scholarly journals Predicting Response to Histone Deacetylase Inhibitors Using High-Throughput Genomics

2015 ◽  
Vol 107 (11) ◽  
pp. djv247 ◽  
Author(s):  
Paul Geeleher ◽  
Andrey Loboda ◽  
Divya Lenkala ◽  
Fan Wang ◽  
Bonnie LaCroix ◽  
...  
2017 ◽  
Vol 19 (suppl_4) ◽  
pp. iv57-iv58
Author(s):  
Viktoria Marquardt ◽  
David Pauck ◽  
Finn K. Hansen ◽  
Daniel Picard ◽  
Jörg Felsberg ◽  
...  

BMC Genomics ◽  
2014 ◽  
Vol 15 (1) ◽  
pp. 528 ◽  
Author(s):  
Ann-Christin Gaupel ◽  
Thomas Begley ◽  
Martin Tenniswood

2011 ◽  
Vol 16 (10) ◽  
pp. 1206-1216 ◽  
Author(s):  
Patricia Haus ◽  
Michael Korbus ◽  
Michael Schröder ◽  
Franz-Josef Meyer-Almes

Histone deacetylases (HDACs) are important epigenetic factors regulating a variety of vital cellular functions such as cell cycle progression, differentiation, cell migration, and apoptosis. Consequently, HDACs have emerged as promising targets for cancer therapy. The drugability of HDACs has been shown by the discovery of several structural classes of inhibitors (HDACis), particularly by the recent approval of two HDACis, vorinostat (ZOLINZA) and romidepsin (Istodax), for the treatment of cutaneous T-cell lymphoma by the US Food and Drug Administration. The outstanding potential of HDACis, with a defined isoform selectivity profile as drugs against a plurality of diseases, vindicates increased effort in developing high-throughput capable assays for screening campaigns. In this study, a dual-competition assay exploiting changes in fluorescence anisotropy and lifetime was used to screen the LOPAC (Sigma-Aldrich, St Louis, MO) library against the bacterial histone deacetylase homologue HDAH from Bordetella, which shares 35% identity with the second deacetylase domain of HDAC6. The binding assay proved to be highly suitable for high-throughput screening campaigns. Several LOPAC compounds have been identified to inhibit HDAH in the lower micromolar range. Most interestingly, some of the hit compounds turned out to be weak but selective inhibitors of human class IIa and IIb HDACs.


2020 ◽  
Vol 11 (1) ◽  
Author(s):  
Francesca Cavallo ◽  
Flavia Troglio ◽  
Giovanni Fagà ◽  
Daniele Fancelli ◽  
Reinald Shyti ◽  
...  

Abstract Background Autism spectrum disorder (ASD) is a highly prevalent neurodevelopmental condition affecting almost 1% of children, and represents a major unmet medical need with no effective drug treatment available. Duplication at 7q11.23 (7Dup), encompassing 26–28 genes, is one of the best characterized ASD-causing copy number variations and offers unique translational opportunities, because the hemideletion of the same interval causes Williams–Beuren syndrome (WBS), a condition defined by hypersociability and language strengths, thereby providing a unique reference to validate treatments for the ASD symptoms. In the above-indicated interval at 7q11.23, defined as WBS critical region, several genes, such as GTF2I, BAZ1B, CLIP2 and EIF4H, emerged as critical for their role in the pathogenesis of WBS and 7Dup both from mouse models and human studies. Methods We performed a high-throughput screening of 1478 compounds, including central nervous system agents, epigenetic modulators and experimental substances, on patient-derived cortical glutamatergic neurons differentiated from our cohort of induced pluripotent stem cell lines (iPSCs), monitoring the transcriptional modulation of WBS interval genes, with a special focus on GTF2I, in light of its overriding pathogenic role. The hits identified were validated by measuring gene expression by qRT-PCR and the results were confirmed by western blotting. Results We identified and selected three histone deacetylase inhibitors (HDACi) that decreased the abnormal expression level of GTF2I in 7Dup cortical glutamatergic neurons differentiated from four genetically different iPSC lines. We confirmed this effect also at the protein level. Limitations In this study, we did not address the molecular mechanisms whereby HDAC inhibitors act on GTF2I. The lead compounds identified will now need to be advanced to further testing in additional models, including patient-derived brain organoids and mouse models recapitulating the gene imbalances of the 7q11.23 microduplication, in order to validate their efficacy in rescuing phenotypes across multiple functional layers within a translational pipeline towards clinical use. Conclusions These results represent a unique opportunity for the development of a specific class of compounds for treating 7Dup and other forms of intellectual disability and autism.


Author(s):  
Shanshan Zhang ◽  
Zhaojian Gong ◽  
Peter O. Oladimeji ◽  
Duane G. Currier ◽  
Qipan Deng ◽  
...  

Abstract Background Medulloblastoma is the most frequently occurring malignant brain tumor in children. Current treatment strategies for medulloblastoma include aggressive surgery, cranio-spinal irradiation and adjuvant chemotherapy. Because current treatments can cause severe long-term side effects and are not curative, successful treatment remains a challenge. Methods In this study, we employed a high-throughput cell viability assay to screen 12,800 compounds and to identify drug candidates with anti-proliferative properties for medulloblastoma cells. We also tested these compounds for attenuating medulloblastoma tumor development using mouse xenografts. Results We identified two histone deacetylase inhibitors (dacinostat and quisinostat) with anti-proliferative properties for medulloblastoma cells. We showed that both compounds induce cytotoxicity, trigger cell apoptosis, and block cell cycle progression at the G2/M phase. In addition, dacinostat and quisinostat attenuated xenograft medulloblastoma growth in mice. Conclusions Our findings suggest that histone deacetylase inhibitors are potent therapeutic agents against medulloblastoma.


Sign in / Sign up

Export Citation Format

Share Document