ArticlePDF Available

Altering Sphingolipid Metabolism Attenuates Cell Death and Inflammatory Response After Myocardial Infarction

Authors:

Abstract and Figures

Background: Sphingolipids have recently emerged as a biomarker of recurrence and mortality after myocardial infarction (MI). The increased ceramide levels in mammalian heart tissues during acute MI, as demonstrated by several groups, is associated with higher cell death rates in the left ventricle and deteriorated cardiac function. Ceramidase, the only enzyme known to hydrolyze proapoptotic ceramide, generates sphingosine, which is then phosphorylated by sphingosine kinase to produce the prosurvival molecule sphingosine-1-phosphate. We hypothesized that Acid Ceramidase (AC) overexpression would counteract the negative effects of elevated ceramide and promote cell survival, thereby providing cardioprotection after MI. Methods: We performed transcriptomic, sphingolipid, and protein analyses to evaluate sphingolipid metabolism and signaling post-MI. We investigated the effect of altering ceramide metabolism through a loss (chemical inhibitors) or gain (modified mRNA [modRNA]) of AC function post hypoxia or MI. Results: We found that several genes involved in de novo ceramide synthesis were upregulated and that ceramide (C16, C20, C20:1, and C24) levels had significantly increased 24 hours after MI. AC inhibition after hypoxia or MI resulted in reduced AC activity and increased cell death. By contrast, enhancing AC activity via AC modRNA treatment increased cell survival after hypoxia or MI. AC modRNA-treated mice had significantly better heart function, longer survival, and smaller scar size than control mice 28 days post-MI. We attributed the improvement in heart function post-MI after AC modRNA delivery to decreased ceramide levels, lower cell death rates, and changes in the composition of the immune cell population in the left ventricle manifested by lowered abundance of proinflammatory detrimental neutrophils. Conclusions: Our findings suggest that transiently altering sphingolipid metabolism through AC overexpression is sufficient and necessary to induce cardioprotection post-MI, thereby highlighting the therapeutic potential of AC modRNA in ischemic heart disease.
Characterizing cell death dynamics and sphingolipid metabolism in mouse hearts after MI. A, Hearts were harvested from sham-operated mice or 1, 2, 4 and 28 days post MI. B, TUNEL assays were performed to assess DNA fragmentation in hearts harvested from either sham-operated mice or 1, 2, 4 or 28 days post MI. Troponin-I immunostaining was used to distinguish between cardiomyocytes and non-cardiomyocytes. Percentage of dead cells in left ventricle at day 1, 2, 4 and 28 post MI was quantified within all cardiac cells (DAPI + cells) n=3 (C); CMs only (DAPI + , Troponin I + ) n=3 (D) and non-CM cells only (DAPI + , Troponin I -) n=3 (E). F, For RNASeq, protein analysis and mass spectrometry, hearts were harvested from shamoperated mice or 4 or 24 hours post MI. RNA, proteins and lipids were extracted for sphingolipid determination. G, Hierarchical clustering dendrogram for the sphingolipid signaling pathway transcriptome in sham-operated hearts and hearts harvested 4 or 24 hours post MI, n=3, 3, 4, respectively. H, Sphingolipid levels measured in the LV of sham-operated mice or 24 hours post MI, n=3. I, Western blot of the AC precursor, AC active subunit β and Sphk1 in the LV of shamoperated hearts and hearts harvested 4 or 24 hours post MI. J, Quantified protein levels of AC precursor, AC β subunit and Sphk1, n=4. K, HPLC-MS/MS determination of AC activity in the LV of sham-operated hearts and hearts harvested 4 or 24 hours post MI, n=3. *, P<0.05, Oneway ANOVA, Tukey's Multiple Comparison Test for (J&K) and Holm-Sidak correction for multiple comparisons (H). Scale bar = 50μm. The results include two independent experiments for H-K.
… 
Content may be subject to copyright.
10.1161/CIRCULATIONAHA.119.041882
1
Altering Sphingolipid Metabolism Attenuates Cell Death and Inflammatory
Response after Myocardial Infarction
Running Title: Hadas et al.; Acid Ceramidase Induce Cardioprotection
Yoav Hadas, et al.
The full author list is available on page 22.
Addresses for Correspondence:
Efrat Eliyahu, PhD
Department of Genetics and Genomic Sciences
1470 Madison Ave, 8th floor, room 202A
New York, NY 10029
Tel: 917-832-2298
Email: efrat.eliyahu@mssm.edu
Lior Zangi, PhD
Department of Cardiology
1470 Madison Ave, 7th floor, room 107
New York, NY 10029
Tel: 646-254-2800
Email lior.zangi@mssm.edu
Downloaded from http://ahajournals.org by on January 30, 2020
10.1161/CIRCULATIONAHA.119.041882
2
Abstract
Background: Sphingolipids have recently emerged as a biomarker of recurrence and mortality
after myocardial infarction (MI). The increased ceramide levels in mammalian heart tissues
during acute MI, as demonstrated by several groups, is associated with higher cell death rates in
the left ventricle (LV) and deteriorated cardiac function. Ceramidase, the only enzyme known to
hydrolyze pro-apoptotic ceramide, generates sphingosine, which is then phosphorylated by
sphingosine kinase (Sphk) to produce the pro-survival molecule sphingosine-1-phosphate (S1P).
We hypothesized that Acid Ceramidase (AC) overexpression would counteract the negative
effects of elevated ceramide and promote cell survival, thereby providing cardioprotection after
MI.
Methods: We performed transcriptomic, sphingolipid and protein analyses to evaluate
sphingolipid metabolism and signaling post MI. We investigated the effect of altering ceramide
metabolism through a loss (chemical inhibitors) or gain (modified mRNA (modRNA)) of AC
function post hypoxia or MI.
Results: We found that several genes involved in de novo ceramide synthesis were upregulated
and that ceramide (C16, C20, C20:1 and C24) levels had significantly increased 24 hours after
MI. AC inhibition post hypoxia or MI resulted in reduced AC activity and increased cell death;
by contrast, enhancing AC activity via AC modRNA treatment increased cell survival post
hypoxia or MI. AC modRNA-treated mice had significantly better heart function, longer survival
and smaller scar size than control mice 28 days post MI. We attributed the improvement in heart
function post MI following AC modRNA delivery to decreased ceramide levels, lower cell death
rates and changes in the composition of the immune cell population in the LV manifested by
lowered abundance of pro-inflammatory detrimental neutrophils.
Conclusions: Our findings suggest that transiently altering sphingolipid metabolism through AC
overexpression is sufficient and necessary to induce cardioprotection post MI, thereby
highlighting the therapeutic potential of AC modRNA in ischemic heart disease.
Key Words: sphingolipid metabolism; Acid Ceramidase; Modified mRNA; myocardial
infarction; Cardioprotection.
Nonstandard Abbreviations and Acronyms
AC- Acid Ceramidase
Sphk- Sphingosine kinase
S1P -sphingosine-1-phosphate
modRNA- modified mRNA
LV- Left ventricle
MI- Myocardial infarction
LAD - Left anterior descending artery
Luc- Luciferase
nrCM- Neonatal rat cardiomyocytes
hiPS- human induce pluripotent stem cells
AAR Area at risk
LVID- Left ventricular internal diameter
EF- Ejection fraction
FS- Fractional Shortening
Downloaded from http://ahajournals.org by on January 30, 2020
10.1161/CIRCULATIONAHA.119.041882
3
Clinical Perspective
What is new?
We show that Acid Ceramidase delivery using modified mRNA alters sphingolipid
metabolism by hydrolyzing ceramides in the heart post myocardial infarction.
This metabolic change significant reduces cell death, alters immune response by limiting
neutrophil infiltration and supports cardiac function.
What are the clinical applications?
Our results suggest that Acid Ceramidase modified mRNA may serve as a cardiac
protection target post myocardial infarction.
Downloaded from http://ahajournals.org by on January 30, 2020
10.1161/CIRCULATIONAHA.119.041882
4
Introduction
Myocardial infarction is an acute, life-threatening medical condition caused by the blockage of a
coronary artery, leading to ischemia and, eventually, to cell death in the affected heart area.
Cardiac tissue has a low regenerative capacity. MI thus leads to cardiac scarring and cardiac
remodeling that increase the risk of heart failure (HF). The extent of MI and the risk of
developing HF after acute MI are positively correlated in patients.1 Several therapeutic strategies
based on inhibiting controlled cell death, attenuating inflammation and inducing regeneration
have been proposed for protecting MI survivors against HF.2, 3
Lipid levels and composition in patient blood during acute MI have been shown to
predict the risk of complications.4 In particular, high plasma ceramide concentrations are
associated with a higher probability of MI recurrence and death.4, 5 Ceramides are simple
membrane sphingolipids that form the backbone of all complex sphingolipids.6 High cellular
ceramide levels can trigger programmed cell death.7 In recent years, a few research groups have
shown that ceramide levels are high in the heart tissues of rodents and humans during acute MI8-
10 and that blocking de novo ceramide synthesis in rodents can improve heart function post MI.8, 9
Ceramidases hydrolyze ceramide to generate free fatty acids and sphingosine, which is
then phosphorylated by sphingosine kinase (Sphk) to produce sphingosine 1-phosphate (S1P), a
pro-survival lipid mediator with both intra- and extracellular functions.11, 12 More specifically,
acid ceramidase (AC) is encoded by the Asah1 gene and catalyzes ceramide hydrolysis to free
fatty acids and sphingosine, which is then phosphorylated by Sphk (1 and 2) to generate S1P.13
Asah1 gene mutations lead to ceramidase deficiency and cause Farber lipogranulomatosis, a
lysosomal storage disease.13 AC is essential for embryogenesis, and Asah1-/- embryos undergo
apoptotic death at the two-cell stage.12 AC is a lysosomal and secreted enzyme, with optimal in
Downloaded from http://ahajournals.org by on January 30, 2020
10.1161/CIRCULATIONAHA.119.041882
5
vitro activity in acidic conditions,14 and belongs to the N-terminal nucleophile hydrolase family.
The autoproteolytic cleavage of AC generates two active subunits: the α subunit with a
molecular weight of ~14 kDa and the β subunit with a molecular weight of ~43 kDa.15 The
autoproteolytic cleavage of the precursor triggers a conformational change that reveals the active
site and activates the enzyme toward sphingolipid metabolism.16 It has been suggested that
interfering with the signal transduction pathways mediated by sphingolipids could prevent cell
death post MI. Recent studies have suggested that S1P could be used as a therapeutic target in
patients with heart failure17 and MI,18 to prolong cardiac cell survival and consequently improve
heart function. While S1P lyase inhibition causes increased cardiac S1P levels and bradycardia in
rats,19 S1P receptor agonist, FTY720, boosts myocardial salvage and enhances heart function in a
porcine model of ischemia/reperfusion injury.20 Inhibiting de novo ceramide synthesis has also
been suggested as a strategy for reducing the pro-apoptotic effect of ceramide post MI.9 Indeed,
inhibiting acid sphingomyelinase, which hydrolyzes spingomyelin to generate ceramide, limits
ceramide accumulation in post-ischemic hearts.10 Moreover, adiponectin seems to exert its anti-
apoptotic effect on CMs through adiponectin receptor-mediated ceramidase activity.21
We investigated using AC and/or Sphk enzymes to inhibit cell death and initiate cell
survival through ceramide hydrolysis and S1P production. Delivering AC or Sphk proteins is
safe and controlled, but their effects are limited by these proteins’ half-lives. Conversely, using
DNA or viruses (DNA or RNA viruses) is not controlled and may elicit an immune response that
could compromise genome integrity. We therefore delivered AC and Sphk via synthetic
modified mRNA (modRNA), a nucleic acid delivery tool, to transiently alter sphingolipid
metabolism. Our group and others have shown that modRNA is a highly efficient system for
delivery to the heart, rapidly yielding transient expression with no signs of innate
Downloaded from http://ahajournals.org by on January 30, 2020
10.1161/CIRCULATIONAHA.119.041882
6
immunoresponse.22-28 More specifically, we have shown that synthesized modRNAs, in which all
uridine residues are replaced with pseudouridine-5'-triphosphate or N1-methylpseudouridine-5'-
triphosphate (1-mψU), result in more efficient translation, with lower immunogenicity and
greater resistance to RNase cleavage than unmodified mRNA in cardiac cells and tissue.27
ModRNA, which has a unique, transient, pulse-like pharmacokinetic profile, is translated within
minutes and the resulting protein remains detectable for ~5-7 days in vitro and ~10 days in
vivo.27 This distinctive kinetic profile makes modRNA approaches ideal for immediately and
transiently altering sphingolipid metabolism in the heart post MI. While insulin-like growth
factor 1 (IGF-1) modRNA delivered immediately after MI has been shown to decrease CM
apoptosis, this treatment’s long-term effects on heart function are remain unclear.22 We recently
showed that delivering IGF-1 modRNA to the heart post MI induces detrimental epicardial
adipose tissue formation by differentiate epicardial derived cells into fat cells 28 days post MI.25
In this study, we investigated sphingolipid level dynamics and the expression of genes
involved in sphingolipid metabolism and signaling pathways post MI. Loss (chemical inhibitors)
or gain (modRNA) of AC function showed that inhibiting AC activity increased cell death and,
by contrast, enhancing AC activity reduced cell death under hypoxia conditions in vitro or during
MI in vivo. Moreover, we show that enhancing AC activity attenuated inflammation post MI by
decreasing detrimental neutrophil levels in the LV, thereby improving heart function and long-
term survival after ischemic injury.
Methods
All methods and materials used in this study are described in detail in the Supplementary
Materials and Methods section. Below please see a brief summary of the most relevant ones.
Downloaded from http://ahajournals.org by on January 30, 2020
10.1161/CIRCULATIONAHA.119.041882
7
Mice
All animal procedures were performed according to protocols approved by the Icahn School of
Medicine at Mount Sinai Institutional Care and Use Committee. CFW male and female mice
were used. For protein expression, mice were injected intramyocardially with 100 g of Luc,
Sphk1 or modRNA in citrate buffer. MI was induced by permanent LAD ligation. When
required, 100 g of modRNA was injected into the border zone immediately after LAD ligation.
For the inhibitor assay, 10mg/kg of ARN14974 was injected IP at the time and 7 hours post LAD
ligation. Heart function was determined using echocardiography and MRI.
ModRNA Synthesis
Clean PCR products generated with plasmid templates (GeneArt, Thermo Fisher Scientific) were
used as the template for mRNA (for a complete list of the open reading frames used in this study,
see Supplementary Table 1). ModRNAs were produced by transcription in vitro with a
customized ribonucleoside blend and DNA PCR products with plasmid templates, followed by
Antarctic Phosphatase treatment and purification. Please see supplemental materials methods
section for more detailes.
nrCM isolation and hPSC differentiation
Ventrical nrCMs were isolated from 3- to 4-day-old Sprague-Dawley rats by multiple digestion
series using 0.1% collagenase II. We obtained hPSCs-derived CMs by using the embryoid bodies
(EBs) formatting method.
In vitro modRNA transfection
Neonatal rat hPSC-derived CMs were transfected with 2.5 g of modRNA per well (on 24 well
plate) encoding nGFP, AC, Sphk1, SphK2 or S1pr2 using RNAiMAX (Life Technologies).
Downloaded from http://ahajournals.org by on January 30, 2020
10.1161/CIRCULATIONAHA.119.041882
8
Sphingolipid composition and AC activity
To determine the sphingolipid composition of mouse hearts, samples were analyzed at the Stony
Brook Lipidomics facility. AC activity in heart tissue lysate was analyzed by HPLC-MS/MS.
Immunofluorescence
Hearts were perfused and fixed with 4% PFA, followed by incubation with 30% sucrose and
mounting in OCT. Transversal 10 m-thick sections were stained overnight with primary
antibodies followed by appropriate secondary antibodies, as listed in Supplementary Table 2,
depending on the experiment. To detect apoptotic cells in the heart tissue, TUNEL assay was
performed according to manufacturer’s protocol. To stain cells, coverslips with previously
seeded cells were fixed with 4%PFA and stained with appropriate antibodies. Images were
quantified using ImageJ software.
Flow cytometry
Infarct zones of AC modRNA- or Luc modRNA-treated hearts were collected at 2, 7 and 14 days
post MI and processed according to a previously described protocol29 with a few modifications.
Isolated cells were first stained with hematopoietic lineage markers followed by a second
staining with specific cell markers. Neutrophils were defined as CD45+ CD11b+ Ly6G+,
Macrophages as CD45+ CD11b+ Lin- F4/80+ Ly6Clow/int and Monocytes as CD45+ CD11b+ Lin-
F4/80- Ly6Clow / Ly6Chi. Data were acquired using Aurora (Cytek) Spectral Flow Cytometer.
Statistical analysis
All statistical analysis was performed with GraphPad-Prism software. Values are reported as
means ± SD. Two-tailed Student's t-tests (*p < 0.05 considered significant) or one-way ANOVA
with Bonferroni correction (*p < 0.05 considered significant) were used for comparisons among
groups. Log-rank (Mantel-Cox) tests were used to analyze survival.
Downloaded from http://ahajournals.org by on January 30, 2020
10.1161/CIRCULATIONAHA.119.041882
9
Results
Cell death and sphingolipid signaling pathway expression dynamics post MI
We characterized the dynamics of cell death and gene expression during and after acute MI by
ligating the left anterior descending artery (LAD) of mouse hearts to cause infarction and then
harvesting hearts at various time points (Fig. 1A).
To assess cell death, we harvested hearts from both sham-operated and ligated mice 1, 2,
4 and 28 days post MI. We used TUNEL staining to assess DNA fragmentation in cardiac cells
and troponin-I (cardiomyocytes marker) immunostaining to distinguish between cardiomyocytes
(CMs) and non-CMs (Fig. 1B). DNA fragmentation levels were highest 24 hours post MI, with
fragmented DNA detected in 9±2% of total cells in the LV (Fig. 1C), 15±3% of CMs (Fig. 1D)
and 4±0.2% of non-CM cells (Fig. 1E). DNA fragmentation levels were lower 48 hours after MI,
in both CMs and non-CM cells, and fell to basal levels by 28 days post MI (Fig. 1C-E).
An unbiased comparison of the LV transcriptome (Fig. 1F) 24 hours post MI identified a
significant change (P=0.02) in the sphingolipid signaling pathway KEGG PATHWAY
map04071.30 This pathway includes 112 genes, 38 of which had significantly changed expression
after MI (Fig. 1G, Supplementary Table 3 and Supplementary Fig. 1F). In addition, 24 hours post
MI, 12 of the 39 (30%) sphingolipid metabolism genes were significantly upregulated
(Supplementary Fig. 1E, F). None of the genes involved in the sphingomyelin hydrolysis
pathway were upregulated at 4 or 24 hours post MI, but four genes involved in the de novo
ceramide synthesis pathway (Sptlc2, CerS2, 3 and 6) displayed 2.2-, 2.3-, 5.2- and 2.2-fold
upregulation, respectively (Supplementary Table 3).
We next determined ceramide levels in the myocardium 24 hours post MI. The levels of
C16-ceramide (8.75-fold increase), C20-ceramide (16.57-fold increase), C20:1-ceramide (8.38-
Downloaded from http://ahajournals.org by on January 30, 2020
10.1161/CIRCULATIONAHA.119.041882
10
fold increase) and C24-ceramide (8.95 increase) were significantly higher 24 hours post MI than
in the absence of MI (Fig. 1H).
In this study, we focused on sphingolipid metabolism and the role of ceramide
degradation by AC and sphingosine phosphorylation to S1P by sphingosine kinase 1 (Sphk1).
We therefore used a separate mouse cohort to measure the mRNA and protein levels
corresponding to these key ceramide metabolism genes. Consistent with our transcriptome
analysis, relative levels of AC mRNA (Supplementary Fig. 1G and Supplementary Table 3) and
AC precursor levels were unaffected after MI (Fig. 1I&J). However, the active AC-enzyme, as
shown by β subunit, was significantly increased along with activity (Fig. 1K). AC-enzyme α
subunit was also upregulated 24 hours post MI (Supplementary Fig. 1I). Moreover, the Sphk1,
mRNA (Supplementary Fig. 1H) and protein (Fig. 1I&J) levels were notably upregulated 24
hours post MI. We also determined relative mRNA levels for AC and Sphk1 as well as Sphk1
protein levels in nrCMs under normoxia or hypoxia (Supplementary Fig. 1A-D). Though we
observed no significant effect on AC and Sphk1 mRNA levels (Supplementary Fig. 1A-B),
Sphk1 protein levels were moderately but significantly higher in cells subjected to hypoxia
(Supplementary Fig. 1C-D).
Modulating sphingolipid metabolism in the heart during acute MI affects cell death
Ceramide metabolites’ effect on cardiomyocyte (CM) death and heart function were investigated
post MI by altering ceramide metabolism via inhibiting or enhancing ceramide degradation and
S1P generation. We used two previously described approaches to limit ceramide degradation in
neonatal rat cardiomyocytes (nrCM): the pan-ceramidase inhibitor B1331 and the acid
ceramidase-specific inhibitor ARN1497432. We also blocked S1P formation with a pan-
sphingosine kinase inhibitor, SK1-II.33 Then, we assessed these inhibitors’ effects under
Downloaded from http://ahajournals.org by on January 30, 2020
10.1161/CIRCULATIONAHA.119.041882
11
normoxia (21% oxygen) and hypoxia (<2% oxygen) (Fig. 2A). In normoxia, inhibiting
ceramidase activity with 50 µM B13 or 20 µM ARN14974 and inhibiting sphingosine kinase
activity with 30 µM SK1-II for 48 hours had no significant effect on nrCM death rates, as shown
by comparison with control cells treated with the solvent used for the inhibitors, DMSO (Fig.
2B). However, increasing B13 concentration to 100 µM or SK1-II concentration to 60 µM
induced nrCM cell death 48 hours after treatment (Fig. 2B). Next, we assessed the combined
inhibition of ceramidase and sphingosine kinase activities. When used together at low doses (50
µM for B13 and 30µM for SK1-II), these two inhibitors induced significantly higher levels of
nrCM cell death than any other treatment except 100 µM B13, for which the same trend was
observed but to a lesser extent (Fig. 2B). In hypoxic conditions, all inhibitor types, at all
concentrations tested, significantly increased CM cell death levels 24 hours after treatment (Fig.
2C). By contrast, 48 hours after treatment, most cells were apoptotic regardless of the presence
or absence of inhibitors, which no longer significantly affected CM death rates (Fig. 2D).
We assessed loss of function in vivo using an AC-specific inhibitor (ARN14974) that has
been shown to reduce enzyme activity in mouse hearts for more than seven hours after
intraperitoneal injection (Fig. 2E).31 First, we first confirmed that the AC-specific inhibitor could
affect AC in our assay (Fig. 2F). We then administered two IP injections (immediately and seven
hours post MI) of the AC-specific inhibitor; this treatment resulted in twice as many cells
containing fragmented DNA 24 hours post MI (Fig. 2G&H). Importantly, lower doses of AC-
specific inhibitor lead to insignificant changes in cell death 24 hours post MI (Supplementary
Fig. 2A-B). To evaluative AC’s role in mouse survival post MI, we compared mouse survival
following treatment with either AC-specific inhibitor or DMSO control post MI. We show
Downloaded from http://ahajournals.org by on January 30, 2020
10.1161/CIRCULATIONAHA.119.041882
12
significantly reduced survival 72 hours post MI and treatment with AC-specific inhibitor (Fig
2I).
Additionally, to investigate the effect of overexpressing AC or Sphk1, alone or together,
in nrCMs under hypoxic conditions (Fig. 3A), we used modRNAs encoding human AC and
Sphk1. First, we confirmed the modRNAs were translated in nrCMs (Fig. 3B). We then cultured
the transfected nrCMs in a hypoxia chamber to assess the modRNAs’ ability to prevent nrCM
cell death. After 48 hours in hypoxia, 60% of the nrCMs transfected with nGFP (the control)
were apoptotic, whereas cell death rates were 22% and 27% lower, respectively, in cells
transfected with the AC modRNA and the Sphk1 modRNA. One of modRNA’s advantages as an
upregulation tool is that several modRNAs can be transfected together into the same cell.25 The
overexpression of both AC and Sphk1 decreased the number of apoptotic cells by 48% (Fig. 3C).
Similarly, we investigated the effects of AC and Sphk1 overexpression on cell death in the LV
post MI (Fig. 3D). We first used immunofluorescence staining to confirm that the various
modRNAs were translated in the LV 24 hours post MI (Fig. 3E). We then confirmed that AC
activity was significantly higher following AC modRNA delivery (Fig. 3F). Finally, we assessed
DNA fragmentation by performing TUNEL assays 48 hours post MI and after the various
treatments (Fig. 3G&H). AC overexpression in the left ventricle immediately after LAD ligation
produced 54% fewer cells with fragmented DNA than hearts treated with Luc modRNA, 48
hours post MI (Fig. 3G&H). Neither Sphk1 overexpression nor lower doses of AC modRNA
significantly changed DNA fragmentation (Fig. 3H and Supplementary Fig. 3A&B), while the
combined approach, upregulating both AC and Sphk1, had an effect similar to that of AC alone
(59% decrease) (Fig. 3G&H). In addition, we evaluated AC modRNA’s effect in human CMs by
transfecting human-induced pluripotent stem cell-derived CMs (hiPS-CMs) with AC modRNA
Downloaded from http://ahajournals.org by on January 30, 2020
10.1161/CIRCULATIONAHA.119.041882
13
(Supplementary Fig. 4A). The AC modRNA was translated to generate AC protein
(Supplementary Fig. 4B), resulting in lower cell death levels under hypoxia (Supplementary Fig.
4C).
Transient AC overexpression improves heart function and survival rate in mice post MI
Based on the beneficial effects of AC both alone and with Sphk1, we investigated how these
modRNAs affect cardiac function post MI by injecting AC, Sphk1, AC+Sphk1 or Luc
modRNAs directly into the LV and evaluating cardiac function post MI (Fig. 4A). Two days post
MI, no significant difference in either area at risk from the left ventricle (AAR/LV) or %FS
(fractional shortening, expressed as a percentage) was observed between groups (Supplementary
Fig. 5A&B). However, 28 days post MI, the %FS of the LV in mice treated with AC, Sphk1 or
AC+ Sphk1 modRNAs was significantly higher than in the control (Supplementary Fig. 5C).
Left ventricular internal diameter end diastole (LVIDd) did not differ significantly between
treated and control mice 28 days post MI (Fig. 4B). Yet left ventricular internal diameter end
systole (LVIDs) was significantly lower in mice treated with AC or AC+Sphk1 modRNAs than
in control mice (Fig. 4C). Comparing %FS values between 2 and 28 days post MI revealed
significantly improved heart function in mice treated with the Sphk1 modRNA and highly
improved heart function in mice treated with AC or AC + Sphk1 modRNAs (Fig. 4D). To
evaluate scar size after MI and the various treatments, we performed Masson trichrome staining
(Fig. 4E) and found that scars were significantly smaller after treatment with AC, Sphk1 or
AC+Sphk1 modRNAs (Fig. 4F). Further, we found no sign of CM hypertrophy in the LV of
treated mice relative to the control (Supplementary Fig. 5D&E). AC modRNA alone yielded the
most significant beneficial effect on the LV post MI. We therefore used magnetic resonance
imaging (MRI) to compare the percent ejection fraction (%EF) of mice treated with AC
Downloaded from http://ahajournals.org by on January 30, 2020
10.1161/CIRCULATIONAHA.119.041882
14
modRNA with that of mice treated with the Luc control modRNA (Fig. 4G). We observed
significantly higher EF (Fig. 4H), with better-preserved wall thickness, specifically in the areas
into which the AC modRNA was injected (posterior and lateral) relative to the injection-free
areas (anterior and septal) (Fig. 4I & Supplementary Movies 1&2). Finally, we determined that
survival rates three months post MI were significantly higher for mice treated with the AC
modRNA than for mice receiving other treatments or for control mice (Fig. 4J).
Transient AC overexpression reduces ceramide levels and attenuates inflammation post MI
To investigate the molecular basis of improved heart function, we collected hearts from mice
treated with AC or Luc control modRNAs and analyzed the effect of AC overexpression on
protein levels and sphingolipid composition 24 hours post MI; we also analyzed whole-
transcriptome dynamics 4, 24 and 48 hours post MI (Fig. 5A). Strikingly, AC overexpression
decreased the levels of all ceramides investigated significantly for C20 and C22 in the LV
post MI (Fig. 5B). We investigated this effect’s possible correlation with apoptosis markers, such
as caspase 3 dimerization,34 and cleavage. We found that caspase 3 dimerization levels were
higher in control mice than in sham-operated mice 24 hours post MI and that AC-treated mice
had lower caspase 3 dimer levels than did control mice (Fig. 5C and Supplementary Fig. 6A).
Unbiased transcriptome analysis revealed that only 30 genes displayed at least 1.5-fold
expression changes (p<0.05) in either direction (Supplementary Fig. 6B and Supplementary
Table 4) four hours after MI. By contrast, the number of genes displaying at least 1.5-fold
expression (p<0.05) in either direction rose to 299 by 24 hours post MI (Supplementary Fig. 6B
and Supplementary Table 5) and to 1675 by 48 hours post MI (Supplementary Fig. 6B and
supplementary Table 6). GO enrichment analysis revealed that AC overexpression altered many
immune-related pathways 24 hours post MI (Supplementary Table 7). Two days post MI, most
Downloaded from http://ahajournals.org by on January 30, 2020
10.1161/CIRCULATIONAHA.119.041882
15
of the genes displaying expression changes were involved in the cell cycle, cell apoptosis and
immune response (Supplementary Fig. 6C). Out of all the genes that were upregulated two days
post MI in mice that received ligation rather than sham, 144 genes had lower expression levels in
AC modRNA-treated hearts than in Luc control modRNA-treated hearts. GO enrichment
analysis with Enrichr35, 36 revealed that among the 144 genes with lower expression levels are
genes involved in lipid metabolism-related genes and cell death. Interestingly, neutrophil
chemotaxis and inflammatory response also show lower expression in the AC modRNA-treated
hearts (Fig. 5D and Supplementary Fig. 6D). The partial transcriptome for sphingolipid
metabolism-related genes was unchanged 4 and 24 hours post MI in AC modRNA-treated hearts
(data not shown). However, 48 hours post MI, we observed significant expression changes for
many of the sphingolipid metabolism-related genes included in RNA sequencing analysis
(Supplementary Fig. 6D). To verify RNAseq data, we selected the Ngp gene encoding the
neutrophilic granule protein, this gene’s expression was downregulated at 4, 24 and 48 hours
post MI in AC modRNA-treated hearts relative to Luc modRNA-treated hearts. We confirmed
changes in this gene’s expression patterns by performing RT-qPCR (please see primers in
Supplemental Table 8) on RNA isolated from the hearts two days post MI; the results obtained
were consistent with our transcriptomic results (Fig. 5E).
The significantly decreased expression of inflammation-related genes in AC modRNA-
treated hearts led us to investigate the composition of the immune cell populations in the infarct
zone of the heart at days 2, 7 and 14 post MI (Fig. 5F). At day 2 post MI, neutrophil infiltration
was reduced significantly in the infarct zone of AC modRNA-treated hearts compared to those
from controls (Fig. 5H-I). No significant differences were found in Ly6Chi and Ly6Clow
monocyte populations (Fig. 5J-K), nor macrophages (Fig. 5L-N). Seven days post MI,
Downloaded from http://ahajournals.org by on January 30, 2020
10.1161/CIRCULATIONAHA.119.041882
16
macrophage numbers increased dramatically whereas neutrophils decreased; both cells remained
near these levels by day 14 post MI. (Supplementary Fig. 6E-F).
Discussion
CM cell death and cardiac inflammation are key to the severity of cardiac dysfunction post MI.
Decreasing CM cell death and inflammation in patients with acute MI or other cardiovascular
diseases is highly desirable.2, 37 There is growing evidence that blood ceramide levels can predict
the extent of cardiovascular disease, complications and mortality.4, 5, 38, 39 Total ceramide levels,
particularly of ceramides with fatty acid chains of 16 or more carbon atoms, are high in the
myocardium in rodent models of MI8, 10 and in human patients with HF.8, 10
We characterized the significant changes to programmed cell death, the expression of
genes involved in sphingolipid metabolism or signaling, sphingolipid levels and AC activity, 24
hours post MI (Fig. 1). We observed significant increases in the levels of C16, C20, C20:1 and
C24 ceramides (Fig. 1H). Further, we found that the heart accommodated the increased ceramide
levels through two different molecular mechanisms. The first involved sharply upregulated
Sphk1 mRNA and protein levels (Fig. 1I&J and Supplementary Fig. 1H), and the second
involved moderately upregulated AC enzymatic activity despite the absence of change in AC
mRNA or protein levels (Fig. 1I-K and Supplementary Fig. 1G&I). These findings suggest that
sphingolipid metabolism, especially Sphk1 and AC enzymatic activity, are important factors in
cardiac tissue response to ischemic conditions.
Moderate concentrations of either ceramidase or sphingosine kinase inhibitors were not
toxic to nrCMs under normal culture conditions. However, incubating nrCM cells with a higher
concentration of either inhibitor separately, or lower concentrations of the two inhibitors
Downloaded from http://ahajournals.org by on January 30, 2020
10.1161/CIRCULATIONAHA.119.041882
17
together, induced cell death. Curtailing these enzymes may have caused ceramide and/or
sphingosine to rise to toxic levels. The significant increase in nrCM cell death observed in
conditions of moderate ceramidase or sphingosine kinase inhibition in hypoxic conditions
suggests that a combination of hypoxia and enzyme inhibition can increase the rate of pro-
apoptotic sphingolipid accumulation and cell death (Fig. 2C). Furthermore, in our in vivo loss-of-
function study, limiting AC activity in mouse hearts led to elevated cardiac cell death 24 hours
post MI. These results suggest that AC activity is a crucial cardioprotective factor in the ischemic
heart (Fig. 2E-H). That inhibiting AC post MI significantly decreases survival indicates AC’s
active role is vital to mouse survival post MI (Fig. 2I).
Ceramide has been shown to induce cell death in various cell types,7 including murine
and human CMs.40, 41 Conversely, the phosphorylation of sphingosine, a ceramide degradation
product, generates a major cell survival and cardioprotective agent, S1P.20,42 Klevsting et al.
suggested that Ceramide accumulation in the myocardium may be partially reliant due to
sphingomyelinase activity, and a heterozygous knockout of Smpd1, which encodes acid
sphingomyelinase, reduced ceramide accumulation in mouse hearts, albeit insufficiently to
improve either heart function or survival 30 days post MI.10 Recent studies in mouse models
have demonstrated constraining de novo ceramide synthesis improves heart function post MI8
and moderately decreases the infarct area and inflammation levels following myocardial I/R
injury.9 Our transcriptomic analysis revealed upregulated genes involved in de novo ceramide
synthesis, including the Sptlc2 and CerS2 3 and 6 genes, with no change in mRNA levels for the
natural sphingomyelinase (Smpd3) and lower transcript levels for other sphingomyelinases
(including Smpd1) 24 hours post MI (Supplementary Table 3). We here use AC for ceramide
hydrolysis43 to prevent ceramide toxicity and promote survival.
Downloaded from http://ahajournals.org by on January 30, 2020
10.1161/CIRCULATIONAHA.119.041882
18
Consistent with its anti-apoptotic effects, sphingosine may disassemble mitochondrial
ceramide channels,44, 45 but it has also been implicated in apoptotic or necrotic cell death.46
Benaim et al. suggested that sphingosine might disturb cellular calcium homeostasis by blocking
the activity of the sarcoendoplasmic reticulum Ca2+-ATPase (SERC2a),47 which plays a crucial
role in correct cardiac function.48, 49 Sphingosine kinase catalyzes the phosphorylation of
sphingosine to generate S1P and has cardioprotective properties.50 Accordingly, adenovirus-
mediated Sphk1 overexpression in rat hearts has been shown to protect treated hearts from I/R
injury.51 In this study, we decided to decrease ceramide levels in the ischemic heart by increasing
ceramide hydrolysis via modRNA used as a gene delivery tool to induce transient overexpression
of AC. This strategy not only decreased ceramide levels but also expanded the sphingosine
reservoir, the principal source of raw materials for producing the pro-survival molecule S1P. We
found that AC modRNA increased AC expression in vitro and in vivo (Fig. 3B, E). The resulting
high levels of AC protein were associated with more AC activity, leading to less cardiac cell
death post MI (Fig. 3C and Fig. 3F-H). We examined AC and/or Sphk1 overexpression’s in vitro
and in vivo effects on reduced cell death. Sphk1 overexpression generated via modRNA was
found to prevent ischemic induced cell death in vitro but not in vivo. (Fig. 3C and Fig. 3H). This
may be because Sphk1 mRNA and protein levels are naturally higher post MI in vivo than in in
vitro hypoxia settings. (Supplementary Fig. 1B-D, H).
Interestingly, S1P receptor 2 (S1pr2) or Sphk2 overexpression did not significantly
reduce cardiac cell death either in vitro or in vivo (Supplementary Fig. 7&8). Sphk1 is, thus,
effective for preventing apoptotic death but not redundant with Sphk2 for this function.
Consistent with this conclusion, Xia et al. showed that Sphk1 interacted with TNF receptor-
associated factor 2 (TRAF2) and that this interaction was required for TRAF2’s anti-apoptotic
Downloaded from http://ahajournals.org by on January 30, 2020
10.1161/CIRCULATIONAHA.119.041882
19
activity.52 Our pathway analysis for sphingolipid signal transduction revealed upregulated
TRAF2 post MI (Supplementary Table 3). Guo et al. recently reported a cardioprotective role for
TRAF253. Future studies are required to investigate the interaction between Sphk1 and TRAF2 in
the context of MI. Cell death levels did not change following S1pr2 upregulation, possibly due to
this receptor’s high physiological expression levels in the heart.
We observed a clear therapeutic effect 28 days after AC modRNA delivery in mice with
MI (Fig. 4), including preserved posterior and lateral wall thicknesses, which were most marked
in the area into which the AC modRNA was injected. AC modRNA had no significant effect on
%LVFS two days post MI (Supplementary Fig. 5A), whereas its effect was significant 28 days
post MI (Supplementary Fig. 5B). This suggests that AC modRNA may not only prevent cell
death immediately post MI but also influence other cardiac functions in this context. We also
show a non-significant reduction of LVIDd 30 days post-MI (Fig. 4b) and reduce scar size (Fig.
4e) that implies but not prove a reduced remodeling. Further investigation on the long-term
effects of AC treatment will clarify the effect on heart remodeling. In further sphingolipid
analyses, we found that AC modRNA affected cell death by decreasing ceramide levels post MI,
particularly for ceramides 20 and 22, (Fig. 5B), and by strongly curtailing caspase 3 dimer
formation and cleavage post MI (Fig. 5C and Supplementary Fig. 6A). In addition, many genes
involved in sphingolipid metabolism, inflammation and neutrophil degranulation displayed
altered expression following AC modRNA treatment, two days post MI (Fig. 5D and
Supplementary Fig. 6C&D). High ceramide levels can lead to neutrophil recruitment54 and
activation.55 Analyzing the immune cell population in the infarct area revealed significantly
lower neutrophil infiltration into the infarct zone of AC modRNA-treated hearts two days post
MI (Fig. 5I), consistent with the post MI altered immune response shown by GO enrichment
Downloaded from http://ahajournals.org by on January 30, 2020
10.1161/CIRCULATIONAHA.119.041882
20
analysis (Fig. 5D) and lowered expression of the Ngp gene, which is expressed predominantly in
neutrophils and promyelocytes56 (Fig. 5E) in these hearts.
Inflammation and immune cell infiltration into infarct cardiac tissue are crucial to healing
after MI.57 Completely inhibiting the recruitment of a particular immune cell population, such as
neutrophils or macrophages, is detrimental to the heart post MI,58, 59 but reducing the numbers of
neutrophils infiltrating the myocardium has been shown to shrink infarct size and promote
adaptation to hypoxic stress.60, 61 Administering AC modRNA immediately after MI decreased
detrimental neutrophil levels, thereby providing an additional mechanism for improving outcome
post MI. It remains unclear how AC modRNA alters the immune cell population in the LV post
MI; AC modRNA may influence chemokine expression directly or indirectly.
Additionally, we do not yet know which S1P receptor (1-5) is activated in the heart
during cell death rate reduction. The two most abundant receptors in the heart, S1pr1 and 3,
increase significantly post MI (Supplementary Table 3). By contrast, S1pr2 levels are low four
hours after MI and return to normal by 24 h post MI. The roles of S1pr1 and S1pr3 in
cardioprotection are well established,50 but the role of S1p2 in heart function remains unclear.
In addition, AC modRNA’s effects post MI have yet to be investigated in large animals.
However, by using hiPS-CMs in a hypoxia chamber, we demonstrated that AC modRNA
reduced cell death to control levels in human settings, an effect similar to that reported here for
nrCMs (Supplementary Fig. 6).
Overall, our data suggest (as summarized in Fig. 5O) that using AC modRNA to achieve
transient overexpression of AC, an essential enzyme in sphingolipid metabolism and cell
survival,12 lowers ceramide levels, decreases cardiac cell death and attenuates but does not
Downloaded from http://ahajournals.org by on January 30, 2020
10.1161/CIRCULATIONAHA.119.041882
21
eliminate detrimental neutrophil infiltration in the LV. Further, these effects improve cardiac
function, leading to improved survival after MI in mice.
Acknowledgments
The authors acknowledge Jason Kondrat, Sunita DSouza and the Pluripotent Stem Cell Core
Facility at the Icahn School of Medicine at Mount Sinai for their help with this manuscript.
Sources of Funding
This work was funded by a cardiology start-up grant and NIH grant R01 HL142768-01 awarded
to the Zangi laboratory and by the Genetics and Genomic Sciences budget in support of the
Eliyahu laboratory.
Disclosures
E.E., L.Z, A.S.V. and Y.H. are Inventors on Provisional Patent Application (MODRNA
ENCODING SPHINGOLIPID METABOLIZING PROTEINS TO PROMOTE CELL
SURVIVAL) 3710/039P, Filed March 2018, which covers the results in this manuscript.
Downloaded from http://ahajournals.org by on January 30, 2020
10.1161/CIRCULATIONAHA.119.041882
22
Authors
Yoav Hadas, PhD1,2,4; Adam S. Vincek, PhD2; Elias Youssef, MSc1,2,4;
Magdalena M. Żak, PhD1,2,4; Elena Chepurko, MDM1,2,4; Nishat Sultana, PhD1,2,4;
Mohammad Tofael Kabir Sharkar, PhD1,2,4; Ningning Guo, PhD2; Rinat Komargodski, MSc1,2,4;
Ann Anu Kurian, MSc1,2,4; Keerat Kaur, PhD1,2,4; Ajit Magadum, PhD1,2,4;
Anthony Fargnoli, PhD1; Michael G. Katz, MD1; Nadia Hossain, MSc1,2,4;
Ephraim Kenigsberg, PhD2; Nicole C. Dubois, PhD3,4; Eric Schadt, PhD2,5; Roger Hajjar, MD6;
Efrat Eliyahu, PhD2,5; Lior Zangi, PhD1,2,4
Co-corresponding authors
Affiliations
1Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY;
2Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New
York, NY; 3Department of Developmental and Regenerative Biology and The Mindich Child
Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY;
4Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY;
5Multiscale Biology Institute, Icahn School of Medicine at Mount Sinai, New York, NY;
6Phospholamban Foundation, Amsterdam, Netherlands
Supplemental Materials
Supplementary Materials and Methods
Supplementary Tables 1-8
Supplementary Figures 1-8
Supplementary Movies 1 and 2
References 62-63
Downloaded from http://ahajournals.org by on January 30, 2020
10.1161/CIRCULATIONAHA.119.041882
23
References
1. Spencer FA, Meyer TE, Gore JM and Goldberg RJ. Heterogeneity in the management
and outcomes of patients with acute myocardial infarction complicated by heart failure: the
National Registry of Myocardial Infarction. Circulation. 2002;105:2605-2610.
2. Hadas Y, Katz MG, Bridges CR and Zangi L. Modified mRNA as a therapeutic tool to
induce cardiac regeneration in ischemic heart disease. Wiley Interdiscip Rev Syst Biol Med.
2017;9: wsbm.1367.
3. Lin Z and Pu WT. Strategies for cardiac regeneration and repair. Sci Transl Med.
2014;6:239rv1.
4. Meeusen JW, Donato LJ and Jaffe AS. Lipid Biomarkers for Risk Assessment in Acute
Coronary Syndromes. Curr Cardiol Rep. 2017;19:48.
5. Yu J, Pan W, Shi R, Yang T, Li Y, Yu G, Bai Y, Schuchman EH, He X and Zhang G.
Ceramide is upregulated and associated with mortality in patients with chronic heart failure. Can
J Cardiol. 2015;31:357-363.
6. Castro BM, Prieto M and Silva LC. Ceramide: a simple sphingolipid with unique
biophysical properties. Prog Lipid Res. 2014;54:53-67.
7. Arana L, Gangoiti P, Ouro A, Trueba M and Gomez-Munoz A. Ceramide and ceramide
1-phosphate in health and disease. Lipids Health Dis. 2010;9:15.
8. Ji R, Akashi H, Drosatos K, Liao X, Jiang H, Kennel PJ, Brunjes DL, Castillero E, Zhang
X, Deng LY, Homma S, George IJ, Takayama H, Naka Y, Goldberg IJ and Schulze PC.
Increased de novo ceramide synthesis and accumulation in failing myocardium. JCI Insight.
2017;2:225-230.
9. Reforgiato MR, Milano G, Fabrias G, Casas J, Gasco P, Paroni R, Samaja M, Ghidoni R,
Caretti A and Signorelli P. Inhibition of ceramide de novo synthesis as a postischemic strategy to
reduce myocardial reperfusion injury. Basic Res Cardiol. 2016;111:12.
10. Klevstig M, Stahlman M, Lundqvist A, Scharin Tang M, Fogelstrand P, Adiels M,
Andersson L, Kolesnick R, Jeppsson A, Boren J and Levin MC. Targeting acid
sphingomyelinase reduces cardiac ceramide accumulation in the post-ischemic heart. J Mol Cell
Cardiol. 2016;93:69-72.
11. Eliyahu E, Shtraizent N, Martinuzzi K, Barritt J, He X, Wei H, Chaubal S, Copperman
AB and Schuchman EH. Acid ceramidase improves the quality of oocytes and embryos and the
outcome of in vitro fertilization. FASEB J. 2010;24:1229-38.
12. Eliyahu E, Park JH, Shtraizent N, He X and Schuchman EH. Acid ceramidase is a novel
factor required for early embryo survival. FASEB J. 2007;21:1403-1409.
13. Koch J, Gartner S, Li CM, Quintern LE, Bernardo K, Levran O, Schnabel D, Desnick RJ,
Schuchman EH and Sandhoff K. Molecular cloning and characterization of a full-length
complementary DNA encoding human acid ceramidase. Identification Of the first molecular
lesion causing Farber disease. J Biol Chem. 1996;271:33110-33115.
14. Bernardo K, Hurwitz R, Zenk T, Desnick RJ, Ferlinz K, Schuchman EH and Sandhoff K.
Purification, characterization, and biosynthesis of human acid ceramidase. J Biol Chem.
1995;270:11098-11102.
15. Shtraizent N, Eliyahu E, Park JH, He X, Shalgi R and Schuchman EH. Autoproteolytic
cleavage and activation of human acid ceramidase. J Biol Chem. 2008;283:11253-11259.
16. Gebai A, Gorelik A, Li Z, Illes K and Nagar B. Structural basis for the activation of acid
ceramidase. Nat Commun. 2018;9:1621.
Downloaded from http://ahajournals.org by on January 30, 2020
10.1161/CIRCULATIONAHA.119.041882
24
17. Mann DL. Sphingosine 1-phosphate as a therapeutic target in heart failure: more
questions than answers. Circulation. 2012;125:2692-2694.
18. Waeber C and Walther T. Sphingosine-1-phosphate as a potential target for the treatment
of myocardial infarction. Circ J. 2014;78:795-802.
19. Harris CM, Mittelstadt S, Banfor P, Bousquet P, Duignan DB, Gintant G, Hart M, Kim Y
and Segreti J. Sphingosine-1-Phosphate (S1P) Lyase Inhibition Causes Increased Cardiac S1P
Levels and Bradycardia in Rats. J Pharmacol Exp Ther. 2016;359:151-158.
20. Santos-Gallego CG, Vahl TP, Goliasch G, Picatoste B, Arias T, Ishikawa K, Njerve IU,
Sanz J, Narula J, Sengupta PP, Hajjar RJ, Fuster V and Badimon JJ. Sphingosine-1-Phosphate
Receptor Agonist Fingolimod Increases Myocardial Salvage and Decreases Adverse
Postinfarction Left Ventricular Remodeling in a Porcine Model of Ischemia/Reperfusion.
Circulation. 2016;133:954-966.
21. Holland WL, Miller RA, Wang ZV, Sun K, Barth BM, Bui HH, Davis KE, Bikman BT,
Halberg N, Rutkowski JM, Wade MR, Tenorio VM, Kuo MS, Brozinick JT, Zhang BB,
Birnbaum MJ, Summers SA and Scherer PE. Receptor-mediated activation of ceramidase
activity initiates the pleiotropic actions of adiponectin. Nat Med. 2011;17:55-63.
22. Huang CL, Leblond AL, Turner EC, Kumar AH, Martin K, Whelan D, O'Sullivan DM
and Caplice NM. Synthetic chemically modified mrna-based delivery of cytoprotective factor
promotes early cardiomyocyte survival post-acute myocardial infarction. Mol Pharm.
2015;12:991-996.
23. Turnbull IC, Eltoukhy AA, Fish KM, Nonnenmacher M, Ishikawa K, Chen J, Hajjar RJ,
Anderson DG and Costa KD. Myocardial Delivery of Lipidoid Nanoparticle Carrying modRNA
Induces Rapid and Transient Expression. Mol Ther. 2016;24:66-75.
24. Zangi L, Lui KO, von Gise A, Ma Q, Ebina W, Ptaszek LM, Spater D, Xu H,
Tabebordbar M, Gorbatov R, Sena B, Nahrendorf M, Briscoe DM, Li RA, Wagers AJ, Rossi DJ,
Pu WT and Chien KR. Modified mRNA directs the fate of heart progenitor cells and induces
vascular regeneration after myocardial infarction. Nat Biotechnol. 2013;31:898-907.
25. Zangi L, Oliveira MS, Ye LY, Ma Q, Sultana N, Hadas Y, Chepurko E, Spater D, Zhou
B, Chew WL, Ebina W, Abrial M, Wang QD, Pu WT and Chien KR. Insulin-Like Growth Factor
1 Receptor-Dependent Pathway Drives Epicardial Adipose Tissue Formation After Myocardial
Injury. Circulation. 2017;135:59-72.
26. Kondrat J, Sultana N and Zangi L. Synthesis of Modified mRNA for Myocardial
Delivery. Methods Mol Biol. 2017;1521:127-138.
27. Sultana N, Magadum A, Hadas Y, Kondrat J, Singh N, Youssef E, Calderon D, Chepurko
E, Dubois N, Hajjar RJ and Zangi L. Optimizing Cardiac Delivery of Modified mRNA. Mol
Ther. 2017;25:1306-1315.
28. Magadum A, Singh N, Kurian AA, Sharkar MTK, Chepurko E and Zangi L. Ablation of
a Single N-Glycosylation Site in Human FSTL 1 Induces Cardiomyocyte Proliferation and
Cardiac Regeneration. Mol Ther Nucleic Acids. 2018;13:133-143.
29. Hulsmans M, Sager HB, Roh JD, Valero-Munoz M, Houstis NE, Iwamoto Y, Sun Y,
Wilson RM, Wojtkiewicz G, Tricot B, Osborne MT, Hung J, Vinegoni C, Naxerova K, Sosnovik
DE, Zile MR, Bradshaw AD, Liao R, Tawakol A, Weissleder R, Rosenzweig A, Swirski FK,
Sam F and Nahrendorf M. Cardiac macrophages promote diastolic dysfunction. J Exp Med.
2018;215:423-440.
Downloaded from http://ahajournals.org by on January 30, 2020
10.1161/CIRCULATIONAHA.119.041882
25
30. Du J, Yuan Z, Ma Z, Song J, Xie X and Chen Y. KEGG-PATH: Kyoto encyclopedia of
genes and genomes-based pathway analysis using a path analysis model. Mol Biosyst.
2014;10:2441-2447.
31. Bhabak KP, Kleuser B, Huwiler A and Arenz C. Effective inhibition of acid and neutral
ceramidases by novel B-13 and LCL-464 analogues. Bioorg Med Chem. 2013;21:874-882.
32. Pizzirani D, Bach A, Realini N, Armirotti A, Mengatto L, Bauer I, Girotto S, Pagliuca C,
De Vivo M, Summa M, Ribeiro A and Piomelli D. Benzoxazolone carboxamides: potent and
systemically active inhibitors of intracellular acid ceramidase. Angew Chem Int Ed Engl.
2015;54:485-489.
33. Rex K, Jeffries S, Brown ML, Carlson T, Coxon A, Fajardo F, Frank B, Gustin D, Kamb
A, Kassner PD, Li S, Li Y, Morgenstern K, Plant M, Quon K, Ruefli-Brasse A, Schmidt J,
Swearingen E, Walker N, Wang Z, Watson JE, Wickramasinghe D, Wong M, Xu G and Wesche
H. Sphingosine kinase activity is not required for tumor cell viability. PLoS One. 2013;8:e68328.
34. Liu H, Chang DW and Yang X. Interdimer processing and linearity of procaspase-3
activation. A unifying mechanism for the activation of initiator and effector caspases. J Biol
Chem. 2005;280:11578-11582.
35. Kuleshov MV, Jones MR, Rouillard AD, Fernandez NF, Duan Q, Wang Z, Koplev S,
Jenkins SL, Jagodnik KM, Lachmann A, McDermott MG, Monteiro CD, Gundersen GW and
Ma'ayan A. Enrichr: a comprehensive gene set enrichment analysis web server 2016 update.
Nucleic Acids Res. 2016;44:W90-W97.
36. Chen EY, Tan CM, Kou Y, Duan Q, Wang Z, Meirelles GV, Clark NR and Ma'ayan A.
Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool. BMC
Bioinformatics. 2013;14:128.
37. Katz MG, Hadas Y, Fargnoli AS, Hadas S, Zangi L and Bridges CR. [Gene Therapy
Potential as a Treatment for Heart Failure]. Harefuah. 2018;157:112-116.
38. Laaksonen R, Ekroos K, Sysi-Aho M, Hilvo M, Vihervaara T, Kauhanen D, Suoniemi M,
Hurme R, Marz W, Scharnagl H, Stojakovic T, Vlachopoulou E, Lokki ML, Nieminen MS,
Klingenberg R, Matter CM, Hornemann T, Juni P, Rodondi N, Raber L, Windecker S, Gencer B,
Pedersen ER, Tell GS, Nygard O, Mach F, Sinisalo J and Luscher TF. Plasma ceramides predict
cardiovascular death in patients with stable coronary artery disease and acute coronary
syndromes beyond LDL-cholesterol. Eur Heart J. 2016;37:1967-1976.
39. Wang DD, Toledo E, Hruby A, Rosner BA, Willett WC, Sun Q, Razquin C, Zheng Y,
Ruiz-Canela M, Guasch-Ferre M, Corella D, Gomez-Gracia E, Fiol M, Estruch R, Ros E,
Lapetra J, Fito M, Aros F, Serra-Majem L, Lee CH, Clish CB, Liang L, Salas-Salvado J,
Martinez-Gonzalez MA and Hu FB. Plasma Ceramides, Mediterranean Diet, and Incident
Cardiovascular Disease in the PREDIMED Trial (Prevencion con Dieta Mediterranea).
Circulation. 2017;135:2028-2040.
40. Usta E, Mustafi M, Artunc F, Walker T, Voth V, Aebert H and Ziemer G. The challenge
to verify ceramide's role of apoptosis induction in human cardiomyocytes--a pilot study. J
Cardiothorac Surg. 2011;6:38.
41. Bielawska AE, Shapiro JP, Jiang L, Melkonyan HS, Piot C, Wolfe CL, Tomei LD,
Hannun YA and Umansky SR. Ceramide is involved in triggering of cardiomyocyte apoptosis
induced by ischemia and reperfusion. Am J Pathol. 1997;151:1257-1263.
42. Levkau B. Cardiovascular effects of sphingosine-1-phosphate (S1P). Handb Exp
Pharmacol. 2013:147-170.
Downloaded from http://ahajournals.org by on January 30, 2020
10.1161/CIRCULATIONAHA.119.041882
26
43. Hassler DF and Bell RM. Ceramidases: enzymology and metabolic roles. Adv Lipid Res.
1993;26:49-57.
44. Siskind LJ, Fluss S, Bui M and Colombini M. Sphingosine forms channels in membranes
that differ greatly from those formed by ceramide. J Bioenerg Biomembr. 2005;37:227-236.
45. Elrick MJ, Fluss S and Colombini M. Sphingosine, a product of ceramide hydrolysis,
influences the formation of ceramide channels. Biophys J. 2006;91:1749-1756.
46. Guicciardi ME, Leist M and Gores GJ. Lysosomes in cell death. Oncogene.
2004;23:2881-2890.
47. Benaim G, Pimentel AA, Felibertt P, Mayora A, Colman L, Sojo F, Rojas H and De
Sanctis JB. Sphingosine inhibits the sarco(endo)plasmic reticulum Ca(2+)-ATPase (SERCA)
activity. Biochem Biophys Res Commun. 2016;473:572-577.
48. del Monte F, Williams E, Lebeche D, Schmidt U, Rosenzweig A, Gwathmey JK,
Lewandowski ED and Hajjar RJ. Improvement in survival and cardiac metabolism after gene
transfer of sarcoplasmic reticulum Ca(2+)-ATPase in a rat model of heart failure. Circulation.
2001;104:1424-1429.
49. Hajjar RJ, Kang JX, Gwathmey JK and Rosenzweig A. Physiological effects of
adenoviral gene transfer of sarcoplasmic reticulum calcium ATPase in isolated rat myocytes.
Circulation. 1997;95:423-429.
50. Cannavo A, Liccardo D, Komici K, Corbi G, de Lucia C, Femminella GD, Elia A,
Bencivenga L, Ferrara N, Koch WJ, Paolocci N and Rengo G. Sphingosine Kinases and
Sphingosine 1-Phosphate Receptors: Signaling and Actions in the Cardiovascular System. Front
Pharmacol. 2017;8:556.
51. Duan HF, Wang H, Yi J, Liu HJ, Zhang QW, Li LB, Zhang T, Lu Y, Wu CT and Wang
LS. Adenoviral gene transfer of sphingosine kinase 1 protects heart against ischemia/reperfusion-
induced injury and attenuates its postischemic failure. Hum Gene Ther. 2007;18:1119-1128.
52. Xia P, Wang L, Moretti PA, Albanese N, Chai F, Pitson SM, D'Andrea RJ, Gamble JR
and Vadas MA. Sphingosine kinase interacts with TRAF2 and dissects tumor necrosis factor-
alpha signaling. J Biol Chem. 2002;277:7996-8003.
53. Guo X, Yin H, Li L, Chen Y, Li J, Doan J, Steinmetz R and Liu Q. Cardioprotective Role
of Tumor Necrosis Factor Receptor-Associated Factor 2 by Suppressing Apoptosis and
Necroptosis. Circulation. 2017;136:729-742.
54. Teichgraber V, Ulrich M, Endlich N, Riethmuller J, Wilker B, De Oliveira-Munding CC,
van Heeckeren AM, Barr ML, von Kurthy G, Schmid KW, Weller M, Tummler B, Lang F,
Grassme H, Doring G and Gulbins E. Ceramide accumulation mediates inflammation, cell death
and infection susceptibility in cystic fibrosis. Nat Med. 2008;14:382-391.
55. Corriden R, Hollands A, Olson J, Derieux J, Lopez J, Chang JT, Gonzalez DJ and Nizet
V. Tamoxifen augments the innate immune function of neutrophils through modulation of
intracellular ceramide. Nat Commun. 2015;6:8369.
56. Moscinski LC and Hill B. Molecular cloning of a novel myeloid granule protein. J Cell
Biochem. 1995;59:431-442.
57. Liu J, Wang H and Li J. Inflammation and Inflammatory Cells in Myocardial Infarction
and Reperfusion Injury: A Double-Edged Sword. Clin Med Insights Cardiol. 2016;10:79-84.
58. Horckmans M, Ring L, Duchene J, Santovito D, Schloss MJ, Drechsler M, Weber C,
Soehnlein O and Steffens S. Neutrophils orchestrate post-myocardial infarction healing by
polarizing macrophages towards a reparative phenotype. Eur Heart J. 2017;38:187-197.
Downloaded from http://ahajournals.org by on January 30, 2020
10.1161/CIRCULATIONAHA.119.041882
27
59. van Amerongen MJ, Harmsen MC, van Rooijen N, Petersen AH and van Luyn MJ.
Macrophage depletion impairs wound healing and increases left ventricular remodeling after
myocardial injury in mice. Am J Pathol. 2007;170:818-829.
60. Liehn EA, Tuchscheerer N, Kanzler I, Drechsler M, Fraemohs L, Schuh A, Koenen RR,
Zander S, Soehnlein O, Hristov M, Grigorescu G, Urs AO, Leabu M, Bucur I, Merx MW,
Zernecke A, Ehling J, Gremse F, Lammers T, Kiessling F, Bernhagen J, Schober A and Weber
C. Double-edged role of the CXCL12/CXCR4 axis in experimental myocardial infarction. J Am
Coll Cardiol. 2011;58:2415-2423.
61. Montecucco F, Lenglet S, Braunersreuther V, Pelli G, Pellieux C, Montessuit C, Lerch R,
Deruaz M, Proudfoot AE and Mach F. Single administration of the CXC chemokine-binding
protein Evasin-3 during ischemia prevents myocardial reperfusion injury in mice. Arterioscler
Thromb Vasc Biol. 2010;30:1371-1377.
62. Bielawski J, Szulc ZM, Hannun YA and Bielawska A. Simultaneous quantitative analysis
of bioactive sphingolipids by high-performance liquid chromatography-tandem mass
spectrometry. Methods. 2006;39:82-91.
63. Yang L, Soonpaa MH, Adler ED, Roepke TK, Kattman SJ, Kennedy M, Henckaerts E,
Bonham K, Abbott GW, Linden RM, Field LJ and Keller GM. Human cardiovascular progenitor
cells develop from a KDR+ embryonic-stem-cell-derived population. Nature. 2008;453:524-528.
Downloaded from http://ahajournals.org by on January 30, 2020
10.1161/CIRCULATIONAHA.119.041882
28
Figure Legends
Figure 1. Characterizing cell death dynamics and sphingolipid metabolism in mouse hearts
after MI. A, Hearts were harvested from sham-operated mice or 1, 2, 4 and 28 days post MI. B,
TUNEL assays were performed to assess DNA fragmentation in hearts harvested from either
sham-operated mice or 1, 2, 4 or 28 days post MI. Troponin-I immunostaining was used to
distinguish between cardiomyocytes and non-cardiomyocytes. Percentage of dead cells in left
ventricle at day 1, 2, 4 and 28 post MI was quantified within all cardiac cells (DAPI+ cells) n=3
(C); CMs only (DAPI+, Troponin I+) n=3 (D) and non-CM cells only (DAPI+, Troponin I-) n=3
(E). F, For RNASeq, protein analysis and mass spectrometry, hearts were harvested from sham-
operated mice or 4 or 24 hours post MI. RNA, proteins and lipids were extracted for sphingolipid
determination. G, Hierarchical clustering dendrogram for the sphingolipid signaling pathway
transcriptome in sham-operated hearts and hearts harvested 4 or 24 hours post MI, n=3, 3, 4,
respectively. H, Sphingolipid levels measured in the LV of sham-operated mice or 24 hours post
MI, n=3. I, Western blot of the AC precursor, AC active subunit β and Sphk1 in the LV of sham-
operated hearts and hearts harvested 4 or 24 hours post MI. J, Quantified protein levels of AC
precursor, AC β subunit and Sphk1, n=4. K, HPLC-MS/MS determination of AC activity in the
LV of sham-operated hearts and hearts harvested 4 or 24 hours post MI, n=3. *, P<0.05, One-
way ANOVA, Tukey's Multiple Comparison Test for (J&K) and Holm-Sidak correction for
multiple comparisons (H). Scale bar = 50μm. The results include two independent experiments
for H-K.
Downloaded from http://ahajournals.org by on January 30, 2020
10.1161/CIRCULATIONAHA.119.041882
29
Figure 2. Inhibiting sphingolipid metabolism increases cell death in cardiac cells in vitro
and after MI. A, Primary cardiomyocytes were isolated from the hearts of two- to three-day-old
rats. Two days after isolation, the cells were treated with ceramidase inhibitors (pan-ceramidase
inhibitor B13 and AC-specific ARN 14974) or sphingosine kinase inhibitor (SK1-II) and
transferred either to normoxia (21% oxygen) for 48 hours or hypoxia (<2% oxygen) for 24 or 48
hours. At the end of the incubation period, the cells were stained with an apoptosis marker
(Annexin 5) and a cell viability marker (DAPI) to assess the effects of inhibitors and oxygen
levels on cell death. Cell death was quantified in cells treated with chemical inhibitors under
normoxia for 48 hours, n=4 (B), or hypoxia for 24 hours, n=4 (C) and 48 hours, n=4 (D). E, AC
inhibitor was injected intraperitoneally at the time of MI and 7 hours post MI. Hearts were
harvested 24 hours post MI. Proteins were extracted for AC activity assays, or tissue was fixed
and stained with TUNEL to assess cell death in the LV. F, HPLC-MS/MS determined AC
activity in LV lysates 24 hours post MI and treatment with various AC inhibitor concentrations,
n=3. G, Representative images of heart sections from mice treated with AC inhibitor or DMSO
(control) obtained 24 hours post MI. H, Quantified cell death levels in the LV after treatment
with AC inhibitor or DMSO control, 24 hours post MI, n=4. I, Short-term post MI survival curve
for mice injected with AC inhibitor or DMSO control (n=8). ****, P<0.0001,***, P<0.001,**,
P<0.01, *, P<0.05, NS, not significant. One-way ANOVA, Tukey's Multiple Comparison Test
(B-D&F), two-tailed Student’s t-test (H), Mantel-Cox log-rank test (I). Scale bar 1mm.
Figure 3. Acid ceramidase overexpression using modRNA decreases cell death in cardiac
cells in vitro and after MI. A, Primary cardiomyocytes were isolated from the hearts of two- to
three-day-old rats. Two days after isolation, the cells were transfected with modRNAs for AC
Downloaded from http://ahajournals.org by on January 30, 2020
10.1161/CIRCULATIONAHA.119.041882
30
and SphK1. B, 18 hours post transfection in normoxia, CMs were fixed and immunostained to
confirm modRNA translation. C, Cell death levels of CMs transfected with the nGFP modRNA
(control), AC modRNA and SphK1 modRNA after 48 hours under hypoxia, n=4. D, modRNAs
for Luc, AC and Sphk1 were injected into mouse hearts at the time of MI. To evaluate injected
modRNAs effects on the left ventricle, immunostaining (24 hours post MI), AC activity (24
hours post MI) and TUNEL assays were performed. E, Immunostaining for AC and SphK1 24
hours post modRNA injection. F, HPLC-MS/MS measurement of AC activity in the LV 24
hours post modRNA injection and MI, n=3 . G-H, Quantification and representative images of
cell death levels in heart sections from mice treated with Luc modRNA (control), AC modRNA,
Sphk1 modRNA or combination of AC + Sphk1 modRNAs 48 hours post injection, n=7. ****,
P<0.0001, **, P<0.01, *, P<0.05, NS, not significant. One-way ANOVA, Tukey's Multiple
Comparison Test (C & H), Two-tailed Student’s t-test (F). Scale bars; 10μm (B), 50μm (E) and
1mm (G).
Figure 4. AC modRNA improves heart function and mouse survival post MI. A, modRNAs
for Luc, AC, Sphk1 or a combination of modRNAs for AC and Sphk1 were injected into the LV
immediately after MI. Heart function was assessed by echocardiography and MRI. At day 29
post MI, hearts were collected for histological analysis. B-C, Echochardiography to measure left
ventricular internal dimension end diastole (LVIDd) and left ventricular internal dimension end
systole (LVIDs) 28 days post MI, n=6. D, Echocardiography to determine changes in left
ventricle fractional shortening (FS) between day 2 and day 28 post MI, n=6 . E, Representative
images of Masson’s trichrome staining and F, percentage of left ventricle area occupied by scar
tissue 28 days post MI and modRNA injection, n=6. G-H, Representative images of MRI scans
Downloaded from http://ahajournals.org by on January 30, 2020
10.1161/CIRCULATIONAHA.119.041882
31
and quantification of left ventricle ejection fraction (LVEF) 29 days post MI and injection with
AC or Luc modRNA. I, MRI-based measurement of fractional wall thickening percentage 29
days post MI and injection with AC or Luc modRNA, n=6. J, Long-term survival of mice after
MI and transfection with different modRNAs, n=10. ****, P<0.0001, **, P<0.01, *, P<0.05,
NS, not significant. One-way ANOVA, Bonferroni post-hoc tests (B, C, D, F) and two-tailed
Student’s t-tests (H). The results include two independent experiments. Scale bar = 1mm.
Figure 5. AC modRNA reduces ceramide levels and alters the immune cell composition in
the left ventricle post MI. A, modRNAs for Luc or AC were injected into mouse hearts
immediately after MI. Lipids and proteins were extracted from the hearts 24 hours post MI to
analyze sphingolipids by mass spectrometry and proteins by western blot. For RNA sequencing,
hearts were harvested 4, 24 or 48 hours post MI. B, Sphingolipid levels after Luc or AC
modRNA treatment, assessed 24 hours post MI, n=3. C, Western blot and quantification of AC α
subunits and caspase 3 dimers in sham-operated hearts and in hearts treated with Luc or AC
modRNA, 24 hours post MI, n=3. D, Scatterplot and GO enrichment analysis of all genes
downregulated in AC-treated vs. Luc-treated out of all upregulated genes 2 days post MI, n=4. E,
Relative Ngp gene expression levels 2 days post MI in the LV of mice treated with Luc or AC,
quantified using qPCR, n=4. F, To analyze modRNA treatment’s effect on immune cell content
after MI, hearts were collected at 2, 7 and 14 days post MI and Luc or AC modRNA injection
and analyzed using flow cytometry. G, Gating strategy for immune cell populations in the infarct
zone of modRNA-treated hearts. Neutrophils are defined as CD45+ CD11b+ Ly6G+, macrophages
as CD45+ CD11b+ Lin- F4/80+ Ly6Clow/int and monocytes as CD45+ CD11b+ Lin- F4/80- Ly6Clow /
Ly6Chi. H, Representative plots of neutrophil populations in Luc modRNA or AC modRNA-
Downloaded from http://ahajournals.org by on January 30, 2020
10.1161/CIRCULATIONAHA.119.041882
32
treated hearts 2 days post MI. I-N, Flow cytometric quantification of immune cell subsets in the
infarct zone of Luc modRNA- and AC modRNA-treated hearts 2 days post MI included
following populations: neutrophils, n=16 (I); Ly6Clow monocytes, n=16 (J); Ly6Chi monocytes,
n=16 (K); total macrophages, n=16 (L), MHCII-, n=16 (M) and MHCII+, n=16 (N) macrophage
subsets, n=16. O, Summary of proposed AC modRNA action mechanism during and after acute
MI. ***, P<0.001, *, P<0.05, n=3 (B, C), n=4 (E), n=16-18 (I-N). Holm-Sidak correction for
multiple comparisons (B), one-way ANOVA, Tukey's Multiple Comparison Test (C) and two-
tailed Student’s t-tests (E&I-N). Scale bar 20μm. The results include two and three (FACS)
independent experiments.
Downloaded from http://ahajournals.org by on January 30, 2020
Downloaded from http://ahajournals.org by on January 30, 2020
Downloaded from http://ahajournals.org by on January 30, 2020
Downloaded from http://ahajournals.org by on January 30, 2020
Downloaded from http://ahajournals.org by on January 30, 2020
Downloaded from http://ahajournals.org by on January 30, 2020
... 117 Further research has shown that activating CDases can accelerate the hydrolysis of Cer, thereby reducing cardiomyocyte apoptosis, improving overall cardiac function, and extending survival post-MI. 30 Additionally, S1P interacts with S1PR2 and S1PR3, playing a critical role in maintaining the normal conduction of action potentials in cardiomyocytes. 118 Furthermore, S1P mediates the mTOR signaling pathway, inducing autophagy in cardiomyocytes post-MI, effectively preventing adverse remodeling and demonstrating significant cardioprotective effects. ...
Article
Full-text available
Sphingolipids are essential components of cell membranes and lipoproteins. They are synthesized de novo in the endoplasmic reticulum and subsequently undergo various enzymatic modifications in different organelles, giving rise to a diverse range of biologically active compounds. These molecules play a critical role in regulating cell growth, senescence, migration, apoptosis, and signaling. In recent years, the sphingolipid metabolic pathway has been recognized as a key factor in heart failure (HF) pathophysiology. Abnormal levels of sphingolipid metabolites, such as ceramide (Cer) and sphingomyelin (SM), contribute to oxidative stress and inflammatory responses, ultimately promoting cardiomyocyte apoptosis. Conversely, sphingosine-1-phosphate (S1P) and ceramide-1-phosphate (C1P) regulate vascular function and influence cardiac remodeling. Additionally, enzymes such as diacylglycerol acyltransferase 1 (DGAT1) and sphingosine-1-phosphate lyase 1 (SGPL1) modulate cardiac lipid metabolism. Given their role in HF progression, monitoring sphingolipid alterations offers potential as valuable biomarkers for assessing disease severity, prognosis, and diagnosis. Given the complexity of sphingolipid metabolism and its involvement in diverse regulatory biological processes, a comprehensive understanding of its roles at both the cellular and organismal levels in physiopathology remains incomplete. Therefore, this review aims to explore the physiological functions, regulatory mechanisms, and therapeutic potential of sphingolipid metabolism. It will summarize the specific molecular mechanisms driving key pathological processes in HF, including ventricular remodeling, myocardial fibrosis, vascular dysfunction, and metabolic disorders. Finally, the review will highlight targeted sphingolipid metabolites as potential therapeutic strategies, offering new insights into HF diagnosis and treatment, with the goal of advancing adjunctive clinical therapies.
... Another study showed that different types of ceramides contribute differently to the risk of heart failure, higher plasma Cer-16 levels were associated with an increased risk of heart failure, while Cer-22 levels were associated with a reduced risk of heart failure (Lemaitre et al., 2019). Ceramide (C16, C20, C20:1, and C24) levels increased significantly 24 h after myocardial infarction, and the role of C18 was not mentioned (Hadas et al., 2020). Fecal metabolites Cer (d18:0/12:0) and Cer (d18:0/14:0) are reduced in patients with RA, which may be protective factor for RA (Yu et al., 2022). ...
Article
Full-text available
Background Doxorubicin (DOX) is a widely used chemotherapeutic agent known for its efficacy against various cancers, but its clinical application is often limited by its cardiotoxic effects. The exact mechanisms of DOX-induced cardiotoxicity remain unclear, requiring further investigation. Early diagnosis is essential to enhance the quality of life and prognosis for patients with malignancies. This study aims to identify biomarkers and therapeutic targets for DOX cardiotoxicity. Methods Heart tissue samples from 20 DOX-treated cardiotoxic mice and 19 normal controls were analyzed using liquid chromatography-mass spectrometry (LC-MS). Multivariate statistical analysis identified differential metabolites. Key metabolites were assessed using a random forest algorithm, and ROC curves evaluated diagnostic value. H9C2 rat cardiomyoblast cells were cultured to investigate the protective effects of these metabolites. Results Among 291 metabolites, significant differences emerged between cardiotoxic and normal mice. Five metabolites-4-hydroxy-valeric acid, 2-methylbutanoic acid, traumatic acid, PI (18:2 (9Z, 12Z)/0:0), and MIPC (t18:0/24:0 (2OH))-showed diagnostic potential. ROC analysis indicated excellent value for 4-hydroxy-valeric acid and PI (18:2 (9Z, 12Z)/0:0) and high discriminatory power for 2-methylbutanoic acid (AUC = 0. 99). Pathway analysis highlighted glycosylphosphatidylinositol-anchor biosynthesis, unsaturated fatty acids biosynthesis, pantothenate and CoA pathways, among others, associated with DOX-induced cardiotoxicity. In addition, we found that the differential metabolite Cer (d18:0/12:0) can improve DOX-induced myocardial cell damage and inhibit apoptosis-related protein expression at the cellular level. Conclusion Heart tissue metabolomics with LC-MS identified critical metabolites and pathways associated with DOX cardiotoxicity, suggesting biomarkers for early diagnosis and potential therapeutic targets to mitigate DOX-related cardiotoxicity and improve clinical outcomes.
... In the present study, FOXO1 was linked to sphingolipid metabolism and antigen processing and presentation. Sphingolipid metabolism is involved in the inflammatory response [60], and antigen processing and presentation suggest a role for immune modulation [61], which supports our hypothesis that immune dysfunction is an important factor in IVDD progression. In addition, FOXO1 was found to be closely related to the core clock genes PER1, PER3, CRY1, and follicular helper T cells in IVDD. ...
Article
Full-text available
Background Low back pain is a significant burden worldwide, and intervertebral disc degeneration (IVDD) is identified as the primary cause. Recent research has emphasized the significant role of circadian rhythms (CRs) and immunity in affecting intervertebral discs (IVD). However, the influence of circadian rhythms and immunity on the mechanism of IVDD remains unclear. This study aimed to identify and validate key rhythm‐related genes in IVDD and analyze their correlation with immune cell infiltration. Methods Two gene expression profiles related to IVDD and rhythm‐related genes were obtained from the Gene Expression Omnibus and GeneCards databases to identify differentially expressed rhythm‐related genes (DERGs). Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) and gene set enrichment analysis (GSEA) were conducted to explore the biological functions of these genes. LASSO regression and SVM algorithms were employed to identify hub genes. We subsequently investigated the correlation between hub rhythm‐related genes and immune cell infiltration. Finally, nucleus pulposus‐derived mesenchymal stem cells (NPMSCs) were isolated from normal and degenerative human IVD tissues. Hub rhythm‐related genes expression in NPMSCs was confirmed by real‐time quantitative PCR (RT‐qPCR). Results Six hub genes related to CRs (CCND1, FOXO1, FRMD8, NTRK2, PRRT1, and TFPI) were screened out. Immune infiltration analysis revealed that the IVDD group had significantly more M0 macrophages and significantly fewer follicular helper T cells than those of the control group. Specifically, M0 macrophages were significantly associated with FRMD8, PRRT1, and TFPI. T follicular helper cells were significantly associated with FRDM8, FOXO1, and CCND1. We further confirmed that CCND1, FRMD8, NTRK2, and TFPI were dysrhythmic within NPMSCs from degenerated IVD in vitro. Conclusion Six genes (CCND1, FOXO1, FRMD8, NTRK2, PRRT1 and TFPI) linked to circadian rhythms associated with IVDD progression, together with immunity. The identification of these DEGs may provide new insights for the diagnosis and treatment of IVDD.
... Myocardial infarction (MI) is one of the leading causes of death worldwide and is caused by a sharp decrease in or cessation of blood flow to cardiomyocytes [176,177]. Timely restoration of the coronary artery blood flow supply is a routine treatment for MI [178,179]. However, reperfusion therapy can cause MIRI, further exacerbating myocardial injury [180][181][182]. ...
Article
Full-text available
Cardiovascular diseases are the leading causes of death worldwide. However, there are still shortcomings in the currently employed treatment methods for these diseases. Therefore, exploring the molecular mechanisms underlying cardiovascular diseases is an important avenue for developing new treatment strategies. Previous studies have confirmed that metabolic and epigenetic alterations are often involved in cardiovascular diseases across patients. Moreover, metabolic and epigenetic factors interact with each other and affect the progression of cardiovascular diseases in a coordinated manner. Lactylation is a novel posttranslational modification (PTM) that links metabolism with epigenetics and affects disease progression. Therefore, analyzing the crosstalk between cellular metabolic and epigenetic factors in cardiovascular diseases is expected to provide insights for the development of new treatment strategies. The purpose of this review is to describe the relationship between metabolic and epigenetic factors in heart development and cardiovascular diseases such as heart failure, myocardial infarction, and atherosclerosis, with a focus on acylation and methylation, and to propose potential therapeutic measures.
... The enhanced activity of acid ceramidase (AC-an enzyme that catalyzes Cerhydrolysis to free fatty acids and sphingosine, which is then phosphorylated by Sphk 1 and 2 to generate S1P; modRNA-treated mice) decreases Cers and inflammation and improves heart function and survival, with reduced scar size when compared to the control mice. Moreover, in vitro experiments evidence that AC overexpression increased cardiomyocyte cell survival after hypoxia [102]. Interestingly, S1P lyase (SPL) activity was shown to be significantly upregulated in the cardiac tissue during ischemia, and the pharmacological inhibition of SPL with tetrahydroxybutylimidazole (THI) mitigated those effects [103]. ...
Article
Full-text available
Lipids are a complex entity of different molecules, among which ceramides (Cers), ubiquitous sphingolipids with remarkable biological activity, can represent a potential additive biomarker that can be used to better understand the underlying mechanisms which drive the onset and development of atherosclerotic damage and plaque vulnerability and facilitate coronary disease management, as possible risk/prognostic biomarkers and targets for therapeutic intervention. Accordingly, this review aims to discuss the available results on the role Cersplay in contributing to atherosclerosis development and acute coronary event precipitation, their impact on complications and adverse prognosis, as well as the impact of treatment options in modulating Cerlevels.
... Key enzymes in lipid metabolism, such as sphingosine kinase (SPK) and ceramide synthase, played critical roles in regulating sphingolipid levels. 82 SPK activators promoted SPH production, which in turn inhibits the abnormal neuronal excitability in epilepsy through downstream lipid signaling pathways. 83 By regulating the synthesis or degradation of these lipids, it may affect seizure frequency and intensity as well as the duration of the chronic phase of epilepsy. ...
Article
Full-text available
Epilepsy encompasses a spectrum of chronic brain disorders characterized by transient central nervous system dysfunctions induced by recurrent, aberrant, synchronized neuronal discharges. Hippocampal sclerosis (HS) is identified as the predominant pathological alteration in epilepsy, particularly in temporal lobe epilepsy. This study investigates the metabolic profiles of epileptic hippocampal tissues using proteomics and lipidomics techniques. An epilepsy model was established in Sprague–Dawley (SD) rats via intraperitoneal injection of pentylenetetrazole (PTZ), with hippocampal tissue samples subsequently extracted for histopathological examination. Proteomics analysis was conducted using isobaric tags for relative and absolute quantitation (iTRAQ) combined with liquid chromatography-tandem mass spectrometry (LC-MS/MS), while lipidomics analysis employed ultrahigh-performance liquid chromatography quadrupole time-of-flight mass spectrometry (UHPLC Q-TOF/MS). Proteomic analysis identified 144 proteins with significant differential expression in acute epileptic hippocampal tissue and 83 proteins in chronic epileptic hippocampal tissue. Key proteins, including neurofilament heavy (Nefh), vimentin (Vim), gelsolin (Gsn), NAD-dependent protein deacetylase (Sirt2), 2′,3′-cyclic-nucleotide 3′-phosphodiesterase (Cnp), myocyte enhancer factor 2D (Mef2d), and Cathepsin D (Ctsd), were pivotal in epileptic hippocampal tissue injury and validated through parallel reaction monitoring (PRM). Concurrently, lipid metabolomics analysis identified 32 metabolites with significant differential expression in acute epileptic hippocampal tissue and 61 metabolites in chronic epileptic hippocampal tissue. Bioinformatics analysis indicated that glycerophospholipid (GP) metabolism, glycosylphosphatidylinositol (GPI)-anchor biosynthesis, and glycerolipid (GL) metabolism were crucial in epileptic hippocampal tissue injury. Integrated proteomics and lipidomics analysis revealed key protein–lipid interactions in acute and chronic epilepsy and identified critical pathways such as sphingolipid signaling, autophagy, and calcium signaling. These findings provide deeper insights into the pathophysiological mechanisms of epileptic hippocampal tissue damage, potentially unveiling novel therapeutic avenues for clinicians.
Article
Ceramides are bioactive lipid mediators involved in apoptosis, inflammation, and fibrosis. This narrative review provides a concise overview of the emerging role of ceramides in cardiovascular disease with an emphasis on atherosclerotic vascular disease and heart failure, suggesting the potential use of ceramides in risk stratification and as putative therapeutic targets. Recent developments based on observational evidence and genetic associations, including Mendelian randomization studies in humans, are summarized and put into context with experimental evidence for the role of ceramides in human and animal models of disease. Emerging scores composed of ceramides and phosphatidylcholines that are based on the length and desaturation of the N-acyl chains are discussed in the light of novel data demonstrating age- and sex-specific differences. Also reviewed is the structural heterogeneity of the sphingoid bases, including non-conventional sphingolipids that are increasingly recognized for their importance in health and disease. Lastly, novel targets and potential modalities for tissue-specific transfer of drugs are discussed.
Article
Background: Arrhythmia after myocardial infarction, a common disease, has a high incidence and lethality in clinical practice, which seriously affects patients’ quality of life and survival time. Based on our previous study and available evidence, berberine plays a role in the treatment and prevention of arrhythmia after myocardial infarction. Thus, in order to clarify the specific mechanism and provide new clinical treatments, we conducted this study. Method: Firstly, we used bioinformatics analysis and system pharmacology to analyze the physicochemical properties and biological activities of berberine in the Molinspiration server. Secondly, we explored the potential molecular mechanism of arrhythmia after myocardial infarction treated with berberine by using network pharmacology technology: (1) obtaining common genes among berberine, myocardial infarction, and arrhythmia through TCMSP, TTD databases, and so forth; (2) constructing protein–protein interaction by using STRING database; (3) using g:Profiler database to conduct GO enrichment analysis of hub genes and pathways; and (4) performing molecular docking and visualization by using AutoDock and Pymol software. Finally, we applied Western blotting analysis and real‐time quantitative polymerase chain reaction to validate the expression of relevant proteins in the TGF‐ β 1‐induced cell models. Results: The results of bioinformatics analysis and system pharmacology of berberine indicated that it had wonderful bioavailability and high biological activities. The results of network pharmacology showed that (1) 70 genes related to berberine against arrhythmia after myocardial infarction were obtained, (2) 31 hub genes were obtained by constructing PPI network, and (3) GO enrichment analysis showed that hub genes were associated with mechanisms such as stimulus and cell death. The analysis of KEGG pathways, Wiki pathways, and Reactome pathways showed that the HIF‐1 signaling pathway and interleukin‐4 and interleukin‐13 signaling pathways were the most likely to exert therapeutic effects. (4) The results of molecular docking indicated that berberine most likely exerted therapeutic effects through acting on NGF. Western blotting analysis and real‐time quantitative polymerase chain reaction techniques showed that berberine could reduce the expression of NGF and α ‐SMA in TGF‐ β 1‐induced cell models, which confirmed the accuracy of the above findings. Conclusion: Berberine can reduce NGF secretion not only by inhibiting the conversion of cardiac fibroblasts to myofibroblasts but also by acting directly on myofibroblasts. Thus, the sympathetic nerve remodeling was inhibited, which can reduce the occurrence of arrhythmia after myocardial infarction. Considering its wonderful bioavailability and high biological activities, we believe that berberine can be a novel potential therapeutic agent with potential for the treatment of arrhythmia after myocardial infarction.
Article
Despite significant advances in cardiology over the past few decades, cardiovascular diseases (CVDs) remain the leading cause of global mortality and morbidity. This underscores the need for novel therapeutic interventions that go beyond symptom management to address the underlying causal mechanisms of CVDs. RNA-based therapeutics represent a new class of drugs capable of regulating specific genetic and molecular pathways, positioning them as strong candidates for targeting the root causes of a wide range of diseases. Moreover, owing to the vast diversity in RNA form and function, these molecules can be utilized to induce changes at different levels of gene expression regulation, making them suitable for a broad array of medical applications, even within a single disease context. Several RNA-based therapies are currently being investigated for their potential to address various CVD pathologies. These include treatments aimed at promoting cardiac revascularization and regeneration, preventing cardiomyocyte apoptosis, reducing harmful circulating cholesterols and fats, lowering blood pressure, reversing cardiac fibrosis and remodeling, and correcting the genetic basis of inherited CVDs. In this review, we discuss the current landscape of RNA therapeutics for CVDs, with an emphasis on their classifications, modes of action, advancements in delivery strategies and considerations for their implementation, as well as CVD targets with proven therapeutic potential.
Article
Full-text available
Adult mammalian hearts have a very limited regeneration capacity, due largely to a lack of cardiomyocyte (CM) proliferation. It was recently reported that epicardial, but not myocardial, follistatin-like 1 (Fstl1) activates CM proliferation and cardiac regeneration after myocardial infarction (MI). Furthermore, bacterially synthesized human FSTL 1 (hFSTL1) was found to induce CM proliferation, whereas hFSTL1 synthesized in mammals did not, suggesting that post-translational modifications (e.g., glycosylation) of the hFSTL1 protein affect its regenerative activity. We used modified mRNA (modRNA) technology to investigate the possible role of specific hFSTL1 N-glycosylation sites in the induction, by hFSTL1, of CM proliferation and cardiac regeneration. We found that the mutation of a single site (N180Q) was sufficient and necessary to increase the proliferation of rat neonatal and mouse adult CMs in vitro and after MI in vivo, respectively. A single administration of the modRNA construct encoding the N180Q mutant significantly increased cardiac function, decreased scar size, and increased capillary density 28 days post-MI. Overall, our data suggest that the delivery of N180Q hFSTL1 modRNA to the myocardium can mimic the beneficial effect of epicardial hFSTL1, triggering marked CM proliferation and cardiac regeneration in a mouse MI model.
Article
Full-text available
Acid ceramidase (aCDase, ASAH1) hydrolyzes lysosomal membrane ceramide into sphingosine, the backbone of all sphingolipids, to regulate many cellular processes. Abnormal function of aCDase leads to Farber disease, spinal muscular atrophy with progressive myoclonic epilepsy, and is associated with Alzheimer's, diabetes, and cancer. Here, we present crystal structures of mammalian aCDases in both proenzyme and autocleaved forms. In the proenzyme, the catalytic center is buried and protected from solvent. Autocleavage triggers a conformational change exposing a hydrophobic channel leading to the active site. Substrate modeling suggests distinct catalytic mechanisms for substrate hydrolysis versus autocleavage. A hydrophobic surface surrounding the substrate binding channel appears to be a site of membrane attachment where the enzyme accepts substrates facilitated by the accessory protein, saposin-D. Structural mapping of disease mutations reveals that most would destabilize the protein fold. These results will inform the rational design of aCDase inhibitors and recombinant aCDase for disease therapeutics.
Article
Full-text available
Macrophages populate the healthy myocardium and, depending on their phenotype, may contribute to tissue homeostasis or disease. Their origin and role in diastolic dysfunction, a hallmark of cardiac aging and heart failure with preserved ejection fraction, remain unclear. Here we show that cardiac macrophages expand in humans and mice with diastolic dysfunction, which in mice was induced by either hypertension or advanced age. A higher murine myocardial macrophage density results from monocyte recruitment and increased hematopoiesis in bone marrow and spleen. In humans, we observed a parallel constellation of hematopoietic activation: circulating myeloid cells are more frequent, and splenic ¹⁸ F-FDG PET/CT imaging signal correlates with echocardiographic indices of diastolic dysfunction. While diastolic dysfunction develops, cardiac macrophages produce IL-10, activate fibroblasts, and stimulate collagen deposition, leading to impaired myocardial relaxation and increased myocardial stiffness. Deletion of IL-10 in macrophages improves diastolic function. These data imply expansion and phenotypic changes of cardiac macrophages as therapeutic targets for cardiac fibrosis leading to diastolic dysfunction.
Article
Full-text available
The sphingosine kinases 1 and 2 (SphK1 and 2) catalyze the phosphorylation of the lipid, sphingosine, generating the signal transmitter, sphingosine 1-phosphate (S1P). The activation of such kinases and the subsequent S1P generation and secretion in the blood serum of mammals represent a major checkpoint in many cellular signaling cascades. In fact, activating the SphK/S1P system is critical for cell motility and proliferation, cytoskeletal organization, cell growth, survival, and response to stress. In the cardiovascular system, the physiological effects of S1P intervene through the binding and activation of a family of five highly selective G protein-coupled receptors, called S1PR1-5. Importantly, SphK/S1P signal is present on both vascular and myocardial cells. S1P is a well-recognized survival factor in many tissues. Therefore, it is not surprising that the last two decades have seen a flourishing of interest and investigative efforts directed to obtain additional mechanistic insights into the signaling, as well as the biological activity of this phospholipid, and of its receptors, especially in the cardiovascular system. Here, we will provide an up-to-date account on the structure and function of sphingosine kinases, discussing the generation, release, and function of S1P. Keeping the bull’s eye on the cardiovascular system, we will review the structure and signaling cascades and biological actions emanating from the stimulation of different S1P receptors. We will end this article with a summary of the most recent, experimental and clinical observations targeting S1PRs and SphKs as possible new therapeutic avenues for cardiovascular disorders, such as heart failure.
Article
Full-text available
Abnormal lipid metabolism may contribute to myocardial injury and remodeling. To determine whether accumulation of very long-chain ceramides occurs in human failing myocardium, we analyzed myocardial tissue and serum from patients with severe heart failure (HF) undergoing placement of left ventricular assist devices and controls. Lipidomic analysis revealed increased total and very long-chain ceramides in myocardium and serum of patients with advanced HF. After unloading, these changes showed partial reversibility. Following myocardial infarction (MI), serine palmitoyl transferase (SPT), the rate-limiting enzyme of the de novo pathway of ceramide synthesis, and ceramides were found increased. Blockade of SPT by the specific inhibitor myriocin reduced ceramide accumulation in ischemic cardiomyopathy and decreased C16, C24:1, and C24 ceramides. SPT inhibition also reduced ventricular remodeling, fibrosis, and macrophage content following MI. Further, genetic deletion of the SPTLC2 gene preserved cardiac function following MI. Finally, in vitro studies revealed that changes in ceramide synthesis are linked to hypoxia and inflammation. In conclusion, cardiac ceramides accumulate in the failing myocardium, and increased levels are detectable in circulation. Inhibition of de novo ceramide synthesis reduces cardiac remodeling. Thus, increased de novo ceramide synthesis contributes to progressive pathologic cardiac remodeling and dysfunction.
Article
Full-text available
Purpose of review: The objective of this review was to summarize evidence gathered for the prognostic value of routine and novel blood lipids and lipoproteins measured in patients with acute coronary syndromes (ACS). Recent findings: Data supports clear association with risk and actionable value for non-high-density lipoprotein (Non-HDL) cholesterol and plasma ceramides in a setting of ACS. The prognostic value and clinical actionability of apolipoprotein B (apoB) and lipoprotein(a) [Lp(a)] in ACS have not been thoroughly tested, while the data for omega-3 fatty acids and oxidized low-density lipoprotein (Ox-LDL) are either untested or more varied. Measuring basic lipids, which should include Non-HDL cholesterol, at the time of presentation for ACS is guideline mandated. Plasma ceramides also provide useful information to guide both treatment decisions and follow-up. Additional studies targeting ACS patients are necessary for apoB, Lp(a), omega-3 fatty acids, and Ox-LDL.
Article
Full-text available
Modified mRNA (modRNA) is a new technology in the field of somatic gene transfer that has been used for the delivery of genes into different tissues, including the heart. Our group and others have shown that modRNAs injected into the heart are robustly translated into the encoded protein and can potentially improve outcome in heart injury models. However, the optimal compositions of the modRNA and the reagents necessary to achieve optimal expression in the heart have not been characterized yet. In this study, our aim was to elucidate those parameters by testing different nucleotide modifications, modRNA doses, and transfection reagents both in vitro and in vivo in cardiac cells and tissue. Our results indicate that optimal cardiac delivery of modRNA is with N1-Methylpseudouridine-5'-Triphosphate nucleotide modification and achieved using 0.013 μg modRNA/mm(2)/500 cardiomyocytes (CMs) transfected with positively charged transfection reagent in vitro and 100 μg/mouse heart (1.6 μg modRNA/μL in 60 μL total) sucrose-citrate buffer in vivo. We have optimized the conditions for cardiac delivery of modRNA in vitro and in vivo. Using the described methods and conditions may allow for successful gene delivery using modRNA in various models of cardiovascular disease.
Article
Introduction: Advances in understanding the molecular biology of heart failure, the evolution of vector technology, as well as defining the targets for therapeutic interventions has placed heart failure within the reach of gene-based therapy. During the last decade the concept of delivering cDNA encoding a therapeutic gene to failing cardiomyocytes has moved from hypothesis to the bench of preclinical applications and clinical trials. However, despite significant promise, several obstacles exist, which are described in this review. We anticipate that advances in the field will improve gene therapy in heart failure in future clinical approaches.
Article
Background: Programmed cell death, including apoptosis, mitochondria-mediated necrosis, and necroptosis, is critically involved in ischemic cardiac injury, pathological cardiac remodeling, and heart failure progression. Whereas apoptosis and mitochondria-mediated necrosis signaling is well established, the regulatory mechanisms of necroptosis and its significance in the pathogenesis of heart failure remain elusive. Methods: We examined the role of tumor necrosis factor receptor-associated factor 2 (Traf2) in regulating myocardial necroptosis and remodeling using genetic mouse models. We also performed molecular and cellular biology studies to elucidate the mechanisms by which Traf2 regulates necroptosis signaling. Results: We identified a critical role for Traf2 in myocardial survival and homeostasis by suppressing necroptosis. Cardiac-specific deletion of Traf2 in mice triggered necroptotic cardiac cell death, pathological remodeling, and heart failure. Plasma tumor necrosis factor α level was significantly elevated in Traf2-deficient mice, and genetic ablation of TNFR1 largely abrogated pathological cardiac remodeling and dysfunction associated with Traf2 deletion. Mechanistically, Traf2 critically regulates receptor-interacting proteins 1 and 3 and mixed lineage kinase domain-like protein necroptotic signaling with the adaptor protein tumor necrosis factor receptor-associated protein with death domain as an upstream regulator and transforming growth factor β-activated kinase 1 as a downstream effector. It is important to note that genetic deletion of RIP3 largely rescued the cardiac phenotype triggered by Traf2 deletion, validating a critical role of necroptosis in regulating pathological remodeling and heart failure propensity. Conclusions: These results identify an important Traf2-mediated, NFκB-independent, prosurvival pathway in the heart by suppressing necroptotic signaling, which may serve as a new therapeutic target for pathological remodeling and heart failure.