Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2024 Feb 27;10(1):24.
doi: 10.1038/s41421-023-00622-3.

The E3 ubiquitin ligase MARCH2 protects against myocardial ischemia-reperfusion injury through inhibiting pyroptosis via negative regulation of PGAM5/MAVS/NLRP3 axis

Affiliations

The E3 ubiquitin ligase MARCH2 protects against myocardial ischemia-reperfusion injury through inhibiting pyroptosis via negative regulation of PGAM5/MAVS/NLRP3 axis

Shuolin Liu et al. Cell Discov. .

Abstract

Inflammasome activation and pyroptotic cell death are known to contribute to the pathogenesis of cardiovascular diseases, such as myocardial ischemia-reperfusion (I/R) injury, although the underlying regulatory mechanisms remain poorly understood. Here we report that expression levels of the E3 ubiquitin ligase membrane-associated RING finger protein 2 (MARCH2) were elevated in ischemic human hearts or mouse hearts upon I/R injury. Genetic ablation of MARCH2 aggravated myocardial infarction and cardiac dysfunction upon myocardial I/R injury. Single-cell RNA-seq analysis suggested that loss of MARCH2 prompted activation of NLRP3 inflammasome in cardiomyocytes. Mechanistically, phosphoglycerate mutase 5 (PGAM5) was found to act as a novel regulator of MAVS-NLRP3 signaling by forming liquid-liquid phase separation condensates with MAVS and fostering the recruitment of NLRP3. MARCH2 directly interacts with PGAM5 to promote its K48-linked polyubiquitination and proteasomal degradation, resulting in reduced PGAM5-MAVS co-condensation, and consequently inhibition of NLRP3 inflammasome activation and cardiomyocyte pyroptosis. AAV-based re-introduction of MARCH2 significantly ameliorated I/R-induced mouse heart dysfunction. Altogether, our findings reveal a novel mechanism where MARCH2-mediated ubiquitination negatively regulates the PGAM5/MAVS/NLRP3 axis to protect against cardiomyocyte pyroptosis and myocardial I/R injury.

PubMed Disclaimer

Conflict of interest statement

The authors declare no competing interest.

Figures

Fig. 1
Fig. 1. MARCH2 protein level is increased in hearts of patients with ICM and mice during myocardial I/R.
a Heatmap showing six differentially expressed E3 ligases in mouse hearts following I/R injury. b Histogram showing the relative mRNA expression of the six ubiquitin-related genes in the indicated groups. c mRNA levels of MARCH2 following I/R injury (45 min/9 h) were analyzed by RT-qPCR (normalized to β-actin). d Western blotting analysis of MARCH2 expression in healthy human hearts and patients with ICM. e Quantitated MARCH2 levels in hearts of healthy human and patients with ICM (n = 6 for each group). f Western blotting analysis of MARCH2 protein levels following I/R injury (45 min/9 h). g Quantitated MARCH2 levels following I/R injury (45 min/9 h). h Representative immunohistochemistry images showing MARCH2 protein levels in human hearts. i Representative immunohistochemistry images showing MARCH2 protein levels in sham- or I/R-treated mouse hearts. Dotted white lines indicate the boundary of the infarct area and border area. j, k Representative western blotting image (j) and quantification analysis (k) of MARCH2 expression in NMCMs subjected to hypoxia (6 h)/reoxygenation (3 h) (n = 6 for each group). l Western blotting analysis showed that MARCH2 protein could not be detected following the deletion of MARCH2 gene. m, n CK-MB level (m) and LDH activity (n) of WT and MARCH2 KO mice with or without myocardial I/R (45 min/24 h) (n = 6 for each group). o Representative images of heart sections by TTC/Evans Blue staining depicting infracted area. p Ratios of area at risk (AAR) to left ventricular (LV) area. q Infarct area normalized to AAR. Data are shown as means ± SEM. Statistical significance was examined by two-way ANOVA with Bonferroni post-test. *P < 0.05, **P < 0.01, ***P < 0.001.
Fig. 2
Fig. 2. MARCH2 KO exacerbates myocardial dysfunction after myocardial I/R injury.
a Representative M-mode recordings of echocardiography of WT and MARCH2 KO mice subjected to I/R. bf Quantitative analyses of echocardiographic measurements performed after I/R injury. n = 6 mice per group. Left ventricular end-systolic diameter (LVESD, b), left ventricular end-diastolic diameter (LVEDD, c), left ventricular end-systolic volume (LVESV, d), ejection fraction (EF, e), and fractional shortening (FS, f). gl Evaluation of mechanical properties of single, acutely isolated cardiomyocytes from I/R-treated mice. Resting cell length (g), peak shortening (h), +dL/dt: maximal velocity of shortening (i), -dL/dt: maximal velocity of re-lengthening (j), TPS90: time-to-90% peak shortening (k); TR90: time-to-90% re-lengthening (l). n = 60 cells from 3 mice per group. m Representative PI-stained images of myocardial sections from WT and MARCH2–/– mice subjected to I/R. Green: PI-positive nuclei; Red: cTnI-stained cardiomyocytes; blue, DAPI-stained nuclei; Scale bar = 20 μm. n Quantitative analysis of PI-positive cells. Experiments were repeated three times with similar results. Data are shown as the means ± SEM. *P < 0.05, **P < 0.01, ***P < 0.001.
Fig. 3
Fig. 3. PGAM5 is identified to interact with MARCH2.
a Comparison of cell type distributions among different groups showing changes in MP and cardiomyocyte clusters after I/R procedure. b, c KEGG enrichment analysis (b) and GO (c) analysis of scRNA-seq data from WT-I/R and MARCH2–/–-I/R hearts. d, e Interaction between NLRP3 and ASC in mitochondria (mito-NLRP3 and mito-ASC) of cardiac tissues of MARCH2 KO and WT mice subjected to I/R was examined by IP-western blotting assay. IP with NLRP3 antibody (d) and IP with ASC antibody (e). f Representative Western blotting analyses of MARCH2, NLRP3, ASC, caspase-1 (procaspase1; cleaved caspase-1), GSDMD (full-length; N-terminal) in cardiac tissues of MARCH2 KO and WT mice subjected to I/R (45 min/9 h). g IL-18 release was measured by enzyme-linked immunosorbent assay (ELISA) in cardiac tissues of MARCH2 KO and WT mice subjected to I/R (45 min/9 h). h Schematic diagram showing MS analysis workflow for identifying targets of MARCH2. i IP analysis with anti-HA antibody and immunoblotting with antibodies of anti-Flag and anti-HA, respectively, in NMCMs transfected with PGAM5-HA or control vector along with MARCH2-Flag. j Endogenous IP of MARCH2 and PGAM5 in NMCMs. k Direct interaction between GST-PGAM5 and His-MARCH2 demonstrated by GST pull-down assays. Both input and pull-down samples were immunoblotted with anti-GST and anti-His antibodies. GST-PGAM5 and His-MARCH2 proteins were expressed in vitro. l Representative immunofluorescence images of MARCH2 and PGAM5 in NMCMs. Data are shown as the means ± SEM. *P < 0.05, **P < 0.01, ***P < 0.001.
Fig. 4
Fig. 4. MARCH2 E3 ligase promotes degradation of PGAM5 through K48-linked polyUb.
a Western blotting analysis of PGAM5-HA and MARCH2-Flag. HL-1 cardiomyocytes were transfected with vector expressing PGAM5-HA or MARCH2-Flag and treated with MCM + H/R. b Western blotting analysis of MARCH2 and PGAM5 proteins in HL-1 cardiomyocytes transfected with MARCH2 siRNA, siRNA-resistant MARCH2 or negative control and treated with MCM + H/R. c Analysis of PGAM5-HA protein levels in MCM + H/R-treated HL-1 cardiomyocytes transfected with either MARCH2 WT or its catalytically inactive mutant (C64/67 S, abbreviated CS). d, e Western blotting analysis of MARCH2 and PGAM5 protein levels and quantification analysis of PGAM5 level. HL-1 cardiomyocytes were transfected with MARCH2 (d) or siRNA-MARCH2 (e) and then treated with MCM + H/R and cycloheximide (CHX; 30 μm). f, g Corresponding quantitation graphs of relative PGAM5 degradation in MCM + H/R-treated HL-1 cardiomyocytes transfected with MARCH2 (f) or siRNA-MARCH2 (g) and CHX. The experiments were repeated three times. Error bars represent standard deviation. h Effects of the indicated polyUb on MARCH2-mediated PGAM5 ubiquitination. HL-1 cardiomyocytes were transfected with the indicated ubiquitin under MCM + H/R treatment. IP analysis with anti-HA antibody and immunoblotting with antibodies of anti-His and anti-HA. i Western blotting of the mapping analysis showing the binding domains of MARCH2 to PGAM5. Various truncated forms of MARCH2 (△1–55, △56–116, △117–214 and △215–246) were expressed and purified. jl Effects of the indicated PGAM5 KR mutants (K88R, K141R, or combined K88-141R) on MARCH2-mediated PGAM5 ubiquitination (j), protein level (k), and degradation (l) under MCM + H/R treatment. PGAM5–/– HL-1 cardiomyocytes were transfected with the indicated constructs, and PGAM5 protein levels were analyzed. Statistical differences were analyzed by unpaired Student’s t-test for comparison between two groups. *P < 0.05, **P < 0.01.
Fig. 5
Fig. 5. PGAM5 mediates the regulation of MARCH2 on NLRP3 inflammasome assembly following myocardial I/R.
a Pearson correlation coefficients between MARCH2 and PGAM5 protein levels in ICM. b, c Representative western blotting (b) and quantitated analysis (c) of PGAM5 expression in NMCMs infected with vector or MARCH2 under MCM + H/R treatment. d Ubiquitination assays determining the ubiquitination of endogenous PGAM5 in the hearts of MARCH2 KO and WT mice subjected to I/R injury. e Immunohistochemistry for MARCH2, PGAM5, and GSDMD in hearts of MARCH2 KO and WT mice following I/R injury. f, g Representative western blotting image (f) and quantitated analysis (g) of PGAM5 in cardiac tissues of MARCH2 KO and WT mice subjected to I/R (45 min/9 h). h, i Interaction between NLRP3 and ASC was examined by IP-western blotting assay in cardiomyocytes with MARCH2 or PGAM5 overexpression following MCM + H/R challenge. IP with NLRP3 antibody (h), IP with ASC antibody (i). j Interaction between PGAM5 and MAVS was examined by IP-western blotting assay in cardiomyocytes with or without MCM + H/R treatment. k Representative immunofluorescence images of MAVS and PGAM5 in HL-1 cells. l Colocalization analysis of PGAM5–MAVS. Pearson’s R value (no threshold) was calculated by ImageJ Fiji software. n = 6 images from 3 biological replicates. Data are shown as the means ± SEM. *P < 0.05, **P < 0.01, ***P < 0.001.
Fig. 6
Fig. 6. PGAM5–MAVS co-condensates form under MCM + H/R challenge and PGAM5 promotes MAVS-dependent NLRP3 activation.
a Time-lapse images of HL-1 cells expressing PGAM5-mcherry. PGAM5 condensate fission and fusion is presented in the boxes. b FRAP analysis of PGAM5-mcherry condensates in HL-1 cells. c Quantification of FRAP in the bleached region of PGAM5-mcherry condensates, show as means ± SD (n = 6). d Time-lapse images of HL-1 cells expressing MAVS-EGFP. MAVS condensate fission and fusion is presented in the boxes. e FRAP analysis of MAVS-EGFP condensates in HL-1 cells. f Quantification of FRAP in the bleached region of PGAM5-mcherry condensates, show as means ± SD (n = 6). g FRAP analysis of MAVS-EGFP condensates in HL-1 cardiomyocytes without (upper) or with (lower) the presence of PGAM5. h Quantification of FRAP in the bleached region of MAVS-EGFP condensates with or without PGAM5, show as means ± SD (n = 6). i Representative fluorescent images of MAVS-EGFP droplets in the presence or absence of PGAM5. j Quantification of fluorescence intensity of MAVS-EGFP liquid droplets in i. k Interaction between NLRP3 and MAVS was examined by IP-western blotting assay in cardiomyocytes in the presence or absence of si-PGAM5 under MCM + H/R treatment. l Immunofluorescence of NLRP3 and MAVS in cardiomyocytes with or without PGAM5 knockdown under MCM + H/R treatment. m Colocalization analysis of NLRP3–MAVS. Pearson’s R value (no threshold) was calculated by ImageJ Fiji software. n = 6 images from 3 biological replicates. np Representative western blotting (n) and quantitated analyses of GSDMD (o) and caspase-1 (p) in cardiomyocytes infected with PGAM5 or si-MAVS under MCM + H/R treatment. q IL-18 release measured by ELISA in cardiomyocytes of the indicated groups underwent MCM + H/R treatment. rt Representative western blotting (r) and quantitated analyses of GSDMD (s) and caspase-1 (t) in cardiomyocytes infected with MARCH2, PGAM5, or MAVS under MCM + H/R treatment. u IL-18 release measured by ELISA in cardiomyocytes of the indicated groups underwent MCM + H/R treatment. Data are shown as the means ± SEM. *P < 0.05, **P < 0.01, ***P < 0.001.
Fig. 7
Fig. 7. MARCH2 overexpression reduces I/R injury and preserves cardiac function via inhibition of PGAM5–MAVS-NLRP3 inflammasome pathway.
a Representative M-mode recordings of echocardiography of WT mice injected with AAV9-cTnT-NC or AAV9-cTnT-MARCH2 (2 × 1011 V.g/mouse) for 3 weeks and then subjected to I/R (45 min/24 h). b, c Quantitative analysis of echocardiographic measurements performed in AAV9-cTnT-NC or AAV9-cTnT-MARCH2 mice subjected to I/R (45 min/24 h) injury (n = 6 mice per group). Left ventricular (LV) ejection fraction (EF, b) and fractional shortening (FS, c). d Representative images of heart sections by TTC/Evans Blue staining depicting infracted area. e Ratios of area at risk (AAR) to left ventricular (LV) area. f Infarct area normalized to AAR (n = 6 mice per group). g, h LDH activity and CK-MB level in AAV9-cTnT-NC or AAV9-cTnT-MARCH2 mice with or without myocardial I/R (45 min/24 h) (n = 6 mice per group). i Ubiquitination assays determining the ubiquitination of endogenous PGAM5 in the hearts of AAV9-cTnT-NC or AAV9-cTnT-MARCH2 mice subjected to I/R injury. j Representative western blotting analysis of PGAM5, caspase-1 (procaspase1; cleaved caspase-1), GSDMD (full-length; N-terminal), and IL-18 in hearts of AAV9-cTnT-NC or AAV9-cTnT-MARCH2 mice subjected to I/R (45 min/9 h). k Quantitated analysis of PGAM5 in hearts of AAV9-cTnT-NC or AAV9-cTnT-MARCH2 mice subjected to I/R (45 min/9 h). l, m Interaction between NLRP3 and ASC in mitochondria of cardiac tissues of AAV9-cTnT-NC or AAV9-cTnT-MARCH2 mice subjected to I/R was examined by IP-western blotting assay. n IL-18 release was measured by ELISA in cardiac tissues of AAV9-cTnT-NC or AAV9-cTnT-MARCH2 mice subjected to I/R. Data are shown as the means ± SEM. *P < 0.05, **P < 0.01, ***P < 0.001.
Fig. 8
Fig. 8. PGAM5/MAVS signaling pathway is essential for the regulation of MARCH2 in I/R injury.
a Prior to I/R injury, WT mice were administrated with AAV9-cTnT-vector, AAV9-cTnT-MARCH2, AAV9-cTnT-PGAM5 or AAV9-cTnT-MAVS (2.0 × 1011 V.g/mouse) for 3 weeks by tail vein injection. TTC/Evans Blue staining is used to depict infracted area. b Ratios of area at risk (AAR) to left ventricular (LV) area. c Infarct area normalized to AAR. No statistical significance for Vector-I/R vs MARCH2-PGAM5-I/R, no statistical significance for Vector-I/R vs MARCH2-MAVS-I/R. d, e Echocardiographic assessment of ejection fraction (EF, d) and fractional shortening (FS, e) in the indicated groups. No statistical significance for Vector-I/R vs MARCH2-PGAM5-I/R, no statistical significance for Vector-I/R vs MARCH2-MAVS-I/R. f, g LDH activity (f) and CK-MB level (g) in mouse with and without myocardial I/R. No statistical significance for Vector-I/R vs MARCH2-PGAM5-I/R; no statistical significance for Vector-I/R vs MARCH2-MAVS-I/R. h Representative western blotting results of MARCH2, PGAM5, mito-MAVS, caspase-1 (procaspase1; cleaved caspase-1), GSDMD (full-length; N-terminal), and IL-18 in hearts of the indicated group mice subjected to I/R (45 min/9 h). i IL-18 release was measured by ELISA in cardiac tissues of the indicated group mice subjected to I/R (45 min/9 h). No statistical significance for Vector-I/R vs MARCH2-PGAM5-I/R, no statistical significance for Vector-I/R vs MARCH2-MAVS-I/R. j A working model for MARCH2 function in I/R injury. MARCH2 interacts with and promotes the degradation of PGAM5 by facilitating its K48-linked polyUb, thus inhibiting the activity of MAVS/NLRP3 inflammasome pathway and cardiomyocyte pyroptosis. Data are shown as the means ± SEM. *P < 0.05, **P < 0.01, ***P < 0.001.

References

    1. Toldo S, Abbate A. The NLRP3 inflammasome in acute myocardial infarction. Nat. Rev. Cardiol. 2018;15:203–214. doi: 10.1038/nrcardio.2017.161. - DOI - PubMed
    1. Deng Y, et al. Targeting mitochondria-inflammation circuit by β-hydroxybutyrate mitigates HFpEF. Circ. Res. 2021;128:232–245. doi: 10.1161/CIRCRESAHA.120.317933. - DOI - PubMed
    1. Shi H, et al. GSDMD-mediated cardiomyocyte pyroptosis promotes myocardial I/R injury. Circ. Res. 2021;129:383–396. doi: 10.1161/CIRCRESAHA.120.318629. - DOI - PMC - PubMed
    1. Sharma BR, Kanneganti TD. NLRP3 inflammasome in cancer and metabolic diseases. Nat. Immunol. 2021;22:550–559. doi: 10.1038/s41590-021-00886-5. - DOI - PMC - PubMed
    1. de Carvalho Ribeiro M, Szabo G. Role of the inflammasome in liver disease. Annu. Rev. Pathol. 2022;17:345–365. doi: 10.1146/annurev-pathmechdis-032521-102529. - DOI - PMC - PubMed