SIRT2 inhibition protects against cardiac hypertrophy and ischemic injury

  1. Xiaoyan Yang
  2. Hsiang-Chun Chang
  3. Yuki Tatekoshi
  4. Amir Mahmoodzadeh
  5. Maryam Balibegloo
  6. Zeinab Najafi
  7. Rongxue Wu
  8. Chunlei Chen
  9. Tatsuya Sato
  10. Jason Shapiro
  11. Hossein Ardehali  Is a corresponding author
  1. Feinberg Cardiovascular and Renal Research Institute, Northwestern University School of Medicine, United States
6 figures, 2 tables and 1 additional file

Figures

Figure 1 with 1 supplement
SIRT2 is upregulated in heart failure (HF).

(A) SIRT1, SIRT2, SIRT3, and SIRT6 in mouse hearts after trans-aortic constriction (TAC). (B) SIRT2 in human hearts from healthy patients and patients with dilated cardiomyopathy. (C) SIRT2 protein levels in the hearts of control individual and patients with ischemic heart failure. *p<0.05 by Student’s t-test. Data presented as mean ± SEM.

Figure 1—figure supplement 1
SIRT2 protein in different mouse tissues, including the heart (A), and in various cell lines, including H9c2 cells (B).
Figure 2 with 1 supplement
Sirt2 deficiency protects the heart against cardiac dysfunction after trans-aortic constriction (TAC).

Sirt2-/- and wild-type (WT) littermates were subjected to TAC and ejection fraction (EF) (A), fractional shortening (FS) (B), and interventricular septal thickness during diastole (C) were assessed 4 weeks later (N=6–9). (D–F) Representative hearts (D), HW/BW (E) (N=3–5), H&E staining, (F) and the summary of cross-sectional area of cardiomyocytes (G) in WT and Sirt2-/- hearts (N=20 cardiomyocytes), *p<0.05 by one-way ANOVA and post hoc Tukey analysis (A, B, C, and E) and unpaired Student’s t-test (G). Bars represent group mean.

Figure 2—source data 1

Ejection fraction (EF) in wild-type (WT) and Sirt2-/- mice after sham or trans-aortic constriction (TAC) as shown in Figure 2A.

https://cdn.elifesciences.org/articles/85571/elife-85571-fig2-data1-v2.csv
Figure 2—source data 2

Fractional shortening (FS) in wild-type (WT) and Sirt2-/- mice after sham or trans-aortic constriction (TAC) as shown in Figure 2B.

https://cdn.elifesciences.org/articles/85571/elife-85571-fig2-data2-v2.csv
Figure 2—source data 3

Interventricular septal (IVS) thickness diastole in wild-type (WT) and Sirt2-/- mice after sham or trans-aortic constriction (TAC) as shown in Figure 2C.

https://cdn.elifesciences.org/articles/85571/elife-85571-fig2-data3-v2.csv
Figure 2—source data 4

HW/BW in wild-type (WT) and Sirt2-/- mice after sham or trans-aortic constriction (TAC) as shown in Figure 2E.

https://cdn.elifesciences.org/articles/85571/elife-85571-fig2-data4-v2.csv
Figure 2—source data 5

CSA in wild-type (WT) and Sirt2-/- hearts as shown in Figure 2G.

https://cdn.elifesciences.org/articles/85571/elife-85571-fig2-data5-v2.csv
Figure 2—figure supplement 1
Expression of protein (A) and mRNA (B) of other sirtuin family members in the hearts of Sirt2-/- mice.

N=6. Data presented as mean ± SEM.

Hearts from Sirt2-/- mice are protected against ischemia-reperfusion (I/R) injury.

Ejection fraction (EF) and fractional shortening (FS) in wild-type (WT) and Sirt2-/- mice 7 (A) and 21 days (B) after I/R (N=4–5). (C) Time course of FS in Sirt2-/- mice after I/R injury (N=4–5). (D, E) Cell death assessed by propidium iodide (PI), in neonatal rat cardiomyocyte (NRCM) treated with control or Sirt2 siRNA and with 500 µM of H2O2. *p<0.05 by ANOVA for all panels expect for panel C, where Student’s t-test was used for comparison between the two time points. Bars represent mean (A, B), and data presented as mean ± SEM.

Figure 3—source data 1

Ejection fraction (EF) and fractional shortening (FS) in wild-type (WT) and Sirt2-/- mice after ischemia-reperfusion (I/R) as shown in Figure 3A.

https://cdn.elifesciences.org/articles/85571/elife-85571-fig3-data1-v2.csv
Figure 3—source data 2

Ejection fraction (EF) and fractional shortening (FS) in wild-type (WT) and Sirt2-/- mice after ischemia-reperfusion (I/R) as shown in Figure 3B.

https://cdn.elifesciences.org/articles/85571/elife-85571-fig3-data2-v2.csv
Figure 3—source data 3

Time course of fractional shortening (FS) in wild-type (WT) and Sirt2-/- mice after ischemia-reperfusion (I/R) as shown in Figure 3C.

https://cdn.elifesciences.org/articles/85571/elife-85571-fig3-data3-v2.csv
Figure 3—source data 4

Propidium iodide (PI) positive cells as shown in Figure 3E.

https://cdn.elifesciences.org/articles/85571/elife-85571-fig3-data4-v2.csv
Figure 4 with 2 supplements
cs-Sirt2-/- hearts are protected against trans-aortic constriction (TAC) and ischemia-reperfusion (I/R).

Ejection fraction (EF) and fractional shortening (FS) in Sirt2f/f and cs-Sirt2-/- mice 7 (A) and 14 days (B) after TAC (N=5–9). (C,D) mRNA levels of Anf (C) and Bnp (D) in the hearts of Sirt2f/f and cs-Sirt2-/- mice 4 weeks after TAC (N=7–8). (E) EF and FS in Sirt2f/f and cs-Sirt2-/- mice 7 and 14 days after I/R (N=4). (F) Necrotic area (representing the degree of ischemic damage) in Sirt2f/f and cs-Sirt2-/- mice 14 days after MI. *p<0.05 by ANOVA for panels A and B, and Student’s t-test was used for panels C and D. Data are presented as mean ± SEM.

Figure 4—source data 1

Ejection fraction (EF) and fractional shortening (FS) in Sirt2f/f and cs-Sirt2-/- mice 7 days after ischemia-reperfusion (I/R) as shown in Figure 4A.

https://cdn.elifesciences.org/articles/85571/elife-85571-fig4-data1-v2.csv
Figure 4—source data 2

Ejection fraction (EF) and fractional shortening (FS) in Sirt2f/f and cs-Sirt2-/- mice 14 days after ischemia-reperfusion (I/R) as shown in Figure 4B.

https://cdn.elifesciences.org/articles/85571/elife-85571-fig4-data2-v2.csv
Figure 4—source data 3

Nppa mRNA in Sirt2f/f and cs-Sirt2-/- hearts as shown in Figure 4C.

https://cdn.elifesciences.org/articles/85571/elife-85571-fig4-data3-v2.csv
Figure 4—source data 4

Nppb mRNA in Sirt2f/f and cs-Sirt2-/- hearts as shown in Figure 4D.

https://cdn.elifesciences.org/articles/85571/elife-85571-fig4-data4-v2.csv
Figure 4—source data 5

Echo parameters in Sirt2f/f and cs-Sirt2-/- hearts as shown in Figure 4E.

https://cdn.elifesciences.org/articles/85571/elife-85571-fig4-data5-v2.xlsx
Figure 4—figure supplement 1
SIRT1, SIRT3, and SIRT2 protein in the hearts of cs-Sirt2-/- mice.
Figure 4—figure supplement 2
cs-Sirt2-/- hearts from female mice are protected against trans-aortic constriction (TAC).

Ejection fraction (EF) and fractional shortening (FS) in female wild-type (WT) and cs-Sirt2-/- mice 7 and 14days after TAC (N=4).

Figure 4—figure supplement 2—source data 1

Ejection fraction (EF) and fractional shortening (FS) in female wild-type (WT) and cs-Sirt2-/- mice 7 and 14 days after TAC as shown in Figure 4—figure supplement 2.

https://cdn.elifesciences.org/articles/85571/elife-85571-fig4-figsupp2-data1-v2.xlsx
Figure 5 with 3 supplements
SIRT2 interacts with nuclear factor (erythroid-derived 2)-like 2 (NRF2) and regulates its activity in the heart.

(A) Co-immunoprecipitation (IP) of SIRT2 and NRF2 in extracts of hearts from wild-type (WT) mice. (B) Endogenous NRF2 acetylation levels in the hearts of WT and Sirt2-/- mice at the baseline. Acetylated proteins were IPed by anti-acetyl antibody followed by immunoblotting with anti-NRF2 antibody. (C) NRF2 protein levels in neonatal rat cardiomyocytes (NRCMs) treated with Sirt2 siRNA. (D) NRF2 protein levels in H9c2 cells treated with control or Sirt2 siRNA and harvested at different time points after treatment with 100 µg/ml of CHX. (E) NRF2 protein levels in the nucleus in NRCMs treated with control or Sirt2 siRNA. (F–H) mRNA levels of NRF2 target genes in pentose phosphate pathway (F), quinone and glutathione-based detoxification (G), thioredoxin production (H) in H9c2 cells overexpressing empty vector (white bars) or SIRT2 (gray bars). *p<0.05 by Student’s t-test.

Figure 5—figure supplement 1
Effects of SIRT2 overexpression on mRNA levels of non-nuclear factor (erythroid-derived 2)-like 2 (NRF2) targeted antioxidant genes.

N=5–6. Data presented as mean ± SEM.

Figure 5—figure supplement 2
SIRT2 regulates nuclear factor (erythroid-derived 2)-like 2 (NRF2) and its target proteins.

(A) NRF2 protein levels in HL-1 cells treated with Sirt2 siRNA. (B–D) mRNA levels of NRF2 target genes in pentose phosphate pathway (B), quinone and glutathione-based detoxification (C), thioredoxin production (D) in HL-1 cells overexpressing empty vector (white bars) or SIRT2 (gray bars). *p<0.05 by Student’s t-test.

Figure 5—figure supplement 3
Reactive oxygen species (ROS) levels as assessed by dihydroethidium (DHE) staining in neonatal rat cardiomyocytes (NRCMs) treated with control or Sirt2 siRNA after treatment with 500 µM H2O2.

N=5–6. Data presented as mean ± SEM.

Nrf2 deletion and SIRT2 inhibitors protected against cardiac damage and cardiac hypertrophy.

Ejection fraction (EF) (A) and fractional shortening (FS) (B) in wild-type (WT), Sirt2-/-, and Sirt2-/-/Nrf2-/- double knockout (KO) mice 28 days after ischemia-reperfusion (I/R) (N=4–5). (C) Protocol for treatment of mice with SIRT2 inhibitor, AGK2. (D) Echo images of hearts from WT mice treated with either vehicle or AGK2. (E–J) EF (E), FS (F), left ventricular diameter during diastole (LVDd) (G), left ventricular diameter during systole (LVDs) (H), IVSd (I), and posterior wall thickness during diastole (PWTd) (J) in WT mice treated with AGK after trans-aortic constriction (TAC) according to the protocol in panel C (N=6–10). *p<0.05 by ANOVA for panels A–B or Student’s t-test for panels E–J.

Figure 6—source data 1

Ejection fraction (EF) in wild-type (WT), Sirt2-/-, and Sirt2-/-/Nrf2-/- mice after ischemia-reperfusion (I/R) as shown in Figure 6A.

https://cdn.elifesciences.org/articles/85571/elife-85571-fig6-data1-v2.csv
Figure 6—source data 2

Fractional shortening (FS) in wild-type (WT), Sirt2-/-, and Sirt2-/-/Nrf2-/- mice after ischemia-reperfusion (I/R) as shown in Figure 6B.

https://cdn.elifesciences.org/articles/85571/elife-85571-fig6-data2-v2.csv
Figure 6—source data 3

Ejection fraction (EF) with AGK2 as shown in Figure 6E.

https://cdn.elifesciences.org/articles/85571/elife-85571-fig6-data3-v2.csv
Figure 6—source data 4

Fractional shortening (FS) with AGK2 as shown in Figure 6F.

https://cdn.elifesciences.org/articles/85571/elife-85571-fig6-data4-v2.csv
Figure 6—source data 5

Left ventricular diameter during diastole (LVDd) with AGK2 as shown in Figure 6G.

https://cdn.elifesciences.org/articles/85571/elife-85571-fig6-data5-v2.csv
Figure 6—source data 6

LVDs with AGK2 as shown in Figure 6H.

https://cdn.elifesciences.org/articles/85571/elife-85571-fig6-data6-v2.csv
Figure 6—source data 7

IVSd with AGK2 as shown in Figure 6I.

https://cdn.elifesciences.org/articles/85571/elife-85571-fig6-data7-v2.csv
Figure 6—source data 8

Posterior wall thickness during diastole (PWTd) with AGK2 as shown in Figure 6J.

https://cdn.elifesciences.org/articles/85571/elife-85571-fig6-data8-v2.csv

Tables

Appendix 1—table 1
Primer sequences.
GenesForward primerReverse primerSpecies
NppaGGGTAGGATTGACAGGATTGGCCTCCTTGGCTGTTATCTTCMouse
18 sAGTCCCTGCCCTTTGTACACACGATCCGAGGGCCTCACTA
ActbCTAAGGCCAACCGTGAAAAGACCAGAGGCATACAGGGACAMouse
NppbATCCGTCAGTCGTTTGGGCAGAGTCAGAAACTGGAGTCMouse
G6pdGGCCAACCGTCTGTTCTACCTCCACTATGATGCGGTTCCAGCRat
PgdCGGGTCATACTGCTCGTGAAAGGTCCTGGCATCTTCTTGTCGRat
Nqo1CACTACGATCCGCCCCCAACGCGTGGGCCAATACAATCAGGRat
GclcGTCAAGGACCGGCACAAGGAGAACATCGCCGCCATTCAGTRat
GclmTGCCACCAGATTTGACTGCATTTTCCTGGAAACTTGCCTCAGAGAGRat
GssGAGGTCCGCAAAGAACCCCAGAGCGTGAATGGGGCATACGRat
GsrTCACCCCGATGTATCACGCTGCCCTGAAGCATCTCATCGCRat
Gpx4AGCAACAGCCACGAGTTCCTATCGATGTCCTTGGCTGCGARat
Gsta1ACTTCGATGGCAGGGGGAGAATGGAACATCAAACTCCCATCATTCCRat
Gsta2TTGACGGGATGAAGCTGGCAGTCAGATCTAAAATGCCTTCGGTGTRat
Gstm1CCAAGTGCCTGGACGCCTTCATAGGTGTTGAGAGGTAGCGGCRat
Gstp1CGTCCACGCAGCTTTGAGTGTAACCACCTCCTCCTTCCAGCRat
Txn1AGTAGACGTGGATGACTGCCAAGCACCAGAGAACTCCCCAACRat
Prdx1TCAGATCCCAAGCGCACCATAGCGGCCAACAGGAAGATCARat
Txnrd1AATGCTGGAGAGGTGACGCAGATGTCTCCCCCAGAACGCTRat
Sirt1CAGTGTCATGGTTCCTTTGCCACCGAGGAACTACCTGATMouse
Sirt3GCTGCTTCTGCGGCTCTATACGAAGGACCTTCGACAGACCGTMouse
Sirt4GTGGAAGAATAAGAATGAGCGGAGGCACAAATAACCCCGAGGMouse
Sirt5CCACCGACAGATTCAGGTTTTTCCCGTTAGTGCCCTGCTTTAMouse
Sirt6ATGTCGGTGAATTATGCAGCAGCTGGAGGACTGCCACATTAMouse
Sirt7CAGGTGTCACGCATCCTGAGGCCCGTGTAGACAACCAAGTMouse
CatCCAGCCAGCGACCAGATGAACCTATTGGGTTCCCGCCTCCRat
Sod1AACTGAAGGCGAGCATGGGTTATGCCTCTCTTCATCCGCTGGRat
Sod2GGGGCCATATCAATCACAGCAGAACCTTGGACTCCCACAGACRat
Sod3ACGTTCTTGGGAGAGCTTGTCTGCTAAGTCGACACCGGACRat
ActbGGCTCCTAGCACCATGAAGACAGTGAGGCCAGGATAGAGCRat
Hprt 1CCCTCAGTCCCAGCGTCGTGCGAGCAAGTCTTTCAGTCCTGTCCRat
B2mCCGTGATCTTTCTGGTGCTTGGAGACACGTAGCAGTTGAGGARat
G6pdGTCTTTGCTCGGTGCTTGTCAGCATAGAGGGCCTTACGGAMouse
Nqo1TCTCTGGCCGATTCAGAGTGCCAGACGGTTTCCAGACGTTMouse
GclmATGACCCGAAAGAACTGCTCTGGGTGTGAGCTGGAGTTAAGMouse
GclcACTGAATGGAGGCGATGTTCTTCAGAGGGTCGGATGGTTGGMouse
GssGCACCGACACGTTCTCAATGTAGCACCACCGCATTAGCTGMouse
GsrATGTTGACTGCCTGCTCTGGATCCGTCTGAATGCCCACTTMouse
Gpx4GTACTGCAACAGCTCCGAGTATGCACACGAAACCCCTGTAMouse
Gsta2CCAGGACTCTCACTAGACCGTCCCGGGCATTGAAGTAGTGAMouse
gstm1ATACACCATGGGTGACGCTCTCCATCCAGGTGGTGCTTTCMouse
Gstp1GTCTACGCAGCACTGAATCCGGGAGCTGCCCATACAGACAMouse
Txn1GCGCTCCGCCCTATTTCTATCCTCCTGAAAAGCTTCCTTGCMouse
Prdx1ACTGACAAACATGGTGAAGTGTGTACAAGAGTTTCTTCTGGCTGCMouse
Txnrd1GAATGGACAGTCCCATCCCGAAGCCCACGACACGTTCATCMouse
ActbTAAAACCCGGCGGCGCAGTCATCCATGGCGAACTGGTMouse
Hprt1AGAGCGTTGGGCTTACCTCATGGTTCATCATCGCTAATCACGMouse
B2mACGCCTGCAGAGTTAAGCATTGATCACATGTCTCGATCCCAGMouse
Appendix 1—key resources table
Reagent type (species) or resourceDesignationSource or referenceIdentifiersAdditional information
Strain, strain background (Mus musculus, male, C57BL/6)Sirt2 knockout miceDr. Gius Labrefer to: SIRT2 Maintains Genome Integrity and Suppresses Tumorigenesis through Regulating APC/C Activity.
Strain, strain background (Mus musculus, male, C57BL/6)Sirt2 flox/flox minceDr. Gius Labrefer to: SIRT2 deletion enhances KRAS-induced tumorigenesis in vivo by regulating K147 acetylation status
Strain, strain background (Mus musculus, male, C57BL/6)Nrf2 knockout micethe Jackson LaboratoryRRID:IMSR_JAX:017009
Strain, strain background (Rattus norvegicus domestica, female)Sprague–Dawley ratCharles River
AntibodyRabbit polyclonal SIRT1 antibodySigma07–131WB (1:1000)
AntibodyRabbit polyclonal SIRT2 antibodySigmaS8447WB (1:1000)
AntibodyRabbit mAb SIRT3 antibodyCell Signaling TechnologyRabbit mAb #5490WB (1:1000)
AntibodyRabbit polyclonal anti-HPRT antibodyProteintech150-59-1-APWB (1:5000)
AntibodyMouse monoclonal anti-GAPDH antibodyProteintech60004–1-IgWB (1:10000)
AntibodyRabbit mAb NRF2 antibodyCell Signaling TechnologyRabbit mAb #20733WB (1:1000)
AntibodyRabbit polycloncal NRF2 antibodyAbcamab31163WB (1:1000)
AntibodyHRP-conjugated donkey polyclonal anti-mouse IgG antibodyJackson
ImmunoResearch
715-035-150WB (1:5000)
AntibodyHRP-conjugated donkey polyclonal anti-rabbit IgG antibodyJackson
ImmunoResearch
711-035-152WB (1:5000)
AntibodyRabbit Anti beta Actin antibodyAbcamab8227WB (1:2000)
AntibodyRabbTBP antibodyAbcamab63766WB (1:2000)
AntibodyRabbit mAb SIRT6 antibodyCell Signaling TechnologyRabbit mAb #12486WB (1:1000)
AntibodyMouse Flag-M2 monoclonal antibodySigmaF1804WB: (1:2000); IP(1:xxx)
AntibodyAcetyl Lysine Antibody, AgaroseimmunechemICP0388-2MGIP: 1:10
Chemical compound, drugPropodium IodineSigmaP4170-10MG
Chemical compound, drugHoechst 34432Life Technology62249
Chemical compound, drugCycloheximideSigma1810
Chemical compound, drugParaformaldehydeThermo Fisher ScientificAC416780250
Chemical compound, drugRIPA BufferThermo Fisher Scientific89901
Chemical compound, drugProteaseArrest Protease InhibitorG-Biosciences786–437
Chemical compound, drug10% formalinFisher ScientificFLSF1004
Chemical compound, drugBrdUSigma19–160
Chemical compound, drugvitamin B12SigmaV2876
Chemical compound, drugFBSBio-TechneS11550
Chemical compound, drugpenicillin–streptomycinCytivaSV30010
Chemical compound, drugLipofectamine 2000 Transfection ReagentInvitrogen, Thermo Fisher Scientific11668027
Chemical compound, drugDharmafect transfection reagentHorizon2001–03
Chemical compound, drugAGK2SelleckchemS7577
Chemical compound, drughydrogen peroxideFisher ScientificH324-500
Chemical compound, drugNormal Rabbit IgGSigma12–370
Chemical compound, drugDMSOSigmaD4540
Cell lineH9c2ATCCCRL-1446
OtherDMEMCorning10-013CV
Commercial assay or kitRNA-STAT60TeltestCs-502
Commercial assay or kitDNAse IAmbionAM2222
Commercial assay or kitPerfeCTa SYBR Green FastMixQuanta95074–05 K
Commercial assay or kitqScript cDNA Synthesis KitQuanta95047–500
Commercial assay or kitSuperSignal West Pico PLUS Chemiluminescent SubstratePierce34579
Commercial assay or kitBCA Protein Assay KitPierce23225
Commercial assay or kitNE-PER Nuclear and Cytoplasmic Extraction ReagentsPiercePI78835
Commercial assay or kitTrichrome Stain (Masson) KitSigmaHT15-1KT
Commercial assay or kitProtein A AgaroseRoche11719408001
Commercial assay or kitdihydroethidium (DHE) assayThermo Fisher ScientificD11347
Sequence-based reagentRat Sirt2 siRNAHorizon DiscoveryM-082072-01-0005siGENOME Rat Sirt2 (361532) siRNA
Recombinant DNA reagentWildtype SIRT2 plasmidDr. Gius Labrefer to: SIRT2 Maintains Genome Integrity and Suppresses Tumorigenesis through Regulating APC/C Activity
Software, algorithmGraphPad PrismGraphPadVersion 9
Software, algorithmImageJNIH1.53 c

Additional files

Download links

A two-part list of links to download the article, or parts of the article, in various formats.

Downloads (link to download the article as PDF)

Open citations (links to open the citations from this article in various online reference manager services)

Cite this article (links to download the citations from this article in formats compatible with various reference manager tools)

  1. Xiaoyan Yang
  2. Hsiang-Chun Chang
  3. Yuki Tatekoshi
  4. Amir Mahmoodzadeh
  5. Maryam Balibegloo
  6. Zeinab Najafi
  7. Rongxue Wu
  8. Chunlei Chen
  9. Tatsuya Sato
  10. Jason Shapiro
  11. Hossein Ardehali
(2023)
SIRT2 inhibition protects against cardiac hypertrophy and ischemic injury
eLife 12:e85571.
https://doi.org/10.7554/eLife.85571