Archives

  • 2018-07
  • 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
  • 2023-08
  • 2023-09
  • 2023-10
  • 2023-11
  • 2023-12
  • 2024-01
  • 2024-02
  • 2024-03
  • 2024-04
  • 2024-05
  • Work is presently underway to unveil mechanisms whereby SAHA

    2021-10-21

    Work is presently underway to unveil mechanisms whereby SAHA-dependent restoration of cardiomyocyte autophagic flux is protective. Another interesting question pertains to mechanisms whereby class I and class II HDAC inhibitors induce autophagy. It has been demonstrated that TSA reduces transverse aortic banding-induced cardiac hypertrophy [37]. Recent data demonstrate that inhibition of class I HDACs with apicidin induces the Boc-D-Asp(OtBu)-OH.DCHA of tuberous sclerosis complex 2 (TSC2), an mTOR inhibitor, which inhibits mTOR-mediated cardiac hypertrophy [38]. Since mTOR is a strong modulator of autophagy [39], inhibition of class I HDACs may induce autophagy at least partially through this pathway. Interestingly, β-hydroxybutyrate (β-OHB) at physiological levels functions as an endogenous HDAC inhibitor [40] and induces autophagy in neurons [41]. In the kidney, β-OHB promotes acetylation of the promoters of the genes coding for FoxO3a and MT2 transcription factors, which upregulates their expression to activate the downstream targets SOD2 and catalase [40]. These, in turn, elicit reductions in ROS and protect the kidney from I/R injury [40]. Delivery of exogenous β-OHB or increasing β-OHB by fasting immediately prior to I/R, reduces cardiac infarct size in rats [42,43]. These findings suggest cardioprotective effects of β-OHB, possibly via activation of autophagy, and point to possible links among metabolism, HDAC biology, and cardiomyocyte I/R tolerance. There are few data on the effects of genetic manipulation of HDACs during cardiac I/R. First, we do not know which HDACs are the functionally relevant targets of HDAC inhibition. Second, HDACs are vital for multiple other cellular functions; most constitutive knockouts and even tissue-specific knockouts manifest baseline phenotypes, including cardiac hypertrophy [44]. Last but not least, it is challenging to mimic the small molecule-dependent, reversible suppression of HDACs with a genetic model. Experimental up-regulation of HDAC4 activity has provided some rationale for genetic manipulation of HDACs in cardiac I/R injury [24], but our understanding of HDAC function during cardiac I/R remains incomplete.
    FDA-approved, pharmaceutical grade, small molecule HDAC inhibitors that could be used in clinical trials As mentioned above, it is challenging to design a clinical trial to evaluate a strategy of myocardial protection during reperfusion injury; indeed, specific requirements have been laid out [5] [9]. SAHA has satisfied most of them: tested at the time of reperfusion; confirmed in large-animal models; safe and available pharmaceutical grade agent; efficacy verified in multiple laboratories; robustness of response; preclinical studies conducted in a randomized, blinded fashion [26]. Two caveats that remain are that SAHA has never been tested in animals with comorbidities, and long-term effects beyond infarct size have not been Boc-D-Asp(OtBu)-OH.DCHA evaluated. However, recent data in diabetic rats demonstrating that pretreatment with TSA significantly reduces infarct size offer hope that SAHA may have efficacy in a myocardial infarct population with diabetes [21]. Furthermore, it has been shown that long-term exposure to low-dose TSA or SAHA reduces post-infarct adverse remodeling, suggesting that SAHA's cardioprotective effects may be enduring and translate into improved clinical outcomes [28,31]. There are several reported side effects of HDAC inhibitors in cancer treatment, including gastrointestinal upset, fatigue, thrombocytopenia, anemia and bone marrow toxicity, and reversible cardiac arrhythmia (FK-228, cyclic depsipeptide) [27]. It is worth noting, however, that SAHA manifests no signs of cardiotoxicity [27]. With respect to cardiac I/R, it seems unlikely that a single dose administered at the time of cardiac reperfusion during MI will elicit untoward effects outside the heart. Furthermore, as there are multiple FDA-approved HDAC inhibitors available or in the pipeline for cancer therapy [18], we may be able to refine our therapeutic approaches for optimal efficacy and minimal off-target effects.