GSDMD is Essential for Myocardial I/R Injury and Myocardial Infarction
Cardiac I/R and MI are associated with pyroptosis and apoptosis [10, 12, 24]. We confirmed that in I/R and MI model in mouse, the expression of pyroptosis-related proteins and apoptosis-related proteins was significantly increased, but decreased after GSDMD knockout (GSDMD-KO) (Figure S1A–S1D). Echocardiography verified that GSDMD loss reduced the impairment of cardiac function in I/R or MI mice (Figure S1E–S1H). Importantly, GSDMD deficiency decreased serum lactate dehydrogenase (LDH) release (Figure S1I), oxidative stress [reactive oxygen species (ROS) levels, Figure S1J)], and the number of terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL)-positive cardiomyocytes (Figure S1K). Taken together, these observations indicated that GSDMD-related pyroptosis and apoptosis play an important role in I/R and MI. Our previous studies have confirmed that GSDMD can induce cardiomyocyte pyroptosis through the Caspase-11/GSDMD pathway. However, how GSDMD mediates cardiomyocyte apoptosis is unknown. Previous studies have shown that DIC model is difficult to induce cardiomyocyte pyroptosis. Therefore, we subsequently investigated whether cardiomyocytes undergo pyroptosis and apoptosis in DIC, and the underlying mechanisms.
DOX Treatment Exacerbates GSDMD mediated Cardiomyocyte Injury
The role of GSDMD mediated-pyroptosis and apoptosis in I/R and MI (Figure S1) indicated that GSDMD participated in the myocardial injury. DIC was also a kind of myocardial injury. To investigate the effect of DOX on cardiomyocytes in vivo, WT mice were injected intraperitoneally with DOX to induce acute cardiotoxicity. Sham mice were injected with normal saline (NS), as a control group. Samples were collected 7 d after DOX treatment. The survival rate, heart weight, and body weight (BW) of DOX-treated mice were lower than those of sham mice (Figure S2A and S2B). Consistently, DOX administration significantly impaired cardiac function (Figure S2C and S2D) and triggered an evident cardiomyocyte contractile dysfunction (Figure S2E and S2F), detected as reduced maximal velocity of shortening/relengthening (+dL/dt), peak shortening (PS), and time to peak shortening (TPS) (all at P<0.001) without affecting time to 90% relengthening (TR90). Moreover, DOX-induced myocardial injury obviously upregulated the expression of cleaved caspase-3 (CC3) and BAX, and decreased the levels of Bcl-2 (Figure S2G and S2H).
We used wheat germ agglutinin (WGA) to visualize cardiomyocyte diameter. Further, since DOX is thought to act as an electron acceptor to generate ROS, we also assessed ROS levels in mouse tissues. WGA staining revealed cardiomyocyte diameter reduction after DOX administration and ROS staining suggested more oxidative stress, as indicated by high fluorescence intensity of dihydroethidium (DHE) in cardiac tissues of the DOX-treated group (Figure S2I). We also used TUNEL assay to analyze left ventricular tissue sections 7 d post DOX administration. Compared with the sham control, the numbers of TUNEL-positive cells were increased in DOX-treated mice (Figure S2J and S2L).
The above results indicated that the DIC model was successfully constructed and DOX administration triggered adverse effects in cardiomyocytes. Finally, we probed GSDMD involvement in DIC. We found that the levels of full-length GSDMD and N-terminus of GSDMD were increased in the DOX-treated group (Figure S2K). Collectively, these observations confirmed that GSDMD plays a role in DIC.
GSDMD Deficiency Alleviates DIC and GSDMD Overexpression Aggravates DIC
To explore the biological function of GSDMD in acute animal models of DIC, we established GSDMD-deficient mice. GSDMD-KO mice and control mice underwent DOX treatment. DOX injection led to a high mortality among WT mice, and GSDMD-KO mice further declined (Fig 1A). Further, the BW reduced lower in DOX-treated KO mice than those in WT mice and DOX-treated KO mice exhibited significantly increased ratios of heart weight, accordingly (Figure S3A and S3B). The DOX-increased serum serum creatine kinase-MB (CK-MB) levels and cardiac troponin T (cTnT) activity were further enhanced in WT mice, compared with the levels in the GSDMD-KO mice (Figure 1B). Further, cardiac function reduction upon DOX injection was significantly improved in GSDMD-KO mice, with an increased ejection fraction (EF) and fractional shortening (FS) in DOX-injected GSDMD-KO mice (Figure 1C, and Figure S3C–S3E). Decreased +dL/dt max and -dL/dt min represent systolic dysfunction and diastolic dysfunction, respectively [25], and we observed that DOX exacerbated cardiac function in WT mice but the effect was not pronounced in KO mice (Figure S3G and S3H). Other contractile indexes displayed a similar tendency (Figure S3F and S3H). Consistently, WGA staining revealed that cardiomyocyte diameter decreased upon DOX treatment, with GSDMD deficiency suppressing the DIC shrinkage (Figure 1C, Figure S3I). Interestingly, LDH levels in WT and KO groups were not significantly different (Figure 1F), indicating low or no pyroptosis in DIC combining with the inapparent protein levels of IL-18 and IL-1β (Figure 1E, Figure S4I). Collectively, these observations demonstrate that GSDMD downregulation mitigates cell death after DOX administration.
Next, we infected adult mice with an adenovirus-associated serotype 9 GSDMD-overexpression construct (AAV9-GSDMD-OE) or an adenovirus-associated normal control construct (AAV-NC) to further verify the role of GSDMD in DOX cardiotoxicity. DOX treatment resulted in increased mortality, cardiac injury, as determined by serum biomarker analysis, and cardiac dysfunction of AAV9-GSDMD-OE mice compared with those in WT mice (Figure 1H–1J, Figure S3P, and Figure S4A). Furthermore, ROS levels in DOX-treated GSDMD-overexpressing mice were enhanced compared with those in the control (Figure 1J, Figure S4E). Compared with the control, the number of TUNEL-positive cardiomyocytes in GSDMD-KO mice treated with DOX was reduced (Figure 1D, Figure S3M), however, this effect was reversed by AAV9-GSDMD-OE infection (Figure 1K, Figure S4H). Further, the levels of apoptosis-related proteins BAX and CC3 were significantly increased in DOX treatment groups, and GSDMD loss attenuated this effect (Figure 1E, Figure S3K and S3L). The decrease in protein levels caused by DOX in GSDMD-KO mice was reversed by GSDMD overexpression (Figure 1L, Figure S4F and S4G). Importantly, cardiomyocyte analysis indicated that the occurrence of cell death was reduced after GSDMD deficiency but significantly increased upon GSDMD overexpression (Figure S6E). In summary, these observations indicated GSDMD involvement in DOX-related cardiomyopathy.
Interestingly, the expression of N-GSDMD was significantly increased after DOX treatment, but the level of cardiomyocyte pyroptosis was not changed. GSDMD deficiency alleviates cardiomyocyte apoptosis whereas GSDMD overexpression aggravates cardiomyocyte apoptosis. Taken together, these results demonstrate that GSDMD-mediated cardiomyocyte apoptosis plays an important role in DIC.
Cardiomyocyte-specific GSDMD Deficiency Alleviates Acute and Chronic DIC
Figure 1 suggested that GSDMD played a vital role in regulating the DOX-associated myocardial injury. Numerous lines of evidence indicate that GSDMD induces cell death [1, 3]. However, whether the same is true in the cardiac context remains unknown. Accordingly, to investigate this, we established mice with cardiomyocyte-specific GSDMD KO (GSDMD-CKO). GSDMD-CKO totally abolished the expression of GSDMD protein (Figure 2M). Cardiac GSDMD deficiency decreased the impairment of DOX-induced myocardial injury. After DOX intervention, compared with the control group, the survival rate of GSDMD-CKO mice was significantly prolonged, BW and heart weight loss were decreased, and the levels of myocardial injury markers CK-MB and cTnT were remarkably decreased (Figure 2A–2C, and Figure 2E). The damage of cardiac function in DOX-treated GSDMD-CKO mice was reduced, with a greater increase in ejection fraction, fractional shortening, and contractile indexes than those seen in control mice (Figure 2D, Figure 2F–2K, Figure S5A). In addition, following DOX treatment, cardiomyocytes in GSDMD-CKO mice were bigger than those in GSDMD(flox/flox) mice (Figure 2L). ROS staining based on DHE fluorescence revealed that ROS levels in DOX-treated GSDMD-CKO mice were lower than those in GSDMD(flox/flox) littermates (Figure 2L). Cardiomyocyte apoptosis after DOX administration in GSDMD-CKO mice was reduced compared to that in GSDMD(flox/flox) mice, as indicated by decreased CC3 and BAX protein levels (Figure 2M and 2O) and the number of TUNEL-positive cells (Figure 2N and 2P).
We next repeated the above experiments in a 6-week model of chronic DOX cardiotoxicity. Consistently with the observations in the acute model, DOX-treated GSDMD-CKO mice had an improved survival rate, and with higher heart weight and BW at the end of the observation period than those in control mice (Figure S5B–S5D). Echocardiography at weeks 2 and 4 did not reveal any significant differences between DOX-treated GSDMD(flox/flox) and GSDMD-CKO mice, however, cardiac function notably improved by week 6 in CKO mice compared with GSDMD(flox/flox) DOX-treated controls (Figure S5E and S5F). Western blotting revealed a remarkable reduction of the levels of apoptosis-related proteins in CKO heart after DOX treatment (Figure S5G and S5H). Collectively, these observations indicated that GSDMD KO in the heart is protective against DIC in both, acute and chronic models of DOX cardiomyopathy.
GSDMD Deficiency Causes Apoptosis in Cardiomyocytes and Exacerbates DIC
We next repeated the experiments investigating DIC in vitro, using cultured adult cardiomyocytes. Cardiomyocytes were treated with 1 mM DOX and analyzed over time. Western blotting revealed that GSDMD protein levels, either full-length or its N-terminus, peaked 6 h after DOX treatment and began to decline 12 h after the treatment (Figure 3A). The same changes were observed for apoptosis-related proteins (Figure 3B). We therefore selected 12 h as the time point for the ensuing detailed analysis. Similar effects of GSDMD deletion on DOX-treated damage of cardiomyocytes were accordantly observed as in vivo. GSDMD KO in cardiomyocytes reduced DOX-induced increase of GSDMD N-terminus, CC3, and BAX protein levels (Figure 3C and 3D), and the number of dead cardiomyocytes, as assessed by propidium iodide staining (Figure 3E and 3F). All these effects were reversed by adenovirus-mediated GSDMD overexpression (Figure 3G–3J). Collectively, these observations confirmed that GSDMD causes apoptosis in cardiomyocytes, thus exacerbating DIC.
GSDMD Promotes DOX-induced Myocardial Autophagy
The contribution of autophagy to DOX-induced cardiac injury is well-documented [15]. To evaluate the effect of DIC on autophagy in GSDMD inhibition hearts, we first performed time-point analysis of hearts 1, 3, 5, and 7 d after DOX injection. Western blotting revealed the same trends in the levels of GSDMD and microtubule-associated protein-1 light chain 3-II (LC3-II), a marker of autophagy. Specifically, protein levels began to increase 1 d after DOX intervention, peaked after 3 d, and began to decline after 5 d (Figure 4A and 4B). We then determined the expression of several autophagy-associated genes, including ATGs, Beclin-1, LC3B, and Sqstm 1/p62 genes. The expression of most genes showed a similar trend to that shown on the protein level (Figure 4C), reflecting the response to DOX. Therefore, we selected day 3 as the time of cardiac intervention in mice for subsequent exploration of autophagy.
We quantified the levels of LC3-II protein in mouse heart. While the levels were increased by DOX administration, the increase was reduced in the GSDMD-CKO background, and partially rescued by AAV9-GSDMD-OE infection (Figure 4D and 4E). LC3-II level increase could be associated with the initiation of autophagy or prevention of LC3-II degradation. To distinguish between these two possibilities, we used bafilomycin A1 (BAFA1), a lysosomal inhibitor of late-stage autophagy that blocks the degradation of LC3-II [26]. We observed that LC3-II levels were obviously increased in DOX-treated heart and further increased upon BAFA1 treatment (Figure 4F). However, the increase was much lower in DOX-treated GSDMD-CKO heart than that in the GSDMD(flox/flox) DOX-treated heart (Figure 4F). Further, the reduced LC3-II levels in GSDMD-deficient heart were rescued by GSDMD overexpression (Figure 4G). These observations suggest that LC3-II accumulation in GSDMD-overexpressing heart and DOX-treated heart is caused by its blocked degradation.
We next used transmission electron microscopy (TEM) to investigate the autophagosomes and autolysosomes in the heart of DOX-treated mice. GSDMD-CKO mice were relatively resistant to DOX injection, with a reduced number of autophagosomes and autolysosomes (Figure 4H). However, a marked increase of autophagosomes and autolysosomes was observed in AAV9-GSDMD-OE DOX-treated mice, compared with control mice (Figure 4I). This indicated that GSDMD promotes DOX-induced myocardial autophagy.
DOX Activates Cardiomyocyte Autophagic Flux and GSDMD Aggravates Myocardial Autophagy Levels in Adult Cardiomyocytes
In vivo studies verified the relationship between GSDMD and autophagy. We next explored whether this effect reproduced in cultured cardiomyocytes. Consistent with the cell experiments described in Figure 3A, the levels of LC3-II protein in DOX-treated cardiomyocytes first increased and then fell, reaching a peak 6–12 h after DOX intervention (Figure 5A and 5B). We therefore chose 12 h as the time point for cardiomyocyte autophagy studies. DOX markedly induced the accumulation of autophagy-associated proteins in the WT group, but the levels were nore reduced in cardiomyocytes isolated from adult GSDMD-KO mice (Figure 5C and 5D). By contrast, LC3-II accumulated in GSDMD-overexpressing cardiomyocytes (Figure 5C and 5E). We concluded that DOX promotes autophagy induction, contributing to a high level of LC3-II expression, with GSDMD enhancing this effect.
To identify the specific stages of autophagic flux induced by DOX, we transfected cardiomyocytes with a tandem double-labeled probe, monomeric red fluorescent protein–green fluorescent protein–LC3 (mRFP-GFP-LC3). The cells were then incubated with DOX for 12 h. Since GFP protein is sensitive to low pH, only red fluorescence is detected upon GFP fluorescence after autophagosome and lysosome fusion, as mRFP fluorescence is stable [26]. This allows the assessment of autophagic flux, distinguishing autophagosomes (yellow) from autolysosomes (red). Accordingly, red puncta are more numerous than yellow puncta upon autophagy induction, with the opposite upon autophagy inhibition.
We observed a remarkable increase in the number of autolysosomes and autophagosomes in DOX-treated WT cardiomyocytes, with the former more significant than the latter (Figure 5F). GSDMD deficiency reduced the autolysosome to autophagosome ratio compared with that in the DOX-treated WT cardiomyocyte group (Figure 5F and 5H), whereas GSDMD overexpression had the opposite effect (Figure 5G and 5H). This suggested that downregulation of GSDMD reduces autophagy. These observations, together with immunoblot analysis (Figure 5A–5E), indicated that DOX treatment promotes autophagy, and that GSDMD overexpression enhances this effect.
To verify the above conclusions, we used BAFA1 to prevent the fusion of autophagosomes with lysosomes. The enhanced expression of LC3-II protein upon DOX treatment further increased in the presence of BAFA1 (Figure 5I–5K). By contrast, as anticipated, LC3-II levels were decreased in GSDMD-KO cardiomyocytes (Figure 5I and 5K), and were significantly increased in GSDMD-overexpressing cells (Figure 5J and 5K). We then repeated the autophagic flux analysis using the mRFP-GFP-LC3 reporter system introduced by AAV infection. BAFA1 drastically reduced the number of autolysosomes in WT, GSDMD-KO-CM, and GSDMD-OE-CM groups compared with the controls, with and without DOX treatment, but increased the number of autophagosomes (Figure 5L–5N). In other words, the autolysosome to autophagosome ratio significantly decreased upon BAFA1 treatment, suggesting that the autophagic flux was inhibited, especially in GSDMD-KO cells. Together with western blot data (Figure 5I–5K), these observations confirmed that BAFA1 induces the aggregation of LC3-II by inhibiting LC3-II degradation at the late stage of autophagy, and that downregulation of GSDMD expression weakens this effect.
GSDMD Deletion Ameliorates DIC by Inhibiting Autophagy
The role of autophagy reduction by GSDMD inhibition was further investigated by using autophagy inhibitor 3-methyladenine (3-MA) and autophagy agonist rapamycin (Rapa) in vivo. 3-MA inhibits the formation of autophagosomes and Rapa is an inhibitor of mechanistic target of rapamycin (mTOR) [27]. As anticipated, intraperitoneal injection of 3-MA and Rapa decreased BW in DOX-treated groups, but the weight loss was more pronounced in Rapa-treated groups and declined to a lesser extent in DOX-treated CKO mice than in other mice, while the heart weight to tibia length (HW/TL) ratio showed the opposite trend (Figure S7A). Cardiac function improved after 3-MA treatment, with an increased ejection fraction and fractional shortening, while Rapa treatment exacerbated cardiac function (Figure S7B, S7E, and S7F). Echocardiography readings for the DOX-treated CKO group were significantly better than those for the control group (Figure S7C). Furthermore, 3-MA markedly decreased the LC3-II levels, regardless of DOX treatment (Figure S7D). Meanwhile, Rapa induced accumulation of LC3-II and apoptosis-related proteins, and fully abolished 3-MA–induced decrease of these protein levels upon DOX administration, these protein levels were slightly decreased in CKO mice (Figure S7D, S7G–S7I). Hence, the downregulation of GSDMD levels in the heart during DIC ameliorates the impairment of autophagy.
RNA-sequencing (RNA-seq) Reveals Potential Target Genes of GSDMD
The above experiments suggest that GSDMD deficiency can play a protective role in the myocardium by inhibiting cardiac autophagy. However, how GSDMD mediates cardiac autophagy remains unclear. To further investigate it, we explored the GSDMD-mediated regulatory genes by using RNA-seq. The analysis revealed a significant change of the expression of ER-related genes between DOX-treated GSDMD CKO mice and control mice (Figure 6A and 6B). ERS can lead to the accumulation of unfolded or misfolded proteins in the ER lumen, and induce autophagy in different ways. Western blot analysis indicated that ERS-related proteins were significantly downregulated after GSDMD inhibition (Figure 6C and 6D), further suggesting that GSDMD may induce cardiac autophagy by regulating ERS. Among these ER injury-related genes, FAM134B might be a special one. FAM134B is a common receptor in ER autophagy, and mediates the recognition and removal of ER by autophagosomes [28, 29]. Downregulation of FAM134B leads to ER-phagy, causing ER expansion, ERS, and cell death [28]28. Real-time PCR and western blotting indicated upregulation of ER-regulatory protein Retreg1 (FAM134B) after DOX intervention and its downregulation during GSDMD deficiency (Figure 6C–6E). We observed that ER marker FAM134B co-located with GSDMD, and presented as positive spots upon immunofluorescence staining (Figure 6F). This revealed that GSDMD and FAM134B were expressed in the cardiomyocyte ER, with FAM134B downregulation upon GSDMD deletion (Figure 6F). GSDMD and LC3 (Figure 6G), FAM134B and Bip (Figure 6H), FAM134B and LC3 (Figure 6I) also have co-location in cardiomyocyte. Taken together, these results suggested that GSDMD may cause cardiac autophagy and cardiomyocyte apoptosis by regulating ER-phagy receptor FAM134B.
GSDMD Promotes Autophagy by Promoting ERS to Activate FAM134B, Thus Aggravating Cardiomyocyte Apoptosis
To determine whether GSDMD plays a role in regulating ERS, we used ERS inhibitor 4-phenylbutyric acid (4PBA) to inhibit ERS and observed changes in cardiac autophagy and cardiomyocyte apoptosis . 4PBA inhibited ERS and autophagy in GSDMD(flox/flox) mice, and neutralized the effects of GSDMD deficiency on ERS and autophagy (Figure 7D and 7E). Further, 4PBA reduced GSDMD(flox/flox) mouse mortality (Figure 7A), improved cardiac function (Figure 7B and 7C), and decreased cardiomyocyte apoptosis (Figure 7D and 7E). In vitro data were consistent with the in vivo observation that 4PBA reduces ERS and apoptosis (Figure 7F–7H). These findings confirmed that GSDMD loss inhibits cardiac autophagy, reduces myocardial apoptosis, improves cardiac function, and alleviates DIC by mediating ERS in cardiomyocytes.
Furthermore, the expression of FAM134B in GSDMD(flox/flox) mice was remarkably downregulated after 4PBA administration, further downregulated in the CKO group (Figure 7D), demonstrating that FAM134B is the key downstream target gene of GSDMD regulating ERS. In summary, the presented results indicate that GSDMD-N activates ERS by promoting ER perforation, stimulating FAM134B to fragment ER, thus promoting autophagy, aggravating cardiomyocyte apoptosis, and ultimately aggravating DIC (Figure 7I).