Archives

  • 2018-07
  • 2018-10
  • 2018-11
  • 2019-04
  • 2019-05
  • 2019-06
  • 2019-07
  • 2019-08
  • 2019-09
  • 2019-10
  • 2019-11
  • 2019-12
  • 2020-01
  • 2020-02
  • 2020-03
  • 2020-04
  • 2020-05
  • 2020-06
  • 2020-07
  • 2020-08
  • 2020-09
  • 2020-10
  • 2020-11
  • 2020-12
  • 2021-01
  • 2021-02
  • 2021-03
  • 2021-04
  • 2021-05
  • 2021-06
  • 2021-07
  • 2021-08
  • 2021-09
  • 2021-10
  • 2021-11
  • 2021-12
  • 2022-01
  • 2022-02
  • 2022-03
  • 2022-04
  • 2022-05
  • 2022-06
  • 2022-07
  • 2022-08
  • 2022-09
  • 2022-10
  • 2022-11
  • 2022-12
  • 2023-01
  • 2023-02
  • 2023-03
  • 2023-04
  • 2023-05
  • 2023-06
  • 2023-07
  • 2023-08
  • 2023-09
  • 2023-10
  • 2023-11
  • 2023-12
  • 2024-01
  • 2024-02
  • 2024-03
  • 2024-04
  • 2024-05
  • br Mechanisms of HDAC inhibition

    2022-08-22


    Mechanisms of HDAC inhibition-dependent cardioprotection Multiple preclinical studies have demonstrated potent cardioprotective benefits of HDAC inhibition in murine models of myocardial stress, including I/R [19,25,29,30]. TSA reduces myocardial infarct size by up to 50% [19,25]. In addition, treatment with the structurally distinct HDAC inhibitor Scriptaid (another class I and class II HDAC inhibitor) resulted in nearly identical protection as TSA when compared with Nullscript (negative control), which strongly suggests a class effect [19]. Given that HDAC inhibitors are so effective in targeting reperfusion injury, they provide opportunities to delineate mechanisms of reperfusion injury. Initially, TSA was thought to activate the p38 pathway during I/R to protect myocardial tissue [25]. Further investigation demonstrated that HDAC inhibitors prevent ischemia-induced activation of gene programs in vivo and in vitro that involve hypoxia-inducible factor-1α, cell death, and vascular permeability, thereby reducing vascular leak and myocardial injury [25]. Furthermore, long-term use of TSA promotes myocardial repair and blunts adverse cardiac remodeling by stimulating endogenous cardiac regeneration and neovascularization, which seems to be dependent on c-kit signaling [31]. However, with recent data showing that c-kit+ Toremifene have minimal contribution to cardiomyocytes but mainly contribute to endothelial cells in the heart [32], these data should be interpreted with caution. We have reported that SAHA increases cardiomyocyte autophagic activity within the infarct border zone as measured by LC3-II levels and formation of GFP-LC3 puncta, findings that were verified by electron microscopy [26]. Furthermore, SAHA increased autophagic flux in the infarct border zone assayed using a tandem fluorescence reporter RFP-GFP-LC3 transgenic mouse. In cultured cardiomyocytes subjected to simulated I/R, SAHA pretreatment reduced cell death by 40%. This reduction in cell death correlated with increased autophagic flux in SAHA-treated cells. RNAi-mediated knockdown of ATG7 and ATG5, essential autophagy proteins, abolished SAHA's cardioprotective effects. Based on these findings, we concluded that the cardioprotective effects of SAHA during I/R occur, at least in part, through induction and maintenance of cardiomyocyte autophagic flux [26]. As noted earlier, SAHA plasma concentrations in the rabbits were similar to those achieved in cancer patients [26]. In aggregate, these data support a model in which HDACs participate in the suppression of autophagic flux during myocardial reperfusion injury, and SAHA-dependent suppression of HDAC activity restores autophagic flux, thereby limiting reperfusion injury. In light of this, we submit that SAHA may emerge Toremifene as an effective therapeutic agent in reperfusion injury, a significant clinical problem that lacks meaningfully, efficacious therapy [4,26,33]. TSA and SAHA are selective inhibitors of class I and II HDAC inhibitors, but non-selective among enzymes within those classes [34]. Other work has shown that the class I-specific HDAC inhibitor entinostat (MS-275) significantly reduced infarct size in an isolated rat heart I/R model [35,36]. It is reported that entinostat increased expression of SOD2 and catalase acting through the transcription factor FoxO3a [35]. Interestingly, selective inhibition of class I HDACs afforded superior cardioprotection when compared with pan-HDAC inhibition in this pretreatment model [35]. The same group tested entinostat delivered at the time of reperfusion. They observed that HDAC1 is present in mitochondria of cardiac myocytes but not those of fibroblasts or endothelial cells [36]. The investigators engineered mitochondria-restricted and mitochondria-excluded HDAC inhibitors and tested both in an ex vivo I/R model. Interestingly, selective inhibition of mitochondrial HDAC1 attenuated I/R injury to the same extent as entinostat, whereas the mitochondria-excluded inhibitor did not. These effects were attributed to a decrease in succinate dehydrogenase (SDHA) activity and subsequent metabolic ROS production in reperfusion [36].