

Cardiomyocyte-specific deletion of Gdf11 leads to left ventricular dilation that progresses with

275

age

276

Due to known sexual dimorphism in body weight and heart size in mice, we analyzed 277

body weight, heart weight, and heart weight/body weight ratio data from males and females 278

separately. Myh6cre/wt; Gdf11fl/fl mice had progressive left ventricular dilation with a significant 279

increase in left ventricular end diastolic volume (Figures 1A (1-2 months), 1B (3-4 months), and 280

1C (6 months)), a significant decrease in septal thickness (Figure 1J) and a non-significant 281

decrease in left ventricular posterior wall thickness (Figure 1L) by echocardiography that was 282

apparent in both genders by 6 months of age. Male Myh6cre/wt; Gdf11fl/fl mice also had 283

significantly increased left ventricular internal diameters at 6 months of age compared to sex-284

(14)

matched Myh6cre/wt mice (Figure 1K). Cardiomyocyte-specific deletion of Gdf11 also led to a 285

decrease in left ventricular function with decreased fractional shortening in Myh6cre/wt; Gdf11fl/fl 286

females compared to sex-matched Gdf11fl/fl controls, and in Myh6cre/wt; Gdf11fl/fl males when 287

compared to sex-matched Myh6cre/wt controls (Figure 1G). The chamber dilation and functional 288

decline were not associated with differences in either estimated left ventricular mass by 289

echocardiography, or heart weight compared to either the Cre-only control genotype (Myh6cre/wt) 290

or the flox-only control genotype (Gdf11fl/fl) at 6 months of age (Figure 1H, 1I, 1F, respectively). 291

Heart weight to body weight ratios were not different across groups at any age (Figures 1D (1-2 292

months), 1E (3-4 months), and 1F (6 months)). There were no significant differences in tibia 293

lengths or heart weight to tibia length ratios at 6 months of age (Figures 1N and 1O, 294

respectively). 295

We also utilized a tamoxifen-inducible cardiomyocyte deletion model with 296

administration of 4OH-tamoxifen or vehicle to 6 month old Gdf11fl/fl or Myh6MCM/wt; Gdf11fl/fl 297

mice for 5 days at 75 mg/kg delivered intraperitoneally, but saw no significant differences in left 298

ventricular end diastolic volume, estimated left ventricular mass, body weight, heart weight, or 299

heart weight to body weight ratio with this treatment regimen (Figures 2E, 2F, 2G, 2I, 2J 300

respectively) despite evidence of recombination by PCR (Figure 2C). We did not see significant 301

changes in Gdf11 mRNA expression following this treatment regimen by qPCR (Figure 2D). 302

However, we observed significant differences in survival across all groups in males (Figure 2B) 303

but not females (Figure 2A). The etiology for the survival difference in 4OH-tamoxifen-treated 304

males is unclear, although we presume it reflects tamoxifen toxicity given the acute nature of the 305

deaths. Survival of only the “healthiest” mice may also confound our long-term results. 4OH-306

tamoxifen significantly increased tibia length, therefore we did not evaluate heart weight to tibia 307

(15)

length ratios in this model (Figure 2H); the effect of tamoxifen on long bone length has been 308

reported previously (26). We also found significantly increased activin A (Inbha) mRNA 309

expression within the spleens of 4OH-TAM treated Myh6MCM/wt; Gdf11fl/fl male mice, and a 310

similar non-significant trend in other TAM-treated groups (Figure 2K), consistent with prior 311

reports that tamoxifen may activate TGF-β signaling (4, 5, 15). We did not study this model in 312

further depth due to the absence of a cardiac phenotype at the age selected (6 months of age). 313

We note that the different findings observed between tamoxifen-inducible Myh6-MCM 314

versus constitutively active Myh6-cre models may be due to the different ages at which Gdf11 315

knockdown occurs in the two models. In addition, application of stress such as with pressure 316

overload could be considered to enhance any potential phenotype caused with tamoxifen-induced 317

Myh6-MCM expression. In the context of increased mortality in mice injected with 4OH-TAM 318

even without surgery, and concerns for confounding effects from 4OH-TAM administration on 319

TGF-β signaling, we chose not to apply pressure overload stress (e.g. transverse aortic 320

constriction model) or repeat this experiment at a younger age in the tamoxifen-inducible 321

cardiomyocyte deletion model and instead focused on the constitutively active Myh6-cre model 322

for the remainder of this study. 323

324

Cardiomyocyte-specific deletion of Gdf11 is not sufficient to decrease Gdf11 mRNA

325

expression in the whole heart

326

Myh6 Cre-induced genomic recombination of Gdf11 was evident in the heart in neonatal

327

mice (Figure 3A). Gdf11 recombination was observed only in the heart by PCR and, as expected, 328

was not seen in the lung, liver, kidney, spleen, or skeletal muscle at 6 months of age (Figure 3B). 329

However, despite evidence of DNA recombination, we did not observe differences in Gdf11 330

(16)

mRNA expression in the whole heart at 6 months of age (Figure 3D), and in fact saw a 331

significant increase in Gdf11 mRNA expression in Myh6cre/wt; Gdf11fl/fl male mice at 3 months of 332

age (Figure 3C). 333

Since approximately half of the whole heart is composed of non-cardiomyocytes 334

(including endothelial, fibroblast and smooth muscle cells) (2), we isolated cardiomyocytes and 335

non-cardiomyocytes from adult (>2 months) mice to evaluate Gdf11 mRNA expression in the 336

two cell populations. In a representative batch, the cardiomyocyte population consisted of 84% 337

cTnT+ cardiomyocytes, while the non-cardiomyocyte population consisted of 95% vimentin+ 338

fibroblasts, as quantified by flow cytometry (data not shown). Gdf11 expression shows 339

substantial variability in Myh6cre/wt mice in this analysis; however, we detected a decreased signal 340

in cardiomyocytes from Myh6cre/wt; Gdf11fl/fl mice and a trend toward differences in Gdf11 341

expression in cardiomyocytes across genotypes (p-value 0.05 by Kruskal-Wallis test) (Figure 342

3E). 343

The high variability in Gdf11 expression in Myh6cre/wt mice suggested that this genotype 344

may be abnormal, which became evident in subsequent analysis. The Gdf11 signal in Myh6cre/wt; 345

Gdf11fl/fl mice was greater than zero likely due to contamination from non-cardiomyocytes in the

346

cardiomyocyte population, though it is possible this reflects incomplete recombination of the 347

floxed alleles. We did not observe any differences in Gdf11 expression in non-cardiomyocytes 348

across genotypes (Figure 3F). Direct comparison across cell types is challenging due to different 349

amounts of RNA per cell in different cell types. If starting with an equal amount of RNA, there 350

was a trend toward increased Gdf11 expression in non-cardiomyocytes compared to 351

cardiomyocytes in all genotypes (data not shown). However, we obtained much lower total RNA 352

after isolation from non-cardiomyocytes (predominantly cardiac fibroblasts) compared to an 353

(17)

equal number of cardiomyocytes, and therefore comparing Gdf11 expression from the same 354

amount of total RNA may be suboptimal. 355

Due to challenges in quantifying protein levels of GDF11 with antibody-based 356

approaches (7), we measured GDF11 and myostatin levels in serum by quantitative mass 357

spectrometry and found that Gdf11fl/fl females had significantly lower circulating levels of 358

GDF11 than Myh6cre/wt females; however, there were no significant differences in GDF11 serum 359

concentrations between Myh6cre/wt; Gdf11fl/fl mice and their sex-matched Cre- or flox-only 360

control groups (Figure 3G). Although the major source(s) of circulating GDF11 remains unclear, 361

prior work from our group demonstrated that the spleen has the highest mRNA levels among 362

organs studied (12); thus it is not unexpected that serum levels of GDF11 were not altered in 363

Myh6cre/wt; Gdf11fl/fl mice, as only cardiomyocyte Gdf11 was targeted for deletion in these

364

animals. Circulating myostatin levels did not differ across groups (Figure 3H). 365

366

Mechanism of left ventricular dilation confounded by adverse effects from Myh6-driven Cre

367

recombinase activity

368

Myh6cre/wt male mice were significantly smaller than both Gdf11fl/fl and Myh6cre/wt;

369

Gdf11fl/fl mice by 6 months of age (Figure 1M), with a plateau in the body weight curve in

370

Myh6cre/wt mice appearing at around 4 months of age in both genders (Figures 4C-D). In addition,

371

there was a non-significant trend toward decreased survival in Myh6cre/wt mice with death in 372

several mice at around 4 months of age (Figures 4A-B). 373

There was a significant increase in cardiac mRNA expression of Mstn in Myh6cre/wt;

374

Gdf11fl/fl mice compared to Myh6cre/wt mice at 2 months (Figure 5A), but not 6 months of age

375

(Figure 5E). In contrast, at 6 months but not 2 months of age, Nppb, which encodes B-type 376

(18)

natriuretic peptide (BNP), a clinically used marker of heart failure, had significantly higher 377

expression in hearts of Myh6cre/wt mice compared to Gdf11fl/fl mice, with the experimental 378

genotype (Myh6cre/wt; Gdf11fl/fl) having an intermediate expression profile (2 months, Figure 5B; 379

6 months, Figure 5F). Expression of Tgfbr1, which encodes one of the receptors for GDF11, was 380

also significantly lower in hearts of Gdf11fl/fl mice compared to both Myh6cre/wt mice and 381

Myh6cre/wt; Gdf11fl/fl mice at 6 months (Figure 5G) but not 2 months of age (Figure 5C). Finally,

382

expression of Gdf15, which was reported to be upregulated following administration of 383

supraphysiologic doses of GDF11 (11), was significantly increased in hearts of Myh6cre/wt mice 384

compared to both Gdf11fl/fl mice and Myh6cre/wt; Gdf11fl/fl mice at 6 months (Figure 5H) but not 2 385

months of age (Figure 5D). These results suggest that cardiomyocyte deletion of Gdf11 has an 386

early effect on Mstn expression, which precedes left ventricular dilation, and is followed later by 387

differential regulation on other proteins involved in TGF-β signaling. However, the differences 388

between the two control genotypes particularly at older ages suggest that the presence of Cre 389

recombinase has adverse effects on the heart and make it difficult to identify molecular 390

mechanisms to explain our echocardiographic findings in the experimental genotype. 391

392

Myh6-driven Cre recombinase expression leads to myocardial fibrosis and increased

393

cardiomyocyte size in males

394

Cardiotoxicity has been previously described by Pugach et al. with prolonged Myh6-395

driven Cre expression in this strain (18), and our results are consistent with these prior findings. 396

We observed a significant increase in global myocardial fibrosis in males compared to females in 397

the Cre-only control genotype (Myh6cre/wt) at 6 months of age, with males more sensitive than 398

females to adverse effects from Cre, consistent with Pugach et al. (18) (Figures 6A and B). There 399

(19)

was a similar non-significant trend observed in Myh6cre/wt; Gdf11fl/fl mice. In addition, there was 400

a non-significant trend of increased global myocardial fibrosis in male mice expressing Cre 401

recombinase compared to the flox-only control (Gdf11fl/fl). Furthermore, the cardiomyocyte 402

cross-sectional area was significantly increased in Myh6cre/wt male mice compared to either 403

Gdf11fl/fl or Myh6cre/wt; Gdf11fl/fl mice (Figure 6C).

404 405

Cre mRNA expression is higher in Myh6cre/wt mice compared to Myh6cre/wt; Gdf11fl/fl mice

406

We evaluated whether Cre is differentially expressed across genotypes as a possible 407

explanation for why Myh6cre/wt mice have a more pronounced phenotype in terms of body 408

weight, myocardial fibrosis and expression of selected genes associated with heart failure such as 409

Nppb. We found that cre mRNA expression is significantly greater in Myh6cre/wt mice compared

410

to Myh6cre/wt; Gdf11fl/fl mice (Figure 6D). In contrast to RNA levels, there were no differences 411

noted in Cre protein expression in Myh6cre/wt mice compared to Myh6cre/wt; Gdf11fl/fl mice (Figure 412

6E). No cre protein expression was detected in Gdf11fl/fl mice. 413

414

Discussion

415 416

We observed a significant increase in left ventricular end diastolic volume in mice with 417

Cre-mediated genetic deletion of Gdf11 in cardiomyocytes. This finding was consistent in both 418

males and females, with LV end diastolic volume significantly increased in Myh6cre/wt; Gdf11fl/fl 419

mice compared to both Myh6cre/wt and Gdf11fl/fl control mice. Left ventricular dilation was

420

associated with a decrease in left ventricular systolic function, suggesting a possible role for 421

GDF11 signaling in dilated cardiomyopathy. We observed that Mstn and Gdf11 both transiently 422

(20)

increase in RNA extracted from whole hearts of young adult Myh6cre/wt; Gdf11fl/fl mice prior to 423

the onset of ventricular dilation, which suggests that there is a developmental component to the 424

phenotype observed. However, our experimental design met numerous unanticipated challenges 425

that prohibit clear interpretation of the underlying molecular mechanisms to explain these 426

findings. First, we did not observe a significant decrease in Gdf11 expression in whole heart 427

extracts from Myh6cre/wt; Gdf11fl/fl mice, suggesting that counter-regulation in non-myocytes may 428

buffer cardiomyocyte-specific loss of Gdf11 in the heart. Second, mice expressing Cre 429

recombinase had a different phenotype from mice not expressing Cre, with decreased body 430

weights and increased global myocardial fibrosis in males, an effect only made clear because we 431

utilized two control genotypes. Third, cre mRNA expression was different between the Cre 432

control genotype and the Cre-containing experimental genotype. Although Cre protein levels 433

were not different by western analysis, in the context of Cre-associated toxicity, further study of 434

protein levels at different ages should be performed in future work to elucidate which 435

perturbations to the cardiac system should be attributed to differences in Cre levels versus Gdf11 436

deletion. 437

Although we observed a non-significant trend toward decreased Gdf11 expression in 438

isolated adult cardiomyocytes of Myh6cre/wt; Gdf11fl/fl mice (complete absence of Gdf11 could not 439

be shown likely due to contamination from non-cardiomyocytes), we did not observe a 440

significant decrease in Gdf11 expression in the whole heart despite constitutively active Myh6-441

driven Cre expression and PCR evidence of Gdf11 recombination. In fact, we observed a 442

transient increase in Gdf11 expression in whole heart extracts from male Myh6cre/wt; Gdf11fl/fl

443

mice compared to male Gdf11fl/fl control mice at 3 months of age and a similar non-significant 444

trend compared to male Myh6cre/wt control mice. A similar effect was previously reported when 445

(21)

Mstn was targeted for deletion in cardiomyocytes using mice with tamoxifen-inducible

Myh6-446

driven Cre expression, where Mstn mRNA expression increased in whole heart extracts due to 447

upregulation in non-myocytes despite cardiomyocyte deficiency of myostatin (3). Due to 448

different amounts of total RNA in cardiomyocytes versus non-cardiomyocytes, and low 449

expression levels of Gdf11 in cardiomyocytes requiring pre-amplification of cDNA to detect a 450

reliable signal by qPCR, we were unable to directly compare expression of Gdf11 in 451

cardiomyocytes to non-cardiomyocytes. Nonetheless, in non-cardiomyocytes, we did not observe 452

a difference in Gdf11 expression levels across genotypes. Taken together, this suggests that 453

Gdf11 may act locally in cardiomyocytes, given the presence of a dilated phenotype even in the

454

absence of expression differences at the organ level. In addition, we observed a significant 455

increase in Mstn mRNA expression in whole heart extracts of 2 month old mice. This suggests 456

that Cre-mediated cardiomyocyte deletion of Gdf11 leads to downstream signaling effects during 457

development which precede the observed phenotype of left ventricular dilation starting at 3-4 458

months of age. We also did not examine other organs given the lack of serum differences in 459

GDF11 at 6 months. Alternative models used in future work should focus on early downstream 460

signaling changes in the heart and other organs (such as the spleen which has higher baseline 461

mRNA expression) as well as measuring circulating GDF11 levels at younger ages to better 462

understand how these changes affect cardiac phenotype at later time points. 463

Myh6cre/wt mice have previously been shown to develop progressive myocardial fibrosis

464

and inflammation due to DNA damage at endogenous “loxP-like” or “pseudo-loxP” sites in the 465

myocardium (18). In that study, the authors identified 227 loxP-like sites within genes that could 466

be potentially recognized by Cre recombinase, when tolerating ≤4 mismatches in the canonical 467

loxP sequence. Of these 227 degenerate loxP sites, 55 are expressed in the heart, leading to

(22)

numerous off-target effects from Cre recombinase including myocardial fibrosis and 469

upregulation of apoptotic markers such as p53 and Bax (18). This previous study also found that 470

males appear to be more sensitive to the off-target effects of Cre expression in cardiomyocytes, 471

with an increased heart weight to body weight ratio in Myh6cre/wt compared to wild type control 472

(C57Bl/6J) male mice at 6 months of age (18). Although we did not have wild type mice as a 473

control in our study, we also observed that Myh6cre/wt males had significantly increased global 474

fibrosis compared to females and Myh6cre/wt males had significantly increased cardiomyocyte 475

cross-sectional area compared to Gdf11fl/fl control mice and Myh6cre/wt; Gdf11fl/fl knockout mice.

476

It remains unclear whether the potency of Cre toxicity is identical in the presence or absence of 477

loxP, or whether in the presence of loxP, there might be fewer off-target effects due to the

478

stoichiometry of Cre:loxP versus Cre:loxP-like or pseudo-loxP sites. These results underscore the 479

importance of inclusion of Cre control mice as well as evaluation of both genders when using 480

Cre-lox technology. 481

We utilized both Myh6cre/wt and Gdf11fl/fl controls to attempt to account for adverse

482

effects from Cre recombinase. Our results demonstrate that without inclusion of both controls, 483

our data would have been easily misinterpreted with potentially incorrect conclusions drawn – a 484

lesson that was learned from experience after we failed to include the Myh6MCM/wt control line in 485

the tamoxifen-inducible study described in Results above. For example, without Gdf11fl/fl 486

controls, we may have incorrectly concluded that deletion of Gdf11 leads to a decrease in Nppb 487

expression. Conversely, without Myh6cre/wt controls, we may have incorrectly concluded that 488

deletion of Gdf11 leads to an increase in Nppb expression. However, with inclusion of both 489

control genotypes, we see that there is actually a confounding effect with significant differences 490

in expression of Nppb between Gdf11fl/fl and Myh6cre/wt control mice. It is challenging to

(23)

reconcile these differences among the different genotypes. It appears that the presence of Cre 492

induces stress on the mouse (with increased mortality, decreased body weight, increased Nppb 493

expression, and increased cardiomyocyte size). Comparing the Myh6cre/wt control mice to 494

Myh6cre/wt; Gdf11fl/fl mice, one might conclude that cardiomyocyte deletion of Gdf11 is actually

495

cardioprotective (with increased survival, increased body weight, decreased Nppb expression and 496

decreased cardiomyocyte size). However, other explanations are possible as well, such as 497

differential off target effects in the presence or absence of true loxP sites, or differential 498

expression of Cre in the two genotypes. Given that comparison of Gdf11fl/fl control mice to 499

Myh6cre/wt; Gdf11fl/fl mice leads to different conclusions than when comparing Myh6cre/wt control

500

mice to Myh6cre/wt; Gdf11fl/fl mice, we are unable to definitively determine the mechanisms by 501

which Gdf11 deletion in cardiomyocytes leads to left ventricular dilation. Inclusion of multiple 502

control groups admittedly adds significant costs and time required to maintain a larger animal 503

colony, but careful selection of control groups is necessary to obtain meaningful results. 504

We observed higher cre mRNA but not protein expression in Myh6cre/wt compared to 505

Myh6cre/wt; Gdf11fl/fl mice in this study. Variable cre RNA expression in different generations has

506

been reported in other Cre lines (19, 23). For example, in an albumin-Cre model, despite 507

transmission of albumin-cre in genomic DNA of successive generations, some mice did not 508

express cre in the liver (23). The authors speculated that that this could be due to multiple 509

homologous recombination events leading to segregation of inactive copies of the transgene, 510

binding of transcriptional inhibitors, or post-transcriptional silencing of cre (23). In addition, 511

cytosine methylation of loxP sites following Cre recombination of a parent can lead to inhibition 512

of Cre-mediated recombination in subsequent generations (19). Finally, although all mice were 513

on a mixed C57Bl/6J and 6N background, there may be generational differences due to different 514

(24)

degrees of backcrossing to the underlying genetic background. It is possible that there is a more 515

complex interaction with Cre and the underlying genetic background which will be difficult to 516

uncover. To try to address this confounding effect, we used littermate controls to compare 517

Gdf11fl/fl with Myh6cre/wt; Gdf11fl/fl mice. However, our breeding strategy paired Myh6cre/wt with

518

wild type (C57Bl/6J) mice and Gdf11fl/fl with Myh6cre/wt; Gdf11fl/fl thus Myh6cre/wt were not

519

littermates with Myh6cre/wt; Gdf11fl/fl mice and may have had varying degrees of methylation or 520

varying genetic backgrounds in the breeders. Future work to understand how Cre mRNA and 521

protein levels change with age in both genotypes is necessary to interpret whether variable Cre 522

levels might be confounding the phenotype seen with cardiomyocyte Gdf11 deletion. 523

In conclusion, deletion of the Gdf11 gene from cardiomyocytes may lead to left 524

ventricular dilation and decreased systolic function, consistent with a dilated cardiomyopathy 525

phenotype. However, due to numerous confounding factors associated with the selected, and 526

commonly used, Cre-lox system as well as challenges in working with Gdf11 itself and its 527

complicated regulatory system, we are unable to attribute a mechanism to this phenotype. These 528

data highlight the importance of using appropriate control groups when using the Cre 529

recombinase system, as opposite conclusions could have been drawn had only one of the two 530

control genotypes been used for comparison. Further work to develop alternative animal models 531

that avoid Cre toxicity as well as investigate alternative proteins involved in regulation of 532

GDF11 expression are warranted. 533

534

Acknowledgements

535

The authors acknowledge Catherine MacGillivray and Diane Faria from the Department of Stem 536

Cell and Regenerative Biology Histology Core for histology processing and staining. 537

(25)

538

Sources of Funding

539

J.C.G. was supported by the John S. LaDue Memorial Fellowship in Cardiology from Harvard 540

Medical School and an NIH T32 fellowship (5T32HL007572). V.B. was supported by the 541

Lemann Foundation Cardiovascular Research Postdoctoral Fellowship through Harvard 542

University/Brigham and Women’s Hospital. J.M.G was supported by an NIH F32 postdoctoral 543

fellowship (F32AG050395). R.G.W. was supported by a T32 fellowship from the NIH 544

(T32HL007208-39). This work was supported by grants from the NIH (AG047131, HL119230, 545

AG048917, and AG057428 to RTL and AG048917 and AG057428 to AJW) and from the Glenn 546

Foundation (to AJW). 547

548

Disclosures

549

Richard Lee, Ryan Walker, and Amy Wagers are co-founders of, members of the scientific 550

advisory board for, and hold private equity in Elevian, Inc, a company that aims to develop 551

medicines to restore regenerative capacity. Elevian also provides sponsored research support to 552

the Lee Lab and Wagers Lab. 553

(26)

References

554 555

1. Ackers-Johnson M, Li PY, Holmes AP, O'Brien SM, Pavlovic D, and Foo RS. A

556

Simplified, Langendorff-Free Method for Concomitant Isolation of Viable Cardiac Myocytes 557

and Nonmyocytes From the Adult Mouse Heart. Circ Res 119: 909-920, 2016. 558

2. Banerjee I, Fuseler JW, Price RL, Borg TK, and Baudino TA. Determination of cell

559

types and numbers during cardiac development in the neonatal and adult rat and mouse. Am J 560

Physiol Heart Circ Physiol 293: H1883-1891, 2007.

561

3. Biesemann N, Mendler L, Wietelmann A, Hermann S, Schafers M, Kruger M,

562

Boettger T, Borchardt T, and Braun T. Myostatin regulates energy homeostasis in the heart

563

and prevents heart failure. Circ Res 115: 296-310, 2014. 564

4. Chen H, Tritton TR, Kenny N, Absher M, and Chiu JF. Tamoxifen induces TGF-beta

565

1 activity and apoptosis of human MCF-7 breast cancer cells in vitro. J Cell Biochem 61: 9-17, 566

1996. 567

5. Costantino JP, Kuller LH, Ives DG, Fisher B, and Dignam J. Coronary heart disease

568

mortality and adjuvant tamoxifen therapy. J Natl Cancer Inst 89: 776-782, 1997. 569

6. Du GQ, Shao ZB, Wu J, Yin WJ, Li SH, Wu J, Weisel RD, Tian JW, and Li RK.

570

Targeted myocardial delivery of GDF11 gene rejuvenates the aged mouse heart and enhances 571

myocardial regeneration after ischemia-reperfusion injury. Basic Res Cardiol 112: 7, 2017. 572

7. Egerman MA, Cadena SM, Gilbert JA, Meyer A, Nelson HN, Swalley SE, Mallozzi

573

C, Jacobi C, Jennings LL, Clay I, Laurent G, Ma S, Brachat S, Lach-Trifilieff E,

574

Shavlakadze T, Trendelenburg AU, Brack AS, and Glass DJ. GDF11 Increases with Age and

575

Inhibits Skeletal Muscle Regeneration. Cell Metab 22: 164-174, 2015. 576

(27)

8. Goldner J. A modification of the masson trichrome technique for routine laboratory

577

purposes. Am J Pathol 14: 237-243, 1938. 578

9. Harper SC, Brack A, MacDonnell S, Franti M, Olwin BB, Bailey BA, Rudnicki MA,

579

and Houser SR. Is Growth Differentiation Factor 11 a Realistic Therapeutic for

Aging-580

Dependent Muscle Defects? Circ Res 118: 1143-1150; discussion 1150, 2016. 581

10. Jackson MF, Luong D, Vang DD, Garikipati DK, Stanton JB, Nelson OL, and

582

Rodgers BD. The aging myostatin null phenotype: reduced adiposity, cardiac hypertrophy,

583

enhanced cardiac stress response, and sexual dimorphism. J Endocrinol 213: 263-275, 2012. 584

11. Jones JE, Cadena SM, Gong C, Wang X, Chen Z, Wang SX, Vickers C, Chen H,

585

Lach-Trifilieff E, Hadcock JR, and Glass DJ. Supraphysiologic Administration of GDF11

586

Induces Cachexia in Part by Upregulating GDF15. Cell Rep 22: 1522-1530, 2018. 587

12. Loffredo FS, Steinhauser ML, Jay SM, Gannon J, Pancoast JR, Yalamanchi P,

588

Sinha M, Dall'Osso C, Khong D, Shadrach JL, Miller CM, Singer BS, Stewart A,

589

Psychogios N, Gerszten RE, Hartigan AJ, Kim MJ, Serwold T, Wagers AJ, and Lee RT.

590

Growth differentiation factor 11 is a circulating factor that reverses age-related cardiac 591

hypertrophy. Cell 153: 828-839, 2013. 592

13. McPherron AC, Huynh TV, and Lee SJ. Redundancy of myostatin and

593

growth/differentiation factor 11 function. BMC Dev Biol 9: 24, 2009. 594

14. McPherron AC, Lawler AM, and Lee SJ. Regulation of anterior/posterior patterning of

595

the axial skeleton by growth/differentiation factor 11. Nat Genet 22: 260-264, 1999. 596

15. Nordenskjold B, Rosell J, Rutqvist LE, Malmstrom PO, Bergh J, Bengtsson NO,

597

Hatschek T, Wallgren A, and Carstensen J. Coronary heart disease mortality after 5 years of

(28)

adjuvant tamoxifen therapy: results from a randomized trial. J Natl Cancer Inst 97: 1609-1610, 599

2005. 600

16. Olson KA, Beatty AL, Heidecker B, Regan MC, Brody EN, Foreman T, Kato S,

601

Mehler RE, Singer BS, Hveem K, Dalen H, Sterling DG, Lawn RM, Schiller NB, Williams

602

SA, Whooley MA, and Ganz P. Association of growth differentiation factor 11/8, putative

anti-603

ageing factor, with cardiovascular outcomes and overall mortality in humans: analysis of the 604

Heart and Soul and HUNT3 cohorts. Eur Heart J 36: 3426-3434, 2015. 605

17. Poggioli T, Vujic A, Yang P, Macias-Trevino C, Uygur A, Loffredo FS, Pancoast JR,

606

Cho M, Goldstein J, Tandias RM, Gonzalez E, Walker RG, Thompson TB, Wagers AJ,

607

Fong YW, and Lee RT. Circulating Growth Differentiation Factor 11/8 Levels Decline With

608

Age. Circ Res 118: 29-37, 2016. 609

18. Pugach EK, Richmond PA, Azofeifa JG, Dowell RD, and Leinwand LA. Prolonged

610

Cre expression driven by the alpha-myosin heavy chain promoter can be cardiotoxic. J Mol Cell 611

Cardiol 86: 54-61, 2015.

612

19. Rassoulzadegan M, Magliano M, and Cuzin F. Transvection effects involving DNA

613

methylation during meiosis in the mouse. EMBO J 21: 440-450, 2002. 614

20. Reisz-Porszasz S, Bhasin S, Artaza JN, Shen R, Sinha-Hikim I, Hogue A, Fielder

615

TJ, and Gonzalez-Cadavid NF. Lower skeletal muscle mass in male transgenic mice with

616

muscle-specific overexpression of myostatin. Am J Physiol Endocrinol Metab 285: E876-888, 617

2003. 618

21. Roh JD, Hobson R, Chaudhari V, Quintero P, Yeri A, Benson M, Xiao C, Zlotoff D,

619

Bezzerides V, Houstis N, Platt C, Damilano F, Lindman BR, Elmariah S, Biersmith M, Lee

(29)

SJ, Seidman CE, Seidman JG, Gerszten RE, Lach-Trifilieff E, Glass DJ, and Rosenzweig

621

A. Activin type II receptor signaling in cardiac aging and heart failure. Sci Transl Med 11: 2019.

622

22. Schafer MJ, Atkinson EJ, Vanderboom PM, Kotajarvi B, White TA, Moore MM,

623

Bruce CJ, Greason KL, Suri RM, Khosla S, Miller JD, Bergen HR, 3rd, and LeBrasseur

624

NK. Quantification of GDF11 and Myostatin in Human Aging and Cardiovascular Disease. Cell

625

Metab 23: 1207-1215, 2016.

626

23. Schulz TJ, Glaubitz M, Kuhlow D, Thierbach R, Birringer M, Steinberg P, Pfeiffer

627

AF, and Ristow M. Variable expression of Cre recombinase transgenes precludes reliable

628

prediction of tissue-specific gene disruption by tail-biopsy genotyping. PLoS One 2: e1013, 629

2007. 630

24. Smith SC, Zhang X, Zhang X, Gross P, Starosta T, Mohsin S, Franti M, Gupta P,

631

Hayes D, Myzithras M, Kahn J, Tanner J, Weldon SM, Khalil A, Guo X, Sabri A, Chen X,

632

MacDonnell S, and Houser SR. GDF11 does not rescue aging-related pathological

633

hypertrophy. Circ Res 117: 926-932, 2015. 634

25. Walker RG, Poggioli T, Katsimpardi L, Buchanan SM, Oh J, Wattrus S, Heidecker

635

B, Fong YW, Rubin LL, Ganz P, Thompson TB, Wagers AJ, and Lee RT. Biochemistry and

636

Biology of GDF11 and Myostatin: Similarities, Differences, and Questions for Future 637

Investigation. Circ Res 118: 1125-1141; discussion 1142, 2016. 638

26. Zhong ZA, Sun W, Chen H, Zhang H, Lay YE, Lane NE, and Yao W. Optimizing

639

tamoxifen-inducible Cre/loxp system to reduce tamoxifen effect on bone turnover in long bones 640

of young mice. Bone 81: 614-619, 2015. 641

(30)

Figure Captions

643 644

Figure 1. Targeted cardiac Gdf11 deletion leads to left ventricular dilation. (A-C) Left

645

ventricular end diastolic volume by echocardiography at 1-2 months (A), 3-4 months (B), and 6 646

months of age (C). (D-F) Heart weight/body weight ratio at 1-2 months (D), 3-4 months (E), and 647

6 months of age (F). (G) Fractional shortening by echocardiography at 6 months of age. (H) 648

Estimated left ventricular mass by echocardiography at 6 months of age. (I) Heart weight at 6 649

months of age. (J) Interventricular septal thickness during diastole at 6 months of age. (K) Left 650

ventricular internal diameter during diastole at 6 months of age. (L) Left ventricular posterior 651

wall thickness during diastole at 6 months of age. (M) Body weight at 6 months of age (just prior 652

to harvest). (N) Tibia length at 6 months of age. (O) Heart weight to tibia length ratio at 6 653

months of age. Sample sizes at 1-2 months: Myh6cre/wt (n=5 females, n=9 males), Gdf11fl/fl (n=11 654

females, n=21 males), and Myh6cre/wt; Gdf11fl/fl (n=12 females, n=15 males) mice; n=4/group for 655

heart weight to body weight ratio parameter. Sample sizes at 3-4 months: Myh6cre/wt (n=9 656

females, n=12 males), Gdf11fl/fl (n=9 females, n=13 males), and Myh6cre/wt; Gdf11fl/fl (n=13 657

females, n=14 males) mice for echocardiography; n=4/group for heart weight to body weight 658

ratio parameter. Sample sizes at 6 months: Myh6cre/wt (n=6 females, n=16 males), Gdf11fl/fl (n=3 659

females, n=5 males), and Myh6cre/wt; Gdf11fl/fl (n=6 females, n=9 males) mice, *p<0.05, 660

**p<0.01 by Kruskal-Wallis test with Tukey post-hoc analysis. 661

662

Figure 2. Survival curves, echocardiographic, heart weight, and body weight parameters in mice

663

with tamoxifen-inducible Myh6-driven Cre expression (Myh6-MCM). (A and B) Kaplan-Meier 664

survival analysis in Gdf11fl/fl (gray), and Myh6MCM/wt; Gdf11fl/fl (black) female (A) and male (B) 665

(31)

mice. Male mice have significant differences between groups with p<0.01 by the Mantel-Cox 666

test. n=12-16/group at start of study. (C) Recombination of Gdf11 gene seen in the heart but not 667

liver, kidney, spleen or skeletal muscle following administration of 4-hydroxytamoxifen (75 668

mg/kg i.p. x 5 days) by polymerase chain reaction (PCR). (D) Gdf11 expression in RNA 669

extracted from whole heart 1 month after 4-hydroxytamoxifen administration in female and male 670

mice by quantitative PCR (qPCR), n=3/group. Data are normalized to TATA-binding protein 671

(Tbp) then to female Gdf11fl/fl vehicle control. For echo and harvest data in (E) to (J), n=7-672

16/group, data collected 1 month after initiation of 4OH-tamoxifen injection (6 months of age). 673

(E) Left ventricular end diastolic volume (µl) by echocardiography. (F) Estimated left ventricular 674

mass (mg) by echocardiography. (G) Body weight (g) at harvest. (H) Tibia length, **p<0.01, 675

***p<0.001 by two-way ANOVA. (I) Heart weight and (J) Heart weight/body weight ratio. (K) 676

Activin A mRNA expression in spleen is significantly increased in male Myh6MCM/wt; Gdf11fl/fl 677

mice 1 month after initiation of 4OH-tamoxifen injection (6 months of age), n=4-6/group, 678

*p<0.05 by Kruskal-Wallis with Dunn’s multiple comparisons test. Data are normalized to Tbp 679

then to female Gdf11fl/fl vehicle control. 680

681

Figure 3. Targeted cardiomyocyte deletion of Gdf11 does not decrease total Gdf11 mRNA

682

expression in mouse hearts. (A) Representative agarose gel image depicting Myh6 (wild type 683

band at 894 bp, Myh6-cre band at 300 bp) and Gdf11 (wild type band at 359 bp, flox band at 393 684

bp, and ∆2-3 (post-recombination) band at 300 bp) alleles seen in the heart on day of life 0-1 by 685

polymerase chain reaction (PCR) in Myh6cre/wt, Gdf11fl/fl, and Myh6cre/wt; Gdf11fl/fl pups. (B)

686

Recombination of Gdf11 at 6 months of age in females and males in the heart but not in lung, 687

liver, kidney, spleen or skeletal muscle by PCR. (C and D) Gdf11 expression in RNA extracted 688

(32)

from whole heart in (C) 3 month old (n=4/group) or (D) 6 month old (n=3-4/group) female and 689

male mice by quantitative PCR (qPCR). Data are normalized to TATA-binding protein (Tbp) 690

then to female Myh6cre/wt control. (E and F) Gdf11 expression in RNA extracted from isolated 691

adult (>2 months old) cardiomyocytes (E) or non-cardiomyocytes (F), with each data point 692

(shown with open symbols) representing isolated cells from a single mouse of the same 693

genotype, n=3 per group (2 males, 1 female). Data normalized to Tbp then Myh6cre/wt control. 694

Cardiomyocytes (E), p-value = not significant (n.s., 0.05); non-cardiomyocytes (F), p-value = 695

n.s. (0.3) by Kruskal-Wallis test. (G and H) Serum levels of GDF11 (G) and myostatin (H) as 696

determined by quantitative mass spectrometry in 6-month-old mice. n=3-4/group, *p<0.05 by 697

Kruskal-Wallis test followed by Tukey’s multiple comparisons test. 698

699

Figure 4. Cre recombinase has adverse effects on survival and body weight. (A and B)

Kaplan-700

Meier survival analysis in (A) female and (B) male Myh6cre/wt (dark gray, dashed line, n=12 701

females, n=24 males), Gdf11fl/fl (light gray, dotted line, n=5 females, n=11 males), and 702

Myh6cre/wt; Gdf11fl/fl (black solid line, n=12 females, n=15 males) mice. (C and D) Body weight

703

versus age in weeks in (C) female and (D) male Myh6cre/wt (gray circles with dashed line, n=5

704

females, n=13 males), Gdf11fl/fl (gray squares with dotted line, n=5 females, n=11 males), and 705

Myh6cre/wt; Gdf11fl/fl (black tringles with solid line, n=12 females, n=15 males) mice. *p<0.05,

706

**p<0.01 by two-way repeated measures ANOVA with Tukey’s multiple comparisons test. 707

708

Figure 5. Gene expression analysis of in 2 month old (A-D) and 6 month old (E-H) from whole

709

hearts of (A and E) Mstn, (B and F) Nppb, (C and G) Tgfbr1, and (D and H) Gdf15 by qPCR. 710

Data are normalized to Tbp expression then to Myh6cre/wt control. No significant differences were 711

(33)

observed between genders therefore data represent combined male and female data. n=7-8/group, 712

*p<0.05, **p<0.01, ***p<0.001 by Kruskal-Wallis analysis with Dunn’s multiple comparisons 713

test. 714

715

Figure 6. Cre mRNA expression is higher in Myh6cre/wt compared to Myh6cre/wt; Gdf11fl/fl mice 716

and is associated with myocardial fibrosis and increased cardiomyocyte size. (A) Representative 717

histology sections of male and female Myh6cre/wt, Gdf11fl/fl, and Myh6cre/wt; Gdf11fl/fl mice stained 718

with Masson’s trichrome stain. (B) Global fibrosis (% blue pixels) shows increased fibrosis in 719

male (n=5-9/group) Myh6cre/wt mice compared to females (n=3-5/group). (C) Cardiomyocyte 720

cross-sectional area is increased in male Myh6cre/wt mice compared to male Gdf11fl/fl and 721

Myh6cre/wt; Gdf11fl/fl mice. Cross-sectional area from 40 cardiomyocytes per mouse were

722

averaged for each mouse then average cardiomyocyte cross-sectional area by mouse data were 723

analyzed with n=3=6 female mice, n=5-10 male mice, *p<0.05, **p<0.01 by two-way ANOVA 724

with Tukey post-hoc analysis. (D) Cre expression by qPCR from male and female 3-6 month old 725

mice. No significant differences were detected between genders therefore data depict combined 726

male and female data. Data are normalized to Tbp then to Myh6cre/wt control. n=16/group,

727

*p<0.05 by Mann-Whitney test. (E) Cre protein detected by Western analysis in Myh6cre/wt, 728

Gdf11fl/fl, and Myh6cre/wt; Gdf11fl/fl 4 month old male mice. Band densitometry analysis

729

comparing Myh6cre/wt mice to Myh6cre/wt; Gdf11fl/fl (not shown) is not significant with p-value 0.2 730

by Mann-Whitney test. 731

Referenties

GERELATEERDE DOCUMENTEN

Het gaat met name om bladluizen, tripsen, schimmels en bacteriën die verantwoordelijk zijn voor virusoverdracht en de soorten virussen die door hen overgebracht kunnen worden

Sterk gebonden biota zijn voor instand- houding in Nederland (op dit moment) afhankelijk van groeves; matig gebonden biota komen vooral voor in groeves, maar ook in andere

Binne ’n psigososiale verstaan is dit dan ook die tydperk waar identiteit gevestig moet word (Erikson, 1968).. Aangesien hierdie prosesse verskillend ontvou vir

Dit document biedt een overzicht van de vondsten gedaan bij de werfbegeleiding van de bouw- werken aan de uitbreiding van de bibliotheek met een foyer en de bouw van een nieuw

The study is designed to find evidence for the application of availability (what comes to mind), recognition (what is similar to what), satisficing (what will do), anchoring and

Indien de BTS door Nederland gebruikt gaat worden als maat voor het aandeel grote vissen, zal niet de doelstelling van de OSPAR EcoQO kunnen worden overgenomen, maar zal een

Veeleer is door deze 'Changing views on a diptych' (H. Vlieghe) de nadruk komen te liggen op de ver- wantschap die er heerst tussen de kunst uit de Noordelijke en

The ultimate goal in the treatment of short bowel syndrome is to wean patients off par- enteral nutrition, by promoting intestinal adaptation.. Intestinal adaptation is the natural