1
|
Valdez BC, Tsimberidou AM, Yuan B, Nieto Y, Baysal MA, Chakraborty A, Andersen CR, Andersson BS. Synergistic cytotoxicity of histone deacetylase and poly-ADP ribose polymerase inhibitors and decitabine in pancreatic cancer cells: Implications for novel therapy. Oncotarget 2024; 15:361-373. [PMID: 38829622 PMCID: PMC11146633 DOI: 10.18632/oncotarget.28588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 05/17/2024] [Indexed: 06/05/2024] Open
Abstract
Histone deacetylase inhibitors (HDACi) can modulate the acetylation status of proteins, influencing the genomic instability exhibited by cancer cells. Poly (ADP ribose) polymerase (PARP) inhibitors (PARPi) have a direct effect on protein poly (ADP-ribosyl)ation, which is important for DNA repair. Decitabine is a nucleoside cytidine analogue, which when phosphorylated gets incorporated into the growing DNA strand, inhibiting methylation and inducing DNA damage by inactivating and trapping DNA methyltransferase on the DNA, thereby activating transcriptionally silenced DNA loci. We explored various combinations of HDACi and PARPi +/- decitabine (hypomethylating agent) in pancreatic cancer cell lines BxPC-3 and PL45 (wild-type BRCA1 and BRCA2) and Capan-1 (mutated BRCA2). The combination of HDACi (panobinostat or vorinostat) with PARPi (talazoparib or olaparib) resulted in synergistic cytotoxicity in all cell lines tested. The addition of decitabine further increased the synergistic cytotoxicity noted with HDACi and PARPi, triggering apoptosis (evidenced by increased cleavage of caspase 3 and PARP1). The 3-drug combination treatments (vorinostat, talazoparib, and decitabine; vorinostat, olaparib, and decitabine; panobinostat, talazoparib, and decitabine; panobinostat, olaparib, and decitabine) induced more DNA damage (increased phosphorylation of histone 2AX) than the individual drugs and impaired the DNA repair pathways (decreased levels of ATM, BRCA1, and ATRX proteins). The 3-drug combinations also altered the epigenetic regulation of gene expression (NuRD complex subunits, reduced levels). This is the first study to demonstrate synergistic interactions between the aforementioned agents in pancreatic cancer cell lines and provides preclinical data to design individualized therapeutic approaches with the potential to improve pancreatic cancer treatment outcomes.
Collapse
Affiliation(s)
- Benigno C. Valdez
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Apostolia M. Tsimberidou
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Bin Yuan
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yago Nieto
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mehmet A. Baysal
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Abhijit Chakraborty
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Clark R. Andersen
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Borje S. Andersson
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
2
|
Shanmugam G, Subramaniyam K, George M, Sarkar K. HDAC inhibition regulates oxidative stress in CD4 +Thelper cells of chronic obstructive pulmonary disease and non-small cell lung cancer patients via mitochondrial transcription factor a (mtTFA) modulating NF-κB/HIF1α axis. Int Immunopharmacol 2023; 122:110661. [PMID: 37473712 DOI: 10.1016/j.intimp.2023.110661] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 07/14/2023] [Accepted: 07/14/2023] [Indexed: 07/22/2023]
Abstract
Histone deacetylases (HDACs) play a crucial role in the epigenetic regulation of gene expression by remodelling chromatin. Isoenzymes of the HDAC family exhibit aberrant regulation in a wide variety of cancers as well as several inflammatory lung disorders like chronic obstructive pulmonary disease (COPD). Inhibition of HDACs is a potential therapeutic strategy that could be used to reverse epigenetic modification. Trichostatin A (TSA), a powerful histone deacetylase (HDAC) inhibitor, has anti-cancer effects in numerous cancer types. However, it is not yet apparent how HDAC inhibitors affect human non-small cell lung cancer cells (NSCLC) and COPD. This study aims to investigate TSA's role in restoring mitochondrial dysfunction and its effect on hypoxia and inflammation in CD4+T cells obtained from patients with COPD and lung cancer. As a result of treatment with TSA, there is a reduction in the expression of inflammatory cytokines and a decreased enrichment of transcriptional factors associated with inflammation at VEGFA gene loci. We have seen a substantial decrease in the expression of NF-κB and HIF1α, which are the critical mediators of inflammation and hypoxia, respectively. Following TSA treatment, mtTFA expression was increased, facilitating patients with COPD and NSCLC in the recovery of their dysfunctional mitochondria. Furthermore, we have discovered that TSA treatment in patients with COPD and NSCLC may lead to immunoprotective ness by inducing Th1ness. Our finding gives a new insight into the existing body of knowledge regarding TSA-based therapeutic methods and highlights the necessity of epigenetic therapy for these devastating lung disorders.
Collapse
Affiliation(s)
- Geetha Shanmugam
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Krishnaveni Subramaniyam
- Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Melvin George
- Department of Clinical Pharmacology, SRM Medical College Hospital and Research Centre, Kattankulathur, Tamil Nadu 603203, India
| | - Koustav Sarkar
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India.
| |
Collapse
|
3
|
Wei Z, Li Y, Ali F, Wang Y, Liu J, Yang Z, Wang Z, Xing Y, Li F. Transcriptomic analysis reveals the key role of histone deacetylation via mediating different phytohormone signalings in fiber initiation of cotton. Cell Biosci 2022; 12:107. [PMID: 35831870 PMCID: PMC9277824 DOI: 10.1186/s13578-022-00840-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 06/28/2022] [Indexed: 12/04/2022] Open
Abstract
Background Histone deacetylation is one of the most important epigenetic modifications and plays diverse roles in plant development. However, the detailed functions and mechanisms of histone deacetylation in fiber development of cotton are still unclear. HDAC inhibitors (HDACi) have been commonly used to study the molecular mechanism underlying histone deacetylation or to facilitate disease therapy in humans through hindering the histone deacetylase catalytic activity. Trichostatin A (TSA)—the most widely used HDACi has been extensively employed to determine the role of histone deacetylation on different developmental stages of plants. Results Through in vitro culture of ovules, we observed that exogenous application of TSA was able to inhibit the fiber initiation development. Subsequently, we performed a transcriptomic analysis to reveal the underlying mechanisms. The data showed that TSA treatment resulted in 4209 differentially expressed genes, which were mostly enriched in plant hormone signal transduction, phenylpropanoid biosynthesis, photosynthesis, and carbon metabolism pathways. The phytohormone signal transduction pathways harbor the most differentially expressed genes. Deeper studies showed that some genes promoting auxin, Gibberellic Acid (GA) signaling were down-regulated, while some genes facilitating Abscisic Acid (ABA) and inhibiting Jasmonic Acid (JA) signaling were up-regulated after the TSA treatments. Further analysis of plant hormone contents proved that TSA significantly promoted the accumulation of ABA, JA and GA3. Conclusions Collectively, histone deacetylation can regulate some key genes involved in different phytohormone pathways, and consequently promoting the auxin, GA, and JA signaling, whereas repressing the ABA synthesis and signaling to improve the fiber cell initiation. Moreover, the genes associated with energy metabolism, phenylpropanoid, and glutathione metabolism were also regulated by histone deacetylation. The above results provided novel clues to illuminate the underlying mechanisms of epigenetic modifications as well as related different phytohormones in fiber cell differentiation, which is also very valuable for the molecular breeding of higher quality cotton. Supplementary Information The online version contains supplementary material available at 10.1186/s13578-022-00840-4.
Collapse
|
4
|
Involvement of inflammatory cytokines and epigenetic modification of the mtTFA complex in T-helper cells of patients' suffering from non-small cell lung cancer and chronic obstructive pulmonary disease. Mol Immunol 2022; 151:70-83. [PMID: 36099831 DOI: 10.1016/j.molimm.2022.08.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/01/2022] [Accepted: 08/14/2022] [Indexed: 11/22/2022]
Abstract
Dysregulated inflammatory response plays a crucial role in the pathogenesis of chronic obstructive pulmonary disease (COPD) and Non-Small cell lung cancer (NSCLC). Hence, the purpose of this research is to uncover the link between alterations in inflammatory cytokine levels and disease progression in CD4+T cells of patients suffering from COPD and lung cancer. We also investigated the epigenetic regulation of mtTFA to delineate the role of oxidative stress-mediated inflammation in Lung cancer and COPD. The RT2 Profiler PCR array was used to examine the differential expression pattern of inflammatory genes in CD4+ T helper (Th) cells from COPD, NSCLC, and control subjects. Candidate inflammatory gene loci were selected and the enrichment of transcriptional factor and histone modifiers was analysed using ChIP-qPCR. In comparison to control subjects, a set of genes (e.g., BMP2, CCL2, IL5, VEGFA, etc.) are over-expressed whereas another set of genes (e.g., AIMP1, IFNG, LTA, LTB, TNF, etc.) are under-expressed in both COPD and NSCLC patients. The increased percent enrichment of inflammation-associated transcription factors including NF-kB, CREB, HIF1, and MYC at the loci of inflammatory genes was revealed by our chromatin immunoprecipitation (ChIP) data. H3K4me3, H3K9me3, H3K14Ac, HDAC1, 2, 3, 6 all showed dysregulated enrichment at the VEGFA gene locus. One of the epigenetic modifications, histone methylation, was found to be abnormal in the mtTFA complex in COPD and NSCLC patients in comparison to controls. Although there is mounting evidence of several links between these disorders, therapeutic options remain inadequate. Our findings contribute to the body of knowledge about therapeutic techniques that use inflammatory cytokines as a prognostic marker and highlight the need for epigenetic therapy for these debilitating lung diseases.
Collapse
|
5
|
Shanmugam G, Rakshit S, Sarkar K. HDAC inhibitors: Targets for tumor therapy, immune modulation and lung diseases. Transl Oncol 2022; 16:101312. [PMID: 34922087 PMCID: PMC8688863 DOI: 10.1016/j.tranon.2021.101312] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 12/06/2021] [Indexed: 12/13/2022] Open
Abstract
Histone deacetylases (HDACs) are enzymes that play a key role in the epigenetic regulation of gene expression by remodeling chromatin. Inhibition of HDACs is a prospective therapeutic approach for reversing epigenetic alteration in several diseases. In preclinical research, numerous types of HDAC inhibitors were discovered to exhibit powerful and selective anticancer properties. However, such research has revealed that the effects of HDAC inhibitors may be far broader and more intricate than previously thought. This review will provide insight into the HDAC inhibitors and their mechanism of action with special emphasis on the significance of HDAC inhibitors in the treatment of Chronic Obstructive Pulmonary Disease and lung cancer. Nanocarrier-mediated HDAC inhibitor delivery and new approaches for targeting HDACs are also discussed.
Collapse
Affiliation(s)
- Geetha Shanmugam
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Sudeshna Rakshit
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Koustav Sarkar
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India.
| |
Collapse
|
6
|
Yang Y, Hu B, Yang Y, Gong K, Wang H, Guo Q, Tang X, Li Y, Wang J. Rational design of selective HDAC2 inhibitors for liver cancer treatment: computational insights into the selectivity mechanism through molecular dynamics simulations and QM/MM calculations. Phys Chem Chem Phys 2021; 23:17576-17590. [PMID: 34369509 DOI: 10.1039/d1cp02264d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The rational design of selective histone deacetylase 2 (HDAC2) inhibitors is beneficial for the therapeutic treatment of liver cancer, though HDAC2 is highly homologous to HDAC8, which may lead to undesired side effects due to the pan-inhibition towards HDAC2 and HDAC8. To clarify the structural basis of selective inhibition towards HDAC2 over HDAC8, we utilized multiple in silico strategies, including sequence alignment, structural comparison, molecular docking, molecular dynamics simulations, free energy calculations, alanine scanning mutagenesis, pharmacophore modeling, protein contacts atlas analysis and QM/MM calculations to study the binding patterns of HDAC2/8 selective inhibitors. Through the whole process described above, it is found that although HDAC2 has conserved GLY154 and PHE210 that also exist within HDAC8, namely GLY151 and PHE208, the two isoforms exhibit diverse binding modes towards their inhibitors. Typically, HDAC2 inhibitors interact with the Zn2+ ions through the core chelate group, while HDAC8 inhibitors adopt a bent conformation within the HDAC8 pocket that inclines to be in contact with the Zn2+ ions through the terminal hydroxamic acid group. In summary, our data comprehensively elucidate the selectivity mechanism towards HDAC2 over HDAC8, which would guide the rational design of selective HDAC2 inhibitors for liver cancer treatment.
Collapse
Affiliation(s)
- Ye Yang
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning 110016, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Kringel D, Malkusch S, Lötsch J. Drugs and Epigenetic Molecular Functions. A Pharmacological Data Scientometric Analysis. Int J Mol Sci 2021; 22:7250. [PMID: 34298869 PMCID: PMC8311652 DOI: 10.3390/ijms22147250] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/25/2021] [Accepted: 06/30/2021] [Indexed: 12/14/2022] Open
Abstract
Interactions of drugs with the classical epigenetic mechanism of DNA methylation or histone modification are increasingly being elucidated mechanistically and used to develop novel classes of epigenetic therapeutics. A data science approach is used to synthesize current knowledge on the pharmacological implications of epigenetic regulation of gene expression. Computer-aided knowledge discovery for epigenetic implications of current approved or investigational drugs was performed by querying information from multiple publicly available gold-standard sources to (i) identify enzymes involved in classical epigenetic processes, (ii) screen original biomedical scientific publications including bibliometric analyses, (iii) identify drugs that interact with epigenetic enzymes, including their additional non-epigenetic targets, and (iv) analyze computational functional genomics of drugs with epigenetic interactions. PubMed database search yielded 3051 hits on epigenetics and drugs, starting in 1992 and peaking in 2016. Annual citations increased to a plateau in 2000 and show a downward trend since 2008. Approved and investigational drugs in the DrugBank database included 122 compounds that interacted with 68 unique epigenetic enzymes. Additional molecular functions modulated by these drugs included other enzyme interactions, whereas modulation of ion channels or G-protein-coupled receptors were underrepresented. Epigenetic interactions included (i) drug-induced modulation of DNA methylation, (ii) drug-induced modulation of histone conformations, and (iii) epigenetic modulation of drug effects by interference with pharmacokinetics or pharmacodynamics. Interactions of epigenetic molecular functions and drugs are mutual. Recent research activities on the discovery and development of novel epigenetic therapeutics have passed successfully, whereas epigenetic effects of non-epigenetic drugs or epigenetically induced changes in the targets of common drugs have not yet received the necessary systematic attention in the context of pharmacological plasticity.
Collapse
Affiliation(s)
- Dario Kringel
- Institute of Clinical Pharmacology, Goethe-University, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (D.K.); (S.M.)
| | - Sebastian Malkusch
- Institute of Clinical Pharmacology, Goethe-University, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (D.K.); (S.M.)
| | - Jörn Lötsch
- Institute of Clinical Pharmacology, Goethe-University, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (D.K.); (S.M.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| |
Collapse
|
8
|
Rahm AK, Wieder T, Gramlich D, Müller ME, Wunsch MN, El Tahry FA, Heimberger T, Sandke S, Weis T, Most P, Katus HA, Thomas D, Lugenbiel P. Differential regulation of K Ca 2.1 (KCNN1) K + channel expression by histone deacetylases in atrial fibrillation with concomitant heart failure. Physiol Rep 2021; 9:e14835. [PMID: 34111326 PMCID: PMC8191401 DOI: 10.14814/phy2.14835] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 01/12/2023] Open
Abstract
Atrial fibrillation (AF) with concomitant heart failure (HF) poses a significant therapeutic challenge. Mechanism‐based approaches may optimize AF therapy. Small‐conductance, calcium‐activated K+ (KCa, KCNN) channels contribute to cardiac action potential repolarization. KCNN1 exhibits predominant atrial expression and is downregulated in chronic AF patients with preserved cardiac function. Epigenetic regulation is suggested by AF suppression following histone deacetylase (HDAC) inhibition. We hypothesized that HDAC‐dependent KCNN1 remodeling contributes to arrhythmogenesis in AF complicated by HF. The aim of this study was to assess KCNN1 and HDAC1–7 and 9 transcript levels in AF/HF patients and in a pig model of atrial tachypacing‐induced AF with reduced left ventricular function. In HL‐1 atrial myocytes, tachypacing and anti‐Hdac siRNAs were employed to investigate effects on Kcnn1 mRNA levels. KCNN1 expression displayed side‐specific remodeling in AF/HF patients with upregulation in left and suppression in right atrium. In pigs, KCNN1 remodeling showed intermediate phenotypes. HDAC levels were differentially altered in humans and pigs, reflecting highly variable epigenetic regulation. Tachypacing recapitulated downregulation of Hdacs1, 3, 4, 6, and 7 with a tendency towards reduced Kcnn1 levels in vitro, indicating that atrial high rates induce remodeling. Finally, Kcnn1 expression was decreased by knockdown of Hdacs2, 3, 6, and 7 and enhanced by genetic Hdac9 inactivation, while anti‐Hdac1, 4, and 5 siRNAs did not affect Kcnn1 transcript levels. In conclusion, KCNN1 and HDAC expression is differentially remodeled in AF complicated by HF. Direct regulation of KCNN1 by HDACs in atrial myocytes provides a basis for mechanism‐based antiarrhythmic therapy.
Collapse
Affiliation(s)
- Ann-Kathrin Rahm
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, Germany.,HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Teresa Wieder
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, Germany.,HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Heidelberg, Germany
| | - Dominik Gramlich
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, Germany.,HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Mara Elena Müller
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, Germany.,HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Maximilian N Wunsch
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, Germany.,HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Fadwa A El Tahry
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Tanja Heimberger
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Steffi Sandke
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Tanja Weis
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Patrick Most
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, Germany.,HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Heidelberg, Germany
| | - Hugo A Katus
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, Germany.,HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Dierk Thomas
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, Germany.,HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Patrick Lugenbiel
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, Germany.,HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
9
|
Rahm AK, Wieder T, Gramlich D, Müller ME, Wunsch MN, El Tahry FA, Heimberger T, Weis T, Most P, Katus HA, Thomas D, Lugenbiel P. HDAC2-dependent remodeling of K Ca2.2 (KCNN2) and K Ca2.3 (KCNN3) K + channels in atrial fibrillation with concomitant heart failure. Life Sci 2020; 266:118892. [PMID: 33310041 DOI: 10.1016/j.lfs.2020.118892] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 11/30/2020] [Accepted: 12/07/2020] [Indexed: 12/16/2022]
Abstract
AIMS Atrial fibrillation (AF) with concomitant heart failure (HF) is associated with prolonged atrial refractoriness. Small-conductance, calcium-activated K+ (KCa, KCNN) channels promote action potential (AP) repolarization. KCNN2 and KCNN3 variants are associated with AF risk. In addition, histone deacetylase (HDAC)-related epigenetic mechanisms have been implicated in AP regulation. We hypothesized that HDAC2-dependent remodeling of KCNN2 and KCNN3 expression contributes to atrial arrhythmogenesis in AF complicated by HF. The objectives were to assess HDAC2 and KCNN2/3 transcript levels in AF/HF patients and in a pig model, and to investigate cellular epigenetic effects of HDAC2 inactivation on KCNN expression. MATERIALS AND METHODS HDAC2 and KCNN2/3 transcript levels were quantified in patients with AF and HF, and in a porcine model of atrial tachypacing-induced AF and reduced left ventricular function. Tachypacing and anti-Hdac2 siRNA treatment were employed in HL-1 atrial myocytes to study effects on KCNN2/3 mRNA and KCa protein abundance. KEY FINDINGS Atrial KCNN2 and KCNN3 expression was reduced in AF/HF patients and in a corresponding pig model. HDAC2 displayed significant downregulation in humans and a tendency towards reduced expression in right atrial tissue of pigs. Tachypacing recapitulated downregulation of Kcnn2/KCa2.2, Kcnn3/KCa2.3 and Hdac2/HDAC2, indicating that high atrial rates trigger epigenetic remodeling mechanisms. Finally, knock-down of Hdac2 in vitro reduced Kcnn3/KCa2.3 expression. SIGNIFICANCE KCNN2/3 and HDAC2 expression is suppressed in AF complicated by HF. Hdac2 directly regulates Kcnn3 mRNA levels in atrial cells. The mechanistic and therapeutic significance of epigenetic electrophysiological effects in AF requires further validation.
Collapse
Affiliation(s)
- Ann-Kathrin Rahm
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Teresa Wieder
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Dominik Gramlich
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Mara Elena Müller
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Maximilian N Wunsch
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Fadwa A El Tahry
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Tanja Heimberger
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Tanja Weis
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Patrick Most
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Hugo A Katus
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Dierk Thomas
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany.
| | - Patrick Lugenbiel
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| |
Collapse
|
10
|
Mehrpouri M, Momeny M, Bashash D. Synergistic effects of BKM120 and panobinostat on pre-B acute lymphoblastic cells: an emerging perspective for the simultaneous inhibition of PI3K and HDACs. J Recept Signal Transduct Res 2020; 42:100-108. [PMID: 33969806 DOI: 10.1080/10799893.2020.1853159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The reputation of conventional treatment in acute lymphoblastic leukemia (ALL) has recently been questioned due to the considerable increment in the number of relapsed patients. The remarkable role of histone deacetylase (HDAC) enzymes in induction of chemo-resistance has provided an opportunity for HDAC inhibitors to be used as a treatment strategy in ALL; however, the compensatory activation of oncogenic pathways may negatively affect their promising effects. In the present study, we found an attenuating effect for PI3K axis on the anti-leukemic effects of panobinostat in pre-B ALL-derived Nalm-6 cells, as the harnessing of this pathway using BKM120 or CAL-101 resulted in a significant reduction in the number of viable cells as well as the metabolic activity. Moreover, we found the altered expression of p21, p27, c-Myc, and CDK4 upon co-treatment of the cells with panobinostat and BKM120, which was associated with a substantial blockage of cell cycle progression at G2/M phase. The companionship of the PI3K inhibitor with HDAC inhibitor also potentiated panobinostat-induced apoptotic cell death and enhanced the mRNA of Foxo3a and Foxo4. Conclusively, this study sheds light on the adjuvantive effects of BKM120 on panobinostat efficacy and outlined that the simultaneous inhibition of PI3K and HDACs may be a promising therapeutic approach to improve the cure rates of ALL.
Collapse
Affiliation(s)
- Mahdieh Mehrpouri
- Department of Laboratory Sciences, School of Allied Medical Sciences, Alborz University of Medical Sciences, Karaj, Iran
| | - Majid Momeny
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Cavallo F, Troglio F, Fagà G, Fancelli D, Shyti R, Trattaro S, Zanella M, D'Agostino G, Hughes JM, Cera MR, Pasi M, Gabriele M, Lazzarin M, Mihailovich M, Kooy F, Rosa A, Mercurio C, Varasi M, Testa G. High-throughput screening identifies histone deacetylase inhibitors that modulate GTF2I expression in 7q11.23 microduplication autism spectrum disorder patient-derived cortical neurons. Mol Autism 2020; 11:88. [PMID: 33208191 PMCID: PMC7677843 DOI: 10.1186/s13229-020-00387-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 10/01/2020] [Indexed: 12/27/2022] Open
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.
Collapse
Affiliation(s)
- Francesca Cavallo
- Department of Oncology and Hemato-Oncology, University of Milan, c/o High Definition Disease Modelling Lab: Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology IRCCS, Via Adamello 16, 20139, Milan, Italy
| | - Flavia Troglio
- High Definition Disease Modelling Lab: Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology IRCCS, Via Adamello 16, 20139, Milan, Italy
| | - Giovanni Fagà
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Via Adamello 16, 20139, Milan, Italy.,IFOM, the FIRC Institute of Molecular Oncology, Via Adamello 16, 20139, Milan, Italy
| | - Daniele Fancelli
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Via Adamello 16, 20139, Milan, Italy.,IFOM, the FIRC Institute of Molecular Oncology, Via Adamello 16, 20139, Milan, Italy
| | - Reinald Shyti
- High Definition Disease Modelling Lab: Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology IRCCS, Via Adamello 16, 20139, Milan, Italy
| | - Sebastiano Trattaro
- Department of Oncology and Hemato-Oncology, University of Milan, c/o High Definition Disease Modelling Lab: Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology IRCCS, Via Adamello 16, 20139, Milan, Italy.,High Definition Disease Modelling Lab: Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology IRCCS, Via Adamello 16, 20139, Milan, Italy
| | - Matteo Zanella
- High Definition Disease Modelling Lab: Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology IRCCS, Via Adamello 16, 20139, Milan, Italy.,Evotec SE, Hamburg, Germany
| | - Giuseppe D'Agostino
- High Definition Disease Modelling Lab: Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology IRCCS, Via Adamello 16, 20139, Milan, Italy.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - James M Hughes
- High Definition Disease Modelling Lab: Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology IRCCS, Via Adamello 16, 20139, Milan, Italy.,FPO - IRCCS, Candiolo Cancer Institute, SP 142 Km 3.95, 10060, Candiolo, TO, Italy
| | - Maria Rosaria Cera
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Via Adamello 16, 20139, Milan, Italy.,IFOM, the FIRC Institute of Molecular Oncology, Via Adamello 16, 20139, Milan, Italy
| | - Maurizio Pasi
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Via Adamello 16, 20139, Milan, Italy.,IFOM, the FIRC Institute of Molecular Oncology, Via Adamello 16, 20139, Milan, Italy
| | - Michele Gabriele
- High Definition Disease Modelling Lab: Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology IRCCS, Via Adamello 16, 20139, Milan, Italy.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, USA
| | - Maddalena Lazzarin
- Department of Oncology and Hemato-Oncology, University of Milan, c/o High Definition Disease Modelling Lab: Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology IRCCS, Via Adamello 16, 20139, Milan, Italy
| | - Marija Mihailovich
- Department of Oncology and Hemato-Oncology, University of Milan, c/o High Definition Disease Modelling Lab: Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology IRCCS, Via Adamello 16, 20139, Milan, Italy.,High Definition Disease Modelling Lab: Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology IRCCS, Via Adamello 16, 20139, Milan, Italy
| | - Frank Kooy
- Department of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | - Alessandro Rosa
- Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, P.le A. Moro 5, 00185, Rome, Italy.,Center for Life Nano Science, Istituto Italiano Di Tecnologia, Viale Regina Elena 291, 00161, Rome, Italy
| | - Ciro Mercurio
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Via Adamello 16, 20139, Milan, Italy.,IFOM, the FIRC Institute of Molecular Oncology, Via Adamello 16, 20139, Milan, Italy
| | - Mario Varasi
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Via Adamello 16, 20139, Milan, Italy.,IFOM, the FIRC Institute of Molecular Oncology, Via Adamello 16, 20139, Milan, Italy
| | - Giuseppe Testa
- High Definition Disease Modelling Lab: Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology IRCCS, Via Adamello 16, 20139, Milan, Italy. .,Department of Oncology and Hemato-Oncology, University of Milan, Via Santa Sofia 9, 20122, Milan, Italy. .,Human Technopole, Via Cristina Belgioioso, 171, 20157, Milan, Italy.
| |
Collapse
|
12
|
Kang DW, Hwang WC, Noh YN, Kang Y, Jang Y, Kim JA, Min DS. Phospholipase D1 is upregulated by vorinostat and confers resistance to vorinostat in glioblastoma. J Cell Physiol 2020; 236:549-560. [PMID: 32869317 PMCID: PMC7692931 DOI: 10.1002/jcp.29882] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 06/03/2020] [Accepted: 06/04/2020] [Indexed: 12/25/2022]
Abstract
Glioblastoma (GBM) is an aggressive brain tumor and drug resistance remains a major barrier for therapeutics. Epigenetic alterations are implicated in GBM pathogenesis, and epigenetic modulators including histone deacetylase (HDAC) inhibitors are exploited as promising anticancer therapies. Here, we demonstrate that phospholipase D1 (PLD1) is a transcriptional target of HDAC inhibitors and confers resistance to HDAC inhibitor in GBM. Treatment of vorinostat upregulates PLD1 through PKCζ‐Sp1 axis. Vorinostat induces dynamic changes in the chromatin structure and transcriptional machinery associated with PLD1 promoter region. Cotreatment of vorinostat with PLD1 inhibitor further attenuates invasion, angiogenesis, colony‐forming capacity, and self‐renewal capacity, compared with those of either treatment. PLD1 inhibitor overcomes resistance to vorinostat in GBM cells intracranial GBM tumors. Our finding provides new insight into the role of PLD1 as a target of resistance to vorinostat, and PLD1 inhibitor might provide the basis for therapeutic combinations with improved efficacy of HDAC inhibitor.
Collapse
Affiliation(s)
- Dong Woo Kang
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan, Republic of Korea
| | - Won Chan Hwang
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan, Republic of Korea.,College of Pharmacy, Yonsei University, Incheon, South Korea
| | - Yu Na Noh
- Institute for Innovative Cancer Research, Biomedical Research Center, Asan Medical Center, Seoul, Republic of Korea
| | - Youra Kang
- College of Pharmacy, Yeungnam University, Gyeongsan, South Korea
| | - Younghoon Jang
- Department of Biology and Chemistry, Changwon National University, Changwon, Korea
| | - Jung-Ae Kim
- College of Pharmacy, Yeungnam University, Gyeongsan, South Korea
| | - Do Sik Min
- College of Pharmacy, Yonsei University, Incheon, South Korea
| |
Collapse
|
13
|
Epigenomic profile and biological age. PAJAR - PAN AMERICAN JOURNAL OF AGING RESEARCH 2020. [DOI: 10.15448/2357-9641.2020.1.37125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Man ages at a constant chronological rate while their biological aging rate is extremely variable. Interventions to improve, or to slow the rate of biological aging are the subject of several research. The broad spectrum of molecules and its intricate role from the biological point of view and its relation with environmental factors are being investigated. Recently, researchers have been putting its efforts to understand the epigenetic mechanisms and how it can interfere with alterations in gene expression that leads to predisposition and, or pathological outcome. Some of these investigations have shed light about how one can determine the biological age from a simple blood sample, just by detecting the epigenetic alterations on only three CpGs sites with a reasonable certainty. Also, the enzymes inhibitors that can interfere with methylation and demethylation were effective to reverse the epigenetic mechanisms. Other studies have shown how the environmental changes since from early life can affect these alterations on the epigenome. Taking all together, some biomolecular markers are already available to determine the genetic background of an individual and this information can be used to guide the lifestyle in order to prevent some future diseases development and/or improve the quality of later life.
Collapse
|
14
|
Singhal S, Pathak M, Agrawala PK, Ojha H. Design and in silico screening of aryl allyl mercaptan analogs as potential histone deacetylases (HDAC) inhibitors. Heliyon 2020; 6:e03517. [PMID: 32426531 PMCID: PMC7225394 DOI: 10.1016/j.heliyon.2020.e03517] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 01/30/2020] [Accepted: 02/27/2020] [Indexed: 12/24/2022] Open
Abstract
The Zn+2 HDACIs show promising anticancer activity. Allyl mercaptan (AM), a metastabilzed monomeric form of diallyl disulphide (DADS) shows better HDACI activity. The present work screens a dataset of aryl AM derivatives 1(a-g) for potential HDACI action viain silico models. DFT calculations predicted the geometrical parameters and frontier orbital calculations suggested better chemical reactivity. Negative chemical potential and NBO hyper conjugative interactions predicted their chemical stability. ADME study confirmed favourable drug likeliness. Molecular docked models suggested the formation of coordinate bond between sulphur of allylmercaptan and Zn2+ cofactor of HDAC8. Besides, models also predicted the dominance of hydrophobic interactions. The aryl AM analogs docked perfectly with HDAC3 as well. The glide score and S-Zn distance of compounds 1a, 1f and 1g were found to be better than allylmercaptan. Therefore, the designed aryl AM analogs filtered as better HDACIs. These could be further used for design and synthesis of new improved HDACIs.
Collapse
Affiliation(s)
- Sugandha Singhal
- Synthetic Organic and Natural Products Laboratory, University School of Basic and Applied Sciences, Guru Gobind Singh Indraprastha University, Sector 16-C, Dwarka, New Delhi, 110078, India
| | - Mallika Pathak
- Department of Chemistry, Miranda House, University of Delhi, Delhi, 110007, India
| | - Paban K Agrawala
- Department of Radiation Genetics and Epigenetics,Division of Radiation Biodosimetry, Institute of Nuclear Medicine & Allied Sciences, Delhi, 110054, India
| | - Himanshu Ojha
- CBRN Protection and Decontamination Research Group, Division of CBRN Defence, Institute of Nuclear Medicine and Allied Sciences (INMAS) DRDO, Delhi, 110054, India
| |
Collapse
|
15
|
Hashemi P, Sadowski I. Diversity of small molecule HIV-1 latency reversing agents identified in low- and high-throughput small molecule screens. Med Res Rev 2020; 40:881-908. [PMID: 31608481 PMCID: PMC7216841 DOI: 10.1002/med.21638] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 08/26/2019] [Accepted: 09/16/2019] [Indexed: 12/12/2022]
Abstract
The latency phenomenon produced by human immunodeficiency virus (HIV-1) prevents viral clearance by current therapies, and consequently development of a cure for HIV-1 disease represents a formidable challenge. Research over the past decade has resulted in identification of small molecules that are capable of exposing HIV-1 latent reservoirs, by reactivation of viral transcription, which is intended to render these infected cells sensitive to elimination by immune defense recognition or apoptosis. Molecules with this capability, known as latency-reversing agents (LRAs) could lead to realization of proposed HIV-1 cure strategies collectively termed "shock and kill," which are intended to eliminate the latently infected population by forced reactivation of virus replication in combination with additional interventions that enhance killing by the immune system or virus-mediated apoptosis. Here, we review efforts to discover novel LRAs via low- and high-throughput small molecule screens, and summarize characteristics and biochemical properties of chemical structures with this activity. We expect this analysis will provide insight toward further research into optimized designs for new classes of more potent LRAs.
Collapse
Affiliation(s)
- Pargol Hashemi
- Biochemistry and Molecular Biology, Molecular Epigenetics, Life Sciences InstituteUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Ivan Sadowski
- Biochemistry and Molecular Biology, Molecular Epigenetics, Life Sciences InstituteUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| |
Collapse
|
16
|
Schröder C, Khatri R, Petry SF, Linn T. Class I and II Histone Deacetylase Inhibitor LBH589 Promotes Endocrine Differentiation in Bone Marrow Derived Human Mesenchymal Stem Cells and Suppresses Uncontrolled Proliferation. Exp Clin Endocrinol Diabetes 2020; 129:357-364. [PMID: 32052390 DOI: 10.1055/a-1103-1900] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Mesenchymal stem cells are useful tools employed in clinical and preclinical medicine. Their beneficial potential in especially degenerative as well as autoimmune diseases is a constant focus of research. Regarding diabetes mellitus, transplantation of stem cells is seen as a possible therapeutic approach to overcome the loss of endocrine pancreatic cells. It was reported that co-transplantation of mesenchymal stem cells with pancreatic islet cells improves function and survival of the graft. However, these multipotent progenitors may be able to form tumors, especially under immunosuppressed conditions. Histone deacetylase inhibitors might offer the potential to overcome this issue. These small molecules can induce cell differentiation and control proliferation. Their potential to control lineage development of stem cells has been distinctly demonstrated in the treatment of cancer, mainly in hematopoietic neoplasias.In this study, we demonstrate that human bone marrow-derived mesenchymal stem cells exhibit low carcinogenic potential in an immunosuppressed condition in vivo. Further, the effect of histone deacetylase inhibitors LBH589, MS-275, and MGCD0103 was examined after normalizing histone deacetylase activities in culture. Interestingly, transcripts of insulin gene enhancer protein and paired-box-gene 6, two markers of pancreatic endocrine differentiation were constitutively expressed in the cell line. The broad spectrum inhibitor of class I and class II histone deacetylases LBH589 upregulated the expression of these transcription factors in a significant way, whereas addition of selective class I histone deacetylase inhibitors MS-275 and MGCD0103 did not result in significant changes in gene expression.In conclusion, we deliver evidence that a combined class I and II histone deacetylase inhibition is able to modulate the transcripts of differentiation markers of mesenchymal stem cells. The treatment holds the capability to facilitate endocrine differentiation in future approaches to replace endocrine cells by stem cell therapy.
Collapse
Affiliation(s)
- Christoph Schröder
- Clinical Research Unit, Centre of Internal Medicine, Justus Liebig University Giessen, Germany.,Medizinische Hochschule Hannover, Hannover, Germany
| | - Rahul Khatri
- Clinical Research Unit, Centre of Internal Medicine, Justus Liebig University Giessen, Germany
| | | | - Thomas Linn
- Clinical Research Unit, Centre of Internal Medicine, Justus Liebig University Giessen, Germany
| |
Collapse
|
17
|
p53 at the Crossroads between Different Types of HDAC Inhibitor-Mediated Cancer Cell Death. Int J Mol Sci 2019; 20:ijms20102415. [PMID: 31096697 PMCID: PMC6567317 DOI: 10.3390/ijms20102415] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/06/2019] [Accepted: 05/13/2019] [Indexed: 12/19/2022] Open
Abstract
Cancer is a complex genetic and epigenetic-based disease that has developed an armada of mechanisms to escape cell death. The deregulation of apoptosis and autophagy, which are basic processes essential for normal cellular activity, are commonly encountered during the development of human tumors. In order to assist the cancer cell in defeating the imbalance between cell growth and cell death, histone deacetylase inhibitors (HDACi) have been employed to reverse epigenetically deregulated gene expression caused by aberrant post-translational protein modifications. These interfere with histone acetyltransferase- and deacetylase-mediated acetylation of both histone and non-histone proteins, and thereby exert a wide array of HDACi-stimulated cytotoxic effects. Key determinants of HDACi lethality that interfere with cellular growth in a multitude of tumor cells are apoptosis and autophagy, which are either mutually exclusive or activated in combination. Here, we compile known molecular signals and pathways involved in the HDACi-triggered induction of apoptosis and autophagy. Currently, the factors that determine the mode of HDACi-elicited cell death are mostly unclear. Correspondingly, we also summarized as yet established intertwined mechanisms, in particular with respect to the oncogenic tumor suppressor protein p53, that drive the interplay between apoptosis and autophagy in response to HDACi. In this context, we also note the significance to determine the presence of functional p53 protein levels in the cancer cell. The confirmation of the context-dependent function of autophagy will pave the way to improve the benefit from HDACi-mediated cancer treatment.
Collapse
|
18
|
Wobser M, Weber A, Glunz A, Tauch S, Seitz K, Butelmann T, Hesbacher S, Goebeler M, Bartz R, Kohlhof H, Schrama D, Houben R. Elucidating the mechanism of action of domatinostat (4SC-202) in cutaneous T cell lymphoma cells. J Hematol Oncol 2019; 12:30. [PMID: 30885250 PMCID: PMC6423872 DOI: 10.1186/s13045-019-0719-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 03/07/2019] [Indexed: 02/06/2023] Open
Abstract
Background Targeting epigenetic modifiers is effective in cutaneous T cell lymphoma (CTCL). However, there is a need for further improvement of this therapeutic approach. Here, we compared the mode of action of romidepsin (FK228), an established class I histone deacetylase inhibitor, and domatinostat (4SC-202), a novel inhibitor of class I HDACs, which has been reported to also target the lysine-specific histone demethylase 1A (LSD1). Methods We performed MTS assays and flow cytometric analyses of propidium iodide or annexin V-stained cells to assess drug impact on cellular proliferation, cell cycle distribution, and survival. Histone acetylation and methylation as well as caspase activation was analyzed by immunoblot. Gene expression analysis was performed using NanosString technology. Knockdown and knockout of LSD1 was achieved with shRNA and CRISPR/Cas9, respectively, while the CRISPR/Cas9 synergistic activation mediator system was used to induce expression of endogenous HDACs and LSD1. Furthermore, time-lapse fluorescence microscopy and an in vitro tubulin polymerization assay were applied. Results While FK228 as well as 4SC-202 potently induced cell death in six different CTCL cell lines, only in the case of 4SC-202 death was preceded by an accumulation of cells in the G2/M phase of the cell cycle. Surprisingly, apoptosis and accumulation of cells with double DNA content occurred already at 4SC-202 concentrations hardly affecting histone acetylation and methylation, and provoking significantly less changes in gene expression compared to biologically equivalent doses of FK228. Indeed, we provide evidence that the 4SC-202-induced G2/M arrest in CTCL cells is independent of de novo transcription. Furthermore, neither enforced expression of HDAC1 nor knockdown or knockout of LSD1 affected the 4SC-202-induced effects. Since time-lapse microscopy revealed that 4SC-202 could affect mitotic spindle formation, we performed an in vitro tubulin polymerization assay revealing that 4SC-202 can directly inhibit microtubule formation. Conclusions We demonstrate that 4SC-202, a drug currently tested in clinical trials, effectively inhibits growth of CTCL cells. The anti-cancer cell activity of 4SC-202 is however not limited to LSD1-inhibition, modulation of histone modifications, and consecutive alteration of gene expression. Indeed, the compound is also a potent microtubule-destabilizing agent. Electronic supplementary material The online version of this article (10.1186/s13045-019-0719-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Marion Wobser
- Department of Dermatology, Venereology and Allergology, University Hospital Wuerzburg, Josef-Schneider-Str. 2, 97080, Wuerzburg, Germany
| | - Alexandra Weber
- Department of Dermatology, Venereology and Allergology, University Hospital Wuerzburg, Josef-Schneider-Str. 2, 97080, Wuerzburg, Germany
| | - Amelie Glunz
- Department of Dermatology, Venereology and Allergology, University Hospital Wuerzburg, Josef-Schneider-Str. 2, 97080, Wuerzburg, Germany
| | - Saskia Tauch
- Department of Dermatology, Venereology and Allergology, University Hospital Wuerzburg, Josef-Schneider-Str. 2, 97080, Wuerzburg, Germany
| | - Kristina Seitz
- Department of Dermatology, Venereology and Allergology, University Hospital Wuerzburg, Josef-Schneider-Str. 2, 97080, Wuerzburg, Germany
| | - Tobias Butelmann
- Department of Dermatology, Venereology and Allergology, University Hospital Wuerzburg, Josef-Schneider-Str. 2, 97080, Wuerzburg, Germany
| | - Sonja Hesbacher
- Department of Dermatology, Venereology and Allergology, University Hospital Wuerzburg, Josef-Schneider-Str. 2, 97080, Wuerzburg, Germany
| | - Matthias Goebeler
- Department of Dermatology, Venereology and Allergology, University Hospital Wuerzburg, Josef-Schneider-Str. 2, 97080, Wuerzburg, Germany
| | - René Bartz
- 4SC company, Planegg-Martinsried, Germany
| | | | - David Schrama
- Department of Dermatology, Venereology and Allergology, University Hospital Wuerzburg, Josef-Schneider-Str. 2, 97080, Wuerzburg, Germany
| | - Roland Houben
- Department of Dermatology, Venereology and Allergology, University Hospital Wuerzburg, Josef-Schneider-Str. 2, 97080, Wuerzburg, Germany.
| |
Collapse
|
19
|
Guo D, Hong D, Wang P, Wang J, Chen L, Zhao W, Zhang L, Yao C, Chu B, Chen S, Li Z, Chen H. Histone deacetylase inhibitor CI-994 inhibits osteoclastogenesis via suppressing NF-κB and the downstream c-Fos/NFATc1 signaling pathways. Eur J Pharmacol 2019; 848:96-104. [PMID: 30682334 DOI: 10.1016/j.ejphar.2019.01.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 01/16/2019] [Accepted: 01/22/2019] [Indexed: 01/09/2023]
Abstract
[4-(acetylamino)-N-(2-amino-phenyl) benzamide] (CI-994) is a histone deacetylase 1-3 specific inhibitor that has been shown to indirectly increase the production of Dickkopf-1, which is an inhibitor of osteoclastic development. However, whether CI-994 has an influence on receptor activator of nuclear factor-kappa B ligand (RANKL)-induced osteoclastogenesis is still unclear; in our study, this mechanism was investigated. In an in vitro study, using a tartrate-resistant acid phosphatase (TRAP) stain, an F-actin ring, bone absorption test, quantitative PCR and Western blotting, the role of CI-994 in osteoclastogenesis and the expression of related genes and proteins were investigated. In an in vivo study, the effect of CI-994 on osteolysis was evaluated using a titanium particle-induced murine calvarial osteolysis model. Our results indicated that CI-994 inhibited osteoclast differentiation and the function of bone resorption without cytotoxic effects. Moreover, CI-994 inhibited the expression of osteoclast-related genes, including ACP5, CTSK, NFATc1, c-Fos, DC-STAMP and V-ATPase-d2. Furthermore, CI-994 suppressed the phosphorylation of IκBα and p65 and the expression of downstream c-Fos and NFATc1. Consistent with the in vitro results described above, our in vivo experiment indicated that CI-994 inhibited Ti-induced osteolysis. In conclusion, CI-994 inhibited osteoclastogenesis by suppressing NF-κB and the downstream c-Fos/NFATc1 signaling pathway. Thus, this study showed the possibility of using CI-994 for the treatment of exorbitant osteoclastic bone resorption.
Collapse
Affiliation(s)
- Di Guo
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai 317000, China; Bone development and metabolism research center of Taizhou Hospital, Linhai 317000, China
| | - Dun Hong
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai 317000, China; Bone development and metabolism research center of Taizhou Hospital, Linhai 317000, China
| | - Peng Wang
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai 317000, China; Bone development and metabolism research center of Taizhou Hospital, Linhai 317000, China
| | - Jiacheng Wang
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai 317000, China; Bone development and metabolism research center of Taizhou Hospital, Linhai 317000, China
| | - Lihua Chen
- Enze Medical Research Center, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai 317000, China; Bone development and metabolism research center of Taizhou Hospital, Linhai 317000, China
| | - Weibo Zhao
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai 317000, China; Bone development and metabolism research center of Taizhou Hospital, Linhai 317000, China
| | - Liwei Zhang
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai 317000, China; Orthopedic Institute, Soochow University of Medicine, Soochow 215008, China
| | - Can Yao
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai 317000, China; Bone development and metabolism research center of Taizhou Hospital, Linhai 317000, China
| | - Binxiang Chu
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai 317000, China; Bone development and metabolism research center of Taizhou Hospital, Linhai 317000, China
| | - Shenao Chen
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai 317000, China; Bone development and metabolism research center of Taizhou Hospital, Linhai 317000, China
| | - Zhiyan Li
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai 317000, China; Bone development and metabolism research center of Taizhou Hospital, Linhai 317000, China
| | - Haixiao Chen
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai 317000, China; Bone development and metabolism research center of Taizhou Hospital, Linhai 317000, China.
| |
Collapse
|
20
|
Epigenetic Targeting of Autophagy via HDAC Inhibition in Tumor Cells: Role of p53. Int J Mol Sci 2018; 19:ijms19123952. [PMID: 30544838 PMCID: PMC6321134 DOI: 10.3390/ijms19123952] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 12/05/2018] [Accepted: 12/06/2018] [Indexed: 12/19/2022] Open
Abstract
Tumor development and progression is the consequence of genetic as well as epigenetic alterations of the cell. As part of the epigenetic regulatory system, histone acetyltransferases (HATs) and deacetylases (HDACs) drive the modification of histone as well as non-histone proteins. Derailed acetylation-mediated gene expression in cancer due to a delicate imbalance in HDAC expression can be reversed by histone deacetylase inhibitors (HDACi). Histone deacetylase inhibitors have far-reaching anticancer activities that include the induction of cell cycle arrest, the inhibition of angiogenesis, immunomodulatory responses, the inhibition of stress responses, increased generation of oxidative stress, activation of apoptosis, autophagy eliciting cell death, and even the regulation of non-coding RNA expression in malignant tumor cells. However, it remains an ongoing issue how tumor cells determine to respond to HDACi treatment by preferentially undergoing apoptosis or autophagy. In this review, we summarize HDACi-mediated mechanisms of action, particularly with respect to the induction of cell death. There is a keen interest in assessing suitable molecular factors allowing a prognosis of HDACi-mediated treatment. Addressing the results of our recent study, we highlight the role of p53 as a molecular switch driving HDACi-mediated cellular responses towards one of both types of cell death. These findings underline the importance to determine the mutational status of p53 for an effective outcome in HDACi-mediated tumor therapy.
Collapse
|
21
|
Ghayad SE, Rammal G, Sarkis O, Basma H, Ghamloush F, Fahs A, Karam M, Harajli M, Rabeh W, Mouawad JE, Zalzali H, Saab R. The histone deacetylase inhibitor Suberoylanilide Hydroxamic Acid (SAHA) as a therapeutic agent in rhabdomyosarcoma. Cancer Biol Ther 2018; 20:272-283. [PMID: 30307360 DOI: 10.1080/15384047.2018.1529093] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Rhabdomyosarcoma (RMS) is an aggressive childhood sarcoma with two distinct subtypes, embryonal (ERMS) and alveolar (ARMS) histologies. More effective treatment is needed to improve outcomes, beyond conventional cytotoxic chemotherapy. The pan-histone deacetylase inhibitor, Suberoylanilide Hydroxamic Acid (SAHA), has shown promising efficacy in limited preclinical studies. We used a panel of human ERMS and ARMS cell lines and xenografts to evaluate the effects of SAHA as a therapeutic agent in both RMS subtypes. SAHA decreased cell viability by inhibiting S-phase progression in all cell lines tested, and induced apoptosis in all but one cell line. Molecularly, SAHA-treated cells showed activation of a DNA damage response, induction of the cell cycle inhibitors p21Cip1 and p27Kip1 and downregulation of Cyclin D1. In a subset of RMS cell lines, SAHA promoted features of cellular senescence and myogenic differentiation. Interestingly, SAHA treatment profoundly decreased protein levels of the driver fusion oncoprotein PAX3-FOXO1 in ARMS cells at a post-translational level. In vivo, SAHA-treated xenografts showed increased histone acetylation and induction of a DNA damage response, along with variable upregulation of p21Cip1 and p27Kip1. However, while the ARMS Rh41 xenograft tumor growth was significantly inhibited, there was no significant inhibition of the ERMS tumor xenograft RD. Thus, our work shows that, while SAHA is effective against ERMS and ARMS tumor cells in vitro, it has divergent in vivo effects . Together with the observed effects on the PAX3-FOXO1 fusion protein, these data suggest SAHA as a possible therapeutic agent for clinical testing in patients with fusion protein-positive RMS.
Collapse
Affiliation(s)
- Sandra E Ghayad
- a Department of Biology, Faculty of Science II , Lebanese University , Fanar , Lebanon
| | - Ghina Rammal
- a Department of Biology, Faculty of Science II , Lebanese University , Fanar , Lebanon.,b Department of Pediatrics and Adolescent Medicine , American University of Beirut , Beirut , Lebanon
| | - Omar Sarkis
- b Department of Pediatrics and Adolescent Medicine , American University of Beirut , Beirut , Lebanon
| | - Hussein Basma
- b Department of Pediatrics and Adolescent Medicine , American University of Beirut , Beirut , Lebanon
| | - Farah Ghamloush
- b Department of Pediatrics and Adolescent Medicine , American University of Beirut , Beirut , Lebanon
| | - Assil Fahs
- a Department of Biology, Faculty of Science II , Lebanese University , Fanar , Lebanon
| | - Mia Karam
- a Department of Biology, Faculty of Science II , Lebanese University , Fanar , Lebanon
| | - Mohamad Harajli
- b Department of Pediatrics and Adolescent Medicine , American University of Beirut , Beirut , Lebanon
| | - Wissam Rabeh
- b Department of Pediatrics and Adolescent Medicine , American University of Beirut , Beirut , Lebanon
| | - Joe E Mouawad
- b Department of Pediatrics and Adolescent Medicine , American University of Beirut , Beirut , Lebanon
| | - Hassan Zalzali
- b Department of Pediatrics and Adolescent Medicine , American University of Beirut , Beirut , Lebanon
| | - Raya Saab
- b Department of Pediatrics and Adolescent Medicine , American University of Beirut , Beirut , Lebanon.,c Department of Anatomy, Cell Biology and Physiology , American University of Beirut , Beirut , Lebanon
| |
Collapse
|
22
|
Sanaei M, Kavoosi F, Roustazadeh A, Shahsavani H. In Vitro Effect of the Histone Deacetylase Inhibitor Valproic Acid on Viability and Apoptosis of the PLC/PRF5 Human Hepatocellular Carcinoma Cell Line. Asian Pac J Cancer Prev 2018; 19:2507-2510. [PMID: 30256044 PMCID: PMC6249479 DOI: 10.22034/apjcp.2018.19.9.2507] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The nucleosome is the fundamental building block of eukaryotic chromatin formed by DNA and histone proteins. Chromatin modifications such as acetylation, methylation, and phosphorylation are necessary for protection, replication, and gene transcription. Histone deacetylases (HDACs) are a group of enzymes that remove acetyl groups to re-establish positive charges on histones and aberrant deacetylation may lead to tumorigenesis in different tissues. Histone deacetylase inhibitors (HDACIs) are a class of chemotherapeutic agent that can reactivate gene expression and inhibit the growth of tumor cells by histone deacetylase inhibition. HDACI valproic acid (VPA) has shown potent anticancer effects in vitro and in vivo. Previously, we reported that VAP can inhibit the growth and induce apoptosis of human colon carcinoma HT 29 and hepatocellular carcinoma HepG 2 cells. The aim of the present study was to access the effect of VPA on proliferation and apoptosis of the human hepatocellular carcinoma (HCC) PLC/PRF5 cell line.
Collapse
Affiliation(s)
- Masumeh Sanaei
- Research Center for Non-Communicable Diseases, Jahrom University of Medical Sciences, Jahrom, Iran.
| | | | | | | |
Collapse
|
23
|
Differences in Functional Expression of Connexin43 and Na V1.5 by Pan- and Class-Selective Histone Deacetylase Inhibition in Heart. Int J Mol Sci 2018; 19:ijms19082288. [PMID: 30081552 PMCID: PMC6121244 DOI: 10.3390/ijms19082288] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 08/01/2018] [Accepted: 08/02/2018] [Indexed: 12/22/2022] Open
Abstract
Class-selective histone deacetylase (HDAC) inhibitors were designed to improve safety profiles and therapeutic effectiveness in the treatment of multiple cancers relative to pan-HDAC inhibitors. However, the underlying mechanisms for their therapeutic and cardiotoxic potentials remain poorly understood. Cardiac sodium currents (INa) and gap junction conductance (gj) were measured by whole cell patch clamp techniques on primary cultures of neonatal cardiomyocytes. Cardiac NaV1.5 sodium channel and connexin43 (Cx43) gap junction protein levels were assessed by Western blot analyses. Panobinostat produced concentration-dependent reductions in ventricular gj, peak INa density, and NaV1.5 protein expression levels. Membrane voltage (Vm)-dependent activation of INa was shifted by +3 to 6 mV with no effect on inactivation. Entinostat (1 μM) did not affect ventricular gj, peak INa density, or INa activation. However, the INa half-inactivation voltage (V½) was shifted by −3.5 mV. Ricolinostat had only minor effects on ventricular gj and INa properties, though INa activation was shifted by −4 mV. Cx43 and NaV1.5 protein expression levels were not altered by class-selective HDAC inhibitors. The lack of effects of class-selective HDAC inhibitors on ventricular gj and INa may help explain the improved cardiac safety profile of entinostat and ricolinostat.
Collapse
|
24
|
Yong KJ, Li A, Ou WB, Hong CKY, Zhao W, Wang F, Tatetsu H, Yan B, Qi L, Fletcher JA, Yang H, Soo R, Tenen DG, Chai L. Targeting SALL4 by entinostat in lung cancer. Oncotarget 2018; 7:75425-75440. [PMID: 27705911 PMCID: PMC5342750 DOI: 10.18632/oncotarget.12251] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 09/13/2016] [Indexed: 11/26/2022] Open
Abstract
The overall survival of lung cancer patients remains dismal despite the availability of targeted therapies. Oncofetal protein SALL4 is a novel cancer target. We herein report that SALL4 was aberrantly expressed in a subset of lung cancer patients with poor survival. SALL4 silencing by RNA interference or SALL4 peptide inhibitor treatment led to impaired lung cancer cell growth. Expression profiling of SALL4-knockdown cells demonstrated that both the EGFR and IGF1R signaling pathways were affected. Connectivity Map analysis revealed the HDAC inhibitor entinostat as a potential drug in treating SALL4-expressing cancers, and this was confirmed in 17 lung cancer cell lines. In summary, we report for the first time that entinostat can target SALL4-positive lung cancer. This lays the foundation for future clinical studies evaluating the therapeutic efficacy of entinostat in SALL4-positive lung cancer patients.
Collapse
Affiliation(s)
- Kol Jia Yong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Ailing Li
- Department of Pathology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Wen-Bin Ou
- Department of Pathology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA.,Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, China
| | - Clarice Kit Yee Hong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Wenxiu Zhao
- Department of Pathology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Fei Wang
- Department of Pathology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Hiro Tatetsu
- Department of Pathology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Benedict Yan
- Department of Laboratory Medicine, National University Hospital, National University Health System, Singapore
| | - Lihua Qi
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Jonathan A Fletcher
- Department of Pathology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Henry Yang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Ross Soo
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Daniel G Tenen
- Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Harvard Stem Cell Institute, Boston, MA, USA
| | - Li Chai
- Department of Pathology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
25
|
Yan M, Qian YM, Yue CF, Wang ZF, Wang BC, Zhang W, Zheng FM, Liu Q. Inhibition of histone deacetylases induces formation of multipolar spindles and subsequent p53-dependent apoptosis in nasopharyngeal carcinoma cells. Oncotarget 2018; 7:44171-44184. [PMID: 27283770 PMCID: PMC5190087 DOI: 10.18632/oncotarget.9922] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 05/16/2016] [Indexed: 11/25/2022] Open
Abstract
Histone deacetylases (HDACs) play crucial roles in the initiation and progression of cancer, offering a promising target for cancer therapy. HDACs inhibitor MGCD0103 (MGCD) exhibits effective anti-tumor activity by blocking proliferation and inducing cell death in malignant cells. However, the molecular mechanisms of HDACs inhibition induces cell death have not been well elucidated. In this study, we showed that MGCD effectively restored histone acetylation, suppressed cell growth and induced apoptosis in two-dimensional (2D) and three-dimensional (3D) cultured CNE1 and CNE2 nasopharyngeal carcinoma (NPC) cells. Importantly, MGCD arrested cell cycle at mitosis (M) phase with formation of multipolar spindles, which was associated with activated p53-mediated postmitotic checkpoint pathway to induce apoptotic cell death. Moreover, MGCD-induced apoptosis was decreased by inhibition of p53 using short interfering RNA (siRNA), suggesting that p53 was required for MGCD-induced cell apoptosis. Consistently, MGCD in combination with Nutlin-3, a MDM2 inhibitor showed synergistic effect on inducing apoptosis in 2D and 3D cultured CNE2 cells. Collectively, our data revealed that MGCD induced p53-dependent cell apoptosis following formation of multipolar spindles in NPC cells, suggesting the therapeutic potential of combinations of HDACs and MDM2 inhibitors for NPC treatment.
Collapse
Affiliation(s)
- Min Yan
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Institute of Cancer Stem Cell, Dalian, China.,Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yuan-Min Qian
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Institute of Cancer Stem Cell, Dalian, China
| | - Cai-Feng Yue
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Institute of Cancer Stem Cell, Dalian, China.,Department of Laboratory Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zi-Feng Wang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Institute of Cancer Stem Cell, Dalian, China
| | - Bi-Cheng Wang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Institute of Cancer Stem Cell, Dalian, China
| | - Wei Zhang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Institute of Cancer Stem Cell, Dalian, China
| | - Fei-Meng Zheng
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Institute of Cancer Stem Cell, Dalian, China.,Department of Medical Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Quentin Liu
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Institute of Cancer Stem Cell, Dalian, China
| |
Collapse
|
26
|
Topper MJ, Vaz M, Chiappinelli KB, DeStefano Shields CE, Niknafs N, Yen RWC, Wenzel A, Hicks J, Ballew M, Stone M, Tran PT, Zahnow CA, Hellmann MD, Anagnostou V, Strissel PL, Strick R, Velculescu VE, Baylin SB. Epigenetic Therapy Ties MYC Depletion to Reversing Immune Evasion and Treating Lung Cancer. Cell 2017; 171:1284-1300.e21. [PMID: 29195073 DOI: 10.1016/j.cell.2017.10.022] [Citation(s) in RCA: 344] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 08/07/2017] [Accepted: 10/13/2017] [Indexed: 12/25/2022]
Abstract
Combining DNA-demethylating agents (DNA methyltransferase inhibitors [DNMTis]) with histone deacetylase inhibitors (HDACis) holds promise for enhancing cancer immune therapy. Herein, pharmacologic and isoform specificity of HDACis are investigated to guide their addition to a DNMTi, thus devising a new, low-dose, sequential regimen that imparts a robust anti-tumor effect for non-small-cell lung cancer (NSCLC). Using in-vitro-treated NSCLC cell lines, we elucidate an interferon α/β-based transcriptional program with accompanying upregulation of antigen presentation machinery, mediated in part through double-stranded RNA (dsRNA) induction. This is accompanied by suppression of MYC signaling and an increase in the T cell chemoattractant CCL5. Use of this combination treatment schema in mouse models of NSCLC reverses tumor immune evasion and modulates T cell exhaustion state towards memory and effector T cell phenotypes. Key correlative science metrics emerge for an upcoming clinical trial, testing enhancement of immune checkpoint therapy for NSCLC.
Collapse
Affiliation(s)
- Michael J Topper
- Department of Oncology, The Johns Hopkins School of Medicine, The Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287, USA; The Graduate Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Michelle Vaz
- Department of Oncology, The Johns Hopkins School of Medicine, The Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287, USA
| | - Katherine B Chiappinelli
- Department of Microbiology, Immunology, and Tropical Medicine, The George Washington University Cancer Center, Washington, DC 20052, USA
| | - Christina E DeStefano Shields
- Department of Oncology, The Johns Hopkins School of Medicine, The Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287, USA
| | - Noushin Niknafs
- Department of Oncology, The Johns Hopkins School of Medicine, The Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287, USA
| | - Ray-Whay Chiu Yen
- Department of Oncology, The Johns Hopkins School of Medicine, The Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287, USA
| | - Alyssa Wenzel
- The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Jessica Hicks
- Department of Urologic Pathology, The Johns Hopkins School of Medicine, The Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287, USA
| | - Matthew Ballew
- Department of Radiation Oncology & Molecular Radiation Sciences, The Johns Hopkins School of Medicine, The Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287, USA
| | - Meredith Stone
- Department of Oncology, The Johns Hopkins School of Medicine, The Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287, USA; The Graduate Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Phuoc T Tran
- Department of Radiation Oncology & Molecular Radiation Sciences, The Johns Hopkins School of Medicine, The Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287, USA
| | - Cynthia A Zahnow
- Department of Oncology, The Johns Hopkins School of Medicine, The Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287, USA
| | - Matthew D Hellmann
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Valsamo Anagnostou
- Department of Oncology, The Johns Hopkins School of Medicine, The Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287, USA
| | - Pamela L Strissel
- Department of Gynecology and Obstetrics, Laboratory for Molecular Medicine, University-Clinic Erlangen, 91054 Erlangen, Germany
| | - Reiner Strick
- Department of Gynecology and Obstetrics, Laboratory for Molecular Medicine, University-Clinic Erlangen, 91054 Erlangen, Germany
| | - Victor E Velculescu
- Department of Oncology, The Johns Hopkins School of Medicine, The Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287, USA
| | - Stephen B Baylin
- Department of Oncology, The Johns Hopkins School of Medicine, The Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287, USA.
| |
Collapse
|
27
|
Lee JW, Yang DH, Park S, Han HK, Park JW, Kim BY, Um SH, Moon EY. Trichostatin A resistance is facilitated by HIF-1α acetylation in HeLa human cervical cancer cells under normoxic conditions. Oncotarget 2017; 9:2035-2049. [PMID: 29416751 PMCID: PMC5788619 DOI: 10.18632/oncotarget.23327] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 12/08/2017] [Indexed: 12/31/2022] Open
Abstract
Trichostatin A (TSA) is an anticancer drug that inhibits histone deacetylases (HDACs). Hypoxia-inducible factor 1 (HIF-1) participates in tumor angiogenesis by upregulating target genes, such as vascular endothelial growth factor (VEGF). In the present study, we investigated whether TSA treatment increases HIF-1α stabilization via acetylation under normoxic conditions, which would lead to VEGF upregulation and resistance to anticancer drugs. TSA enhanced total HIF-1α and VEGF-HRE reporter activity under normoxic conditions. When cells were transfected with GFP-HIF-1α, treatment with TSA increased the number of green fluorescence protein (GFP)-positive cells. TSA also enhanced the nuclear translocation of HIF-1α protein, as assessed by immunoblotting and as evidenced by increased nuclear localization of GFP-HIF-1α. An increase in the interaction between HIF-1α and the VEGF promoter, which was assessed by a chromatin immunoprecipitation (ChIP) assay, led to activation of the VEGF promoter. TSA acetylated HIF-1α at lysine (K) 674, which led to an increase in TSA-induced VEGF-HRE reporter activity. In addition, TSA-mediated cell death was reduced by the overexpression of HIF-1α but it was rescued by transfection with a HIF-1α mutant (K674R). These data demonstrate that HIF-1α may be stabilized and translocated into the nucleus for the activation of VEGF promoter by TSA-mediated acetylation at K674 under normoxic conditions. These findings suggest that HIF-1α acetylation may lead to resistance to anticancer therapeutics, such as HDAC inhibitors, including TSA.
Collapse
Affiliation(s)
- Jae-Wook Lee
- Department of Bioscience and Biotechnology, Sejong University, Seoul 05006, South Korea
| | - Dong Hee Yang
- Department of Bioscience and Biotechnology, Sejong University, Seoul 05006, South Korea
| | - Sojin Park
- Department of Bioscience and Biotechnology, Sejong University, Seoul 05006, South Korea
| | - Hae-Kyoung Han
- Department of Bioscience and Biotechnology, Sejong University, Seoul 05006, South Korea
| | - Jong-Wan Park
- Department of Pharmacology, College of Medicine, Seoul National University, Seoul 03080, South Korea
| | - Bo Yeon Kim
- World Class Institute, Anticancer Agents Research Center, Korea Research Institute of Bioscience and Biotechnology, Ochang, Cheongwon 28116, South Korea
| | - Sung Hee Um
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi-do 16419, South Korea.,Department of Health Sciences and Technology, SAIHST, Samsung Medical Center, Sungkyunkwan University, Seoul 06351, South Korea
| | - Eun-Yi Moon
- Department of Bioscience and Biotechnology, Sejong University, Seoul 05006, South Korea
| |
Collapse
|
28
|
Ganai SA, Abdullah E, Rashid R, Altaf M. Combinatorial In Silico Strategy towards Identifying Potential Hotspots during Inhibition of Structurally Identical HDAC1 and HDAC2 Enzymes for Effective Chemotherapy against Neurological Disorders. Front Mol Neurosci 2017; 10:357. [PMID: 29170627 PMCID: PMC5684606 DOI: 10.3389/fnmol.2017.00357] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 10/19/2017] [Indexed: 11/30/2022] Open
Abstract
Histone deacetylases (HDACs) regulate epigenetic gene expression programs by modulating chromatin architecture and are required for neuronal development. Dysregulation of HDACs and aberrant chromatin acetylation homeostasis have been implicated in various diseases ranging from cancer to neurodegenerative disorders. Histone deacetylase inhibitors (HDACi), the small molecules interfering HDACs have shown enhanced acetylation of the genome and are gaining great attention as potent drugs for treating cancer and neurodegeneration. HDAC2 overexpression has implications in decreasing dendrite spine density, synaptic plasticity and in triggering neurodegenerative signaling. Pharmacological intervention against HDAC2 though promising also targets neuroprotective HDAC1 due to high sequence identity (94%) with former in catalytic domain, culminating in debilitating off-target effects and creating hindrance in the defined intervention. This emphasizes the need of designing HDAC2-selective inhibitors to overcome these vicious effects and for escalating the therapeutic efficacy. Here we report a top-down combinatorial in silico approach for identifying the structural variants that are substantial for interactions against HDAC1 and HDAC2 enzymes. We used extra-precision (XP)-molecular docking, Molecular Mechanics Generalized Born Surface Area (MMGBSA) for predicting affinity of inhibitors against the HDAC1 and HDAC2 enzymes. Importantly, we employed a novel in silico strategy of coupling the state-of-the-art molecular dynamics simulation (MDS) to energetically-optimized structure based pharmacophores (e-Pharmacophores) method via MDS trajectory clustering for hypothesizing the e-Pharmacophore models. Further, we performed e-Pharmacophores based virtual screening against phase database containing millions of compounds. We validated the data by performing the molecular docking and MM-GBSA studies for the selected hits among the retrieved ones. Our studies attributed inhibitor potency to the ability of forming multiple interactions and infirm potency to least interactions. Moreover, our studies delineated that a single HDAC inhibitor portrays differential features against HDAC1 and HDAC2 enzymes. The high affinity and selective HDAC2 inhibitors retrieved through e-Pharmacophores based virtual screening will play a critical role in ameliorating neurodegenerative signaling without hampering the neuroprotective isoform (HDAC1).
Collapse
Affiliation(s)
- Shabir Ahmad Ganai
- Chromatin and Epigenetics Lab, Department of Biotechnology, University of Kashmir, Srinagar, India
| | - Ehsaan Abdullah
- Chromatin and Epigenetics Lab, Department of Biotechnology, University of Kashmir, Srinagar, India
| | - Romana Rashid
- Chromatin and Epigenetics Lab, Department of Biotechnology, University of Kashmir, Srinagar, India
| | - Mohammad Altaf
- Chromatin and Epigenetics Lab, Department of Biotechnology, University of Kashmir, Srinagar, India
| |
Collapse
|
29
|
Abstract
Glioblastoma multiforme (GBM) is the most lethal primary brain tumor in adults despite contemporary gold-standard first-line treatment strategies. This type of tumor recurs in virtually all patients and no commonly accepted standard treatment exists for the recurrent disease. Therefore, advances in all scientific and clinical aspects of GBM are urgently needed. Epigenetic mechanisms are one of the major factors contributing to the pathogenesis of cancers, including glioblastoma. Epigenetic modulators that regulate gene expression by altering the epigenome and non-histone proteins are being exploited as therapeutic drug targets. Over the last decade, numerous preclinical and clinical studies on histone deacetylase (HDAC) inhibitors have shown promising results in various cancers. This article provides an overview of the anticancer mechanisms of HDAC inhibitors and the role of HDAC isoforms in GBM. We also summarize current knowledge on HDAC inhibitors on the basis of preclinical studies and emerging clinical data.
Collapse
|
30
|
Histone Deacetylase Inhibitor-Induced Autophagy in Tumor Cells: Implications for p53. Int J Mol Sci 2017; 18:ijms18091883. [PMID: 30563957 PMCID: PMC5618532 DOI: 10.3390/ijms18091883] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 08/20/2017] [Accepted: 08/28/2017] [Indexed: 02/07/2023] Open
Abstract
Autophagy is an essential process of the eukaryotic cell allowing degradation and recycling of dysfunctional cellular components in response to either physiological or pathological changes. Inhibition of autophagy in combination with chemotherapeutic treatment has emerged as a novel approach in cancer treatment leading to cell cycle arrest, differentiation, and apoptosis. Suberoyl hydroxamic acid (SAHA) is a broad-spectrum histone deacetylase inhibitor (HDACi) suppressing family members in multiple HDAC classes. Increasing evidence indicates that SAHA and other HDACi can, in addition to mitochondria-mediated apoptosis, also promote caspase-independent autophagy. SAHA-induced mTOR inactivation as a major regulator of autophagy activating the remaining autophagic core machinery is by far the most reported pathway in several tumor models. However, the question of which upstream mechanisms regulate SAHA-induced mTOR inactivation that consequently initiate autophagy has been mainly left unexplored. To elucidate this issue, we recently initiated a study clarifying different modes of SAHA-induced cell death in two human uterine sarcoma cell lines which led to the conclusion that the tumor suppressor protein p53 could act as a molecular switch between SAHA-triggered autophagic or apoptotic cell death. In this review, we present current research evidence about HDACi-mediated apoptotic and autophagic pathways, in particular with regard to p53 and its therapeutic implications.
Collapse
|
31
|
Histone Deacetylase Inhibitors as Anticancer Drugs. Int J Mol Sci 2017; 18:ijms18071414. [PMID: 28671573 PMCID: PMC5535906 DOI: 10.3390/ijms18071414] [Citation(s) in RCA: 848] [Impact Index Per Article: 106.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 06/11/2017] [Accepted: 06/27/2017] [Indexed: 12/11/2022] Open
Abstract
Carcinogenesis cannot be explained only by genetic alterations, but also involves epigenetic processes. Modification of histones by acetylation plays a key role in epigenetic regulation of gene expression and is controlled by the balance between histone deacetylases (HDAC) and histone acetyltransferases (HAT). HDAC inhibitors induce cancer cell cycle arrest, differentiation and cell death, reduce angiogenesis and modulate immune response. Mechanisms of anticancer effects of HDAC inhibitors are not uniform; they may be different and depend on the cancer type, HDAC inhibitors, doses, etc. HDAC inhibitors seem to be promising anti-cancer drugs particularly in the combination with other anti-cancer drugs and/or radiotherapy. HDAC inhibitors vorinostat, romidepsin and belinostat have been approved for some T-cell lymphoma and panobinostat for multiple myeloma. Other HDAC inhibitors are in clinical trials for the treatment of hematological and solid malignancies. The results of such studies are promising but further larger studies are needed. Because of the reversibility of epigenetic changes during cancer development, the potency of epigenetic therapies seems to be of great importance. Here, we summarize the data on different classes of HDAC inhibitors, mechanisms of their actions and discuss novel results of preclinical and clinical studies, including the combination with other therapeutic modalities.
Collapse
|
32
|
The Effect of Histone Hyperacetylation on Viability of Basal-Like Breast Cancer Cells MDA-MB-231. RAZAVI INTERNATIONAL JOURNAL OF MEDICINE 2017. [DOI: 10.5812/rijm.55455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
33
|
Inhibiting HDAC1 Enhances the Anti-Cancer Effects of Statins through Downregulation of GGTase-Iβ Expression. Int J Mol Sci 2017; 18:ijms18051010. [PMID: 28481295 PMCID: PMC5454923 DOI: 10.3390/ijms18051010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Revised: 04/23/2017] [Accepted: 05/01/2017] [Indexed: 02/05/2023] Open
Abstract
Hydroxy-methyl-glutaryl-coenzyme A (HMG-CoA) reductase inhibitors, namely statins, are potential anti-tumor agents. Previously, we showed that a pan-histone deacetylase (HDAC) inhibitor enhances the anti-tumor effects of the HMG-CoA inhibitor. However, the underlying mechanisms were not fully understood. Cancer cell lines (CAL-27 and SACC-83) were exposed to pan-HDAC inhibitor, or HDAC1 inhibitor, or geranylgeranyl transferase type I (GGTase-I) inhibitor alone or in combination with statin. Cell viability, apoptosis, migration, and invasion were assessed by Cell Count Kit-8, 4′,6-diamidino-2-phenylindole staining, and transwell assay, respectively. A xenograft model was used for assessing tumor growth in vivo. Western blot and real-time PCR were used to assess the expression of genes. We observed that inhibiting HDAC1 could enhance the anti-tumor effects of statins both in vitro and in vivo. Inhibiting HDAC1 blocked the statin-induced upregulation of geranylgeranyl transferase type Iβ subunit (GGTase-Iβ), resulting in an enhancement of the anti-cancer effects of statin. Overexpression of GGTase-Iβ or constitutively active RhoA abolished the enhancement by inhibiting HDAC1 on anti-tumor effects of statins. The HDAC1 inhibitor failed to enhance cytotoxicity in non-tumor primary cells treated with statin. Inhibiting HDAC1 enhanced the anti-cancer effects of statins through downregulation of GGTase-Iβ expression, and thus further inactivation of RhoA. A combination of statin with HDAC1 or GGTase-I inhibitor would be a new strategy for cancer chemotherapy.
Collapse
|
34
|
Ahmad M, Aga MA, Bhat JA, Kumar B, Rouf A, Capalash N, Mintoo MJ, Kumar A, Mahajan P, Mondhe DM, Nargotra A, Sharma PR, Zargar MA, Vishwakarma RA, Shah BA, Taneja SC, Hamid A. Exploring Derivatives of Quinazoline Alkaloid l-Vasicine as Cap Groups in the Design and Biological Mechanistic Evaluation of Novel Antitumor Histone Deacetylase Inhibitors. J Med Chem 2017; 60:3484-3497. [DOI: 10.1021/acs.jmedchem.7b00322] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Mudassier Ahmad
- Cancer Pharmacology
Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu-180001, India
| | | | - Javeed Ahmad Bhat
- Cancer Pharmacology
Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu-180001, India
- Department of Biotechnology, Panjab University, Chandigarh-160014, India
| | | | | | - Neena Capalash
- Department of Biotechnology, Panjab University, Chandigarh-160014, India
| | - Mubashir Javeed Mintoo
- Cancer Pharmacology
Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu-180001, India
- CSIR-Academy of Scientific & Innovative Research, New Delhi, India
| | | | - Priya Mahajan
- Discovery Informatics Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu-180001, India
| | - Dilip Manikrao Mondhe
- Cancer Pharmacology
Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu-180001, India
- CSIR-Academy of Scientific & Innovative Research, New Delhi, India
| | - Amit Nargotra
- Discovery Informatics Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu-180001, India
| | | | - Mohmmad Afzal Zargar
- Department of Biochemistry, University of Kashmir, Srinagar, Jammu and Kashmir-190006, India
| | | | - Bhahwal Ali Shah
- CSIR-Academy of Scientific & Innovative Research, New Delhi, India
| | | | - Abid Hamid
- Cancer Pharmacology
Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu-180001, India
- CSIR-Academy of Scientific & Innovative Research, New Delhi, India
| |
Collapse
|
35
|
Ma X, Wang J, Liu J, Mo Q, Yan X, Ma D, Duan H. Targeting CD146 in combination with vorinostat for the treatment of ovarian cancer cells. Oncol Lett 2017; 13:1681-1687. [PMID: 28454309 PMCID: PMC5403387 DOI: 10.3892/ol.2017.5630] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 10/14/2016] [Indexed: 02/03/2023] Open
Abstract
Drug resistance is the predominant cause of mortality in late-stage patients with ovarian cancer. Histone deacetylase inhibitors (HDACis) have emerged as a novel type of second line drug with high specificity for tumor cells, including ovarian cancer cells. However, HDACis usually exhibit relatively low potencies when used as a single agent. The majority of current clinical trials are combination strategies. These strategies are more empirical than mechanism-based applications. Previously, it was reported that the adhesion molecule cluster of differentiation 146 (CD146) is significantly induced in HDACi-treated tumor cells. The present study additionally confirmed that the induction of CD146 is a common phenomenon in vorinostat-treated ovarian cancer cells. AA98, an anti-CD146 monoclonal antibody (mAb), was used to target CD146 function. Synergistic antitumoral effects between AA98 and vorinostat were examined in vitro and in vivo. The potential effect of combined AA98 and vorinostat treatment on the protein kinase B (Akt) pathway was determined by western blotting. The present study found that targeting of CD146 substantially enhanced vorinostat-induced killing via the suppression of activation of Akt pathways in ovarian cancer cells. AA98 in combination with vorinostat significantly inhibited cell proliferation and increased apoptosis. In vivo, AA98 synergized with vorinostat to inhibit tumor growth and prolong survival in ovarian cancer. These data suggest that an undesired induction of CD146 may serve as a protective response to offset the antitumor efficacy of vorinostat. By contrast, targeting CD146 in combination with vorinostat may be exploited as a novel strategy to more effectively kill ovarian cancer cells.
Collapse
Affiliation(s)
- Xiaoli Ma
- Gynecological Minimal Invasive Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100006, P.R. China
| | - Jiandong Wang
- Gynecological Minimal Invasive Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100006, P.R. China
| | - Jia Liu
- Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Qingqing Mo
- Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Xiyun Yan
- Center of Molecular Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, P.R. China
| | - Ding Ma
- Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Hua Duan
- Gynecological Minimal Invasive Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100006, P.R. China
| |
Collapse
|
36
|
Xu Q, Patel D, Zhang X, Veenstra RD. Changes in cardiac Nav1.5 expression, function, and acetylation by pan-histone deacetylase inhibitors. Am J Physiol Heart Circ Physiol 2016; 311:H1139-H1149. [PMID: 27638876 DOI: 10.1152/ajpheart.00156.2016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 08/24/2016] [Indexed: 12/19/2022]
Abstract
Histone deacetylase (HDAC) inhibitors are small molecule anticancer therapeutics that exhibit limiting cardiotoxicities including QT interval prolongation and life-threatening cardiac arrhythmias. Because the molecular mechanisms for HDAC inhibitor-induced cardiotoxicity are poorly understood, we performed whole cell patch voltage-clamp experiments to measure cardiac sodium currents (INa) from wild-type neonatal mouse ventricular or human-induced pluripotent stem cell-derived cardiomyocytes treated with trichostatin A (TSA), vorinostat (VOR), or romidepsin (FK228). All three pan-HDAC inhibitors dose dependently decreased peak INa density and shifted the voltage activation curve 3- to 8-mV positive. Increases in late INa were not observed despite a moderate slowing of the inactivation rate at low activating potentials (<-40 mV). Scn5a mRNA levels were not significantly altered but NaV1.5 protein levels were significantly reduced. Immunoprecipitation with anti-NaV1.5 and Western blotting with anti-acetyl-lysine antibodies indicated that NaV1.5 acetylation is increased in vivo after HDAC inhibition. FK228 inhibited total cardiac HDAC activity with two apparent IC50s of 5 nM and 1.75 μM, consistent with previous findings with TSA and VOR. FK228 also decreased ventricular gap junction conductance (gj), again consistent with previous findings. We conclude that pan-HDAC inhibition reduces cardiac INa density and NaV1.5 protein levels without affecting late INa amplitude and, thus, probably does not contribute to the reported QT interval prolongation and arrhythmias associated with pan-HDAC inhibitor therapies. Conversely, reductions in gj may enhance the occurrence of triggered activity by limiting electrotonic inhibition and, combined with reduced INa, slow myocardial conduction and increase vulnerability to reentrant arrhythmias.
Collapse
Affiliation(s)
- Qin Xu
- Department of Pharmacology, State University of New York (SUNY) Upstate Medical University, Syracuse, New York; and
| | - Dakshesh Patel
- Department of Pharmacology, State University of New York (SUNY) Upstate Medical University, Syracuse, New York; and
| | - Xian Zhang
- Department of Pharmacology, State University of New York (SUNY) Upstate Medical University, Syracuse, New York; and
| | - Richard D Veenstra
- Department of Pharmacology, State University of New York (SUNY) Upstate Medical University, Syracuse, New York; and .,Department of Cell and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York
| |
Collapse
|
37
|
An epithelial marker promoter induction screen identifies histone deacetylase inhibitors to restore epithelial differentiation and abolishes anchorage independence growth in cancers. Cell Death Discov 2016; 2:16041. [PMID: 27551531 PMCID: PMC4979427 DOI: 10.1038/cddiscovery.2016.41] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 05/07/2016] [Indexed: 12/20/2022] Open
Abstract
Epithelial–mesenchymal transition (EMT), a crucial mechanism in development, mediates aggressiveness during carcinoma progression and therapeutic refractoriness. The reversibility of EMT makes it an attractive strategy in designing novel therapeutic approaches. Therefore, drug discovery pipelines for EMT reversal are in need to discover emerging classes of compounds. Here, we outline a pre-clinical drug screening platform for EMT reversal that consists of three phases of drug discovery and validation. From the Phase 1 epithelial marker promoter induction (EpI) screen on a library consisting of compounds being approved by Food and Drug Administration (FDA), Vorinostat (SAHA), a histone deacetylase inhibitor (HDACi), is identified to exert EMT reversal effects by restoring the expression of an epithelial marker, E-cadherin. An expanded screen on 41 HDACi further identifies 28 compounds, such as class I-specific HDACi Mocetinosat, Entinostat and CI994, to restore E-cadherin and ErbB3 expressions in ovarian, pancreatic and bladder carcinoma cells. Mocetinostat is the most potent HDACi to restore epithelial differentiation with the lowest concentration required for 50% induction of epithelial promoter activity (EpIC-50).The HDACi exerts paradoxical effects on EMT transcriptional factors such as SNAI and ZEB family and the effects are context-dependent in epithelial- and mesenchymal-like cells. In vitro functional studies further show that HDACi induced significant increase in anoikis and decrease in spheroid formation in ovarian and bladder carcinoma cells with mesenchymal features. This study demonstrates a robust drug screening pipeline for the discovery of compounds capable of restoring epithelial differentiation that lead to significant functional lethality.
Collapse
|
38
|
Zhao H, Han Z, Ji X, Luo Y. Epigenetic Regulation of Oxidative Stress in Ischemic Stroke. Aging Dis 2016; 7:295-306. [PMID: 27330844 PMCID: PMC4898926 DOI: 10.14336/ad.2015.1009] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 10/09/2015] [Indexed: 12/11/2022] Open
Abstract
The prevalence and incidence of stroke rises with life expectancy. However, except for the use of recombinant tissue-type plasminogen activator, the translation of new therapies for acute stroke from animal models into humans has been relatively unsuccessful. Oxidative DNA and protein damage following stroke is typically associated with cell death. Cause-effect relationships between reactive oxygen species and epigenetic modifications have been established in aging, cancer, acute pancreatitis, and fatty liver disease. In addition, epigenetic regulatory mechanisms during stroke recovery have been reviewed, with focuses mainly on neural apoptosis, necrosis, and neuroplasticity. However, oxidative stress-induced epigenetic regulation in vascular neural networks following stroke has not been sufficiently explored. Improved understanding of the epigenetic regulatory network upon oxidative stress may provide effective antioxidant approaches for treating stroke. In this review, we summarize the epigenetic events, including DNA methylation, histone modification, and microRNAs, that result from oxidative stress following experimental stroke in animal and cell models, and the ways in which epigenetic changes and their crosstalk influence the redox state in neurons, glia, and vascular endothelial cells, helping us to understand the foregone and vicious epigenetic regulation of oxidative stress in the vascular neural network following stroke.
Collapse
Affiliation(s)
- Haiping Zhao
- 1Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Ziping Han
- 1Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Xunming Ji
- 22Department of Neurosurgery, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Yumin Luo
- 1Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; 3Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China
| |
Collapse
|
39
|
Rasmussen RD, Gajjar MK, Jensen KE, Hamerlik P. Enhanced efficacy of combined HDAC and PARP targeting in glioblastoma. Mol Oncol 2016; 10:751-63. [PMID: 26794465 DOI: 10.1016/j.molonc.2015.12.014] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 12/23/2015] [Accepted: 12/30/2015] [Indexed: 01/07/2023] Open
Abstract
Recent clinical trials have demonstrated that targeting chromatin remodeling factors is as a promising strategy for the treatment of glioblastoma (GBM). We and others have shown constitutive activation of DNA damage response (DDR) pathways in gliomas and suggested that targeting the DDR may improve the currently grim prognosis for patients. Based on our previous findings that inhibition of poly(ADP-ribose) polymerase (PARP) increases radio-sensitivity of the notoriously radio-resistant GBM cells, we hypothesized that epigenetic down-regulation of the DDR responses and induction of oxidative stress via HDAC inhibition would contribute to more efficient targeting of this deadly disease. Our data show that SAHA, an HDAC class I + II inhibitor, in combination with olaparib (PARP inhibitor): i) enhanced inhibition of GBM cell survival, ii) induced apoptosis, and iii) impaired cell cycle progression. These results provide a pre-clinical rationale for combined administration of SAHA and olaparib, which are already individually in clinical trials.
Collapse
Affiliation(s)
- Rikke D Rasmussen
- Brain Tumor Biology Group, Danish Cancer Society Research Center, Strandboulevarden 49, 2100-DK, Copenhagen, Denmark
| | - Madhavsai K Gajjar
- Brain Tumor Biology Group, Danish Cancer Society Research Center, Strandboulevarden 49, 2100-DK, Copenhagen, Denmark
| | - Kamilla E Jensen
- Brain Tumor Biology Group, Danish Cancer Society Research Center, Strandboulevarden 49, 2100-DK, Copenhagen, Denmark
| | - Petra Hamerlik
- Brain Tumor Biology Group, Danish Cancer Society Research Center, Strandboulevarden 49, 2100-DK, Copenhagen, Denmark; Department of Radiation Biology, The Finsen Center, Copenhagen University Hospital, Blegdamsvej 9, 2100-DK, Copenhagen, Denmark.
| |
Collapse
|
40
|
Ling Y, Xu C, Luo L, Cao J, Feng J, Xue Y, Zhu Q, Ju C, Li F, Zhang Y, Zhang Y, Ling X. Novel β-Carboline/Hydroxamic Acid Hybrids Targeting Both Histone Deacetylase and DNA Display High Anticancer Activity via Regulation of the p53 Signaling Pathway. J Med Chem 2015; 58:9214-27. [PMID: 26555243 DOI: 10.1021/acs.jmedchem.5b01052] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A novel series of hybrids from β-carboline and hydroxamic acid were designed and synthesized. Several compounds (5m, 11b-d, and 11h) not only exerted significant antiproliferation activity against four human colorectal cancer (CRC) cell lines but also showed histone deacetylase inhibitory effects in vitro. The most potent compound, 11c, exhibited anticancer potency sevenfold higher than that of SAHA. 11c triggered more significant cancer cell apoptosis than did SAHA by cleavage of both PARP and caspase 3 in a dose-dependent manner. Furthermore, 11c simultaneously increased the acetylation of histone H3 and α-tubulin, enhanced expression of DNA damage markers histone H2AX phosphorylation and p-p53 (Ser15), and activated p53 signaling pathway in HCT116 cells. Finally, 11c showed low acute toxicity in mice and inhibited the growth of implanted human CRC in mice more potently than did SAHA. Together, 11c possessed potent antitumor activity and may be a promising candidate for the potential treatment of human CRC.
Collapse
Affiliation(s)
- Yong Ling
- State Key Laboratory of Natural Medicines, China Pharmaceutical University , Nanjing 210009, P.R. China
| | - Chenjun Xu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University , Nanjing 210009, P.R. China
| | | | | | | | | | | | | | - Fengzhi Li
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute , Buffalo, New York, USA
| | - Yihua Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University , Nanjing 210009, P.R. China
| | | | - Xiang Ling
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute , Buffalo, New York, USA
| |
Collapse
|
41
|
Chen HP, Zhao YT, Zhao TC. Histone deacetylases and mechanisms of regulation of gene expression. Crit Rev Oncog 2015; 20:35-47. [PMID: 25746103 DOI: 10.1615/critrevoncog.2015012997] [Citation(s) in RCA: 201] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In recent years it has become widely recognized that histone modification plays a pivotal role in controlling gene expression and is involved in a wide spectrum of disease regulation. Histone acetylation is a major modification that affects gene transcription and is controlled by histone acetyltransferases (HATs) and histone deacetylases (HDACs). HATs acetylate lysines of histone proteins, resulting in the relaxation of chromatin structure, and they also facilitate gene activation. Conversely, HDACs remove acetyl groups from hyperacetylated histones and suppress general gene transcription. In addition to histones, numerous nonhistone proteins can be acetylated and deacetylated, and they also are involved in the regulation of a wide range of diseases. To date there are 18 HDACs in mammals classified into 4 classes based on homology to yeast HDACs. Accumulating evidence has revealed that HDACs play crucial roles in a variety of biological processes including inflammation, cell proliferation, apoptosis, and carcinogenesis. In this review we summarize the current state of knowledge of HDACs in carcinogenesis and describe the involvement of HDACs in cancer-associated molecular processes. It is hoped than an understanding of the role of HDACs in cancer will lead to the design of more potent and specific drugs targeting selective HDAC proteins for the treatment of the disease.
Collapse
Affiliation(s)
- Hong Ping Chen
- Department of Surgery, Boston University Medical School, Boston University, Roger Williams Medical Center, Providence, RI; Department of Histology and Embryology, Medical College, Nanchang University, Nanchang, China
| | - Yu Tina Zhao
- Department of Surgery, Boston University Medical School, Boston University, Roger Williams Medical Center, Providence, RI
| | - Ting C Zhao
- Department of Surgery, Boston University Medical School, Boston University, Roger Williams Medical Center, Providence, RI
| |
Collapse
|
42
|
Chao MW, Lai MJ, Liou JP, Chang YL, Wang JC, Pan SL, Teng CM. The synergic effect of vincristine and vorinostat in leukemia in vitro and in vivo. J Hematol Oncol 2015; 8:82. [PMID: 26156322 PMCID: PMC4504084 DOI: 10.1186/s13045-015-0176-7] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 06/17/2015] [Indexed: 02/08/2023] Open
Abstract
Background Combination therapy is a key strategy for minimizing drug resistance, a common problem in cancer therapy. The microtubule-depolymerizing agent vincristine is widely used in the treatment of acute leukemia. In order to decrease toxicity and chemoresistance of vincristine, this study will investigate the effects of combination vincristine and vorinostat (suberoylanilide hydroxamic acid (SAHA)), a pan-histone deacetylase inhibitor, on human acute T cell lymphoblastic leukemia cells. Methods Cell viability experiments were determined by 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) assay, and cell cycle distributions as well as mitochondria membrane potential were analyzed by flow cytometry. In vitro tubulin polymerization assay was used to test tubulin assembly, and immunofluorescence analysis was performed to detect microtubule distribution and morphology. In vivo effect of the combination was evaluated by a MOLT-4 xenograft model. Statistical analysis was assessed by Bonferroni’s t test. Results Cell viability showed that the combination of vincristine and SAHA exhibited greater cytotoxicity with an IC50 value of 0.88 nM, compared to each drug alone, 3.3 and 840 nM. This combination synergically induced G2/M arrest, followed by an increase in cell number at the sub-G1 phase and caspase activation. Moreover, the results of vincristine combined with an HDAC6 inhibitor (tubastatin A), which acetylated α-tubulin, were consistent with the effects of vincristine/SAHA co-treatment, thus suggesting that SAHA may alter microtubule dynamics through HDAC6 inhibition. Conclusion These findings indicate that the combination of vincristine and SAHA on T cell leukemic cells resulted in a change in microtubule dynamics contributing to M phase arrest followed by induction of the apoptotic pathway. These data suggest that the combination effect of vincristine/SAHA could have an important preclinical basis for future clinical trial testing. Electronic supplementary material The online version of this article (doi:10.1186/s13045-015-0176-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Min-Wu Chao
- Pharmacological Institute, College of Medicine, National Taiwan University, No. 1, Jen-Ai Road, Sec. 1, Taipei, Taiwan
| | - Mei-Jung Lai
- Center for Translational Medicine, Taipei Medical University, No. 250, Wu-hsing Street, Taipei, 11031, Taiwan
| | - Jing-Ping Liou
- School of Pharmacy, College of Pharmacy, Taipei Medical University, No. 250, Wu-hsing Street, Taipei, 11031, Taiwan
| | - Ya-Ling Chang
- Pharmacological Institute, College of Medicine, National Taiwan University, No. 1, Jen-Ai Road, Sec. 1, Taipei, Taiwan
| | - Jing-Chi Wang
- The Ph.D. Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, No. 250, Wu-hsing St., Taipei, 11031, Taiwan
| | - Shiow-Lin Pan
- The Ph.D. Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, No. 250, Wu-hsing St., Taipei, 11031, Taiwan.
| | - Che-Ming Teng
- Pharmacological Institute, College of Medicine, National Taiwan University, No. 1, Jen-Ai Road, Sec. 1, Taipei, Taiwan.
| |
Collapse
|
43
|
Histone Deacetylases Inhibitors in the Treatment of Retinal Degenerative Diseases: Overview and Perspectives. J Ophthalmol 2015; 2015:250812. [PMID: 26137316 PMCID: PMC4468288 DOI: 10.1155/2015/250812] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 09/09/2014] [Indexed: 01/08/2023] Open
Abstract
Retinal degenerative diseases are one of the important refractory ophthalmic diseases, featured with apoptosis of photoreceptor cells. Histone acetylation and deacetylation can regulate chromosome assembly, gene transcription, and posttranslational modification, which are regulated by histone acetyltransferases (HATs) and histone deacetylases (HDACs), respectively. The histone deacetylase inhibitors (HDACis) have the ability to cause hyperacetylation of histone and nonhistone proteins, resulting in a variety of effects on cell proliferation, differentiation, anti-inflammation, and anti-apoptosis. Several HDACis have been approved for clinical trials to treat cancer. Studies have shown that HDACis have neuroprotective effects in nervous system damage. In this paper, we will summarize the neuroprotective effects of common HDACis in retinal degenerative diseases and make a prospect to the applications of HDACis in the treatment of retinal degenerative diseases in the future.
Collapse
|
44
|
Khaopha S, Jogloy S, Patanothai A, Senawong T. Histone Deacetylase Inhibitory Activity of Peanut Testa Extracts against Human Cancer Cell Lines. J Food Biochem 2015. [DOI: 10.1111/jfbc.12128] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Somprasong Khaopha
- Department of Biochemistry; Faculty of Science; Khon Kaen University; Khon Kaen 40002 Thailand
| | - Sanun Jogloy
- Department of Plant Science and Agricultural Resources; Faculty of Agriculture; Khon Kaen University; Khon Kaen 40002 Thailand
| | - Aran Patanothai
- Department of Plant Science and Agricultural Resources; Faculty of Agriculture; Khon Kaen University; Khon Kaen 40002 Thailand
| | - Thanaset Senawong
- Department of Biochemistry; Faculty of Science; Khon Kaen University; Khon Kaen 40002 Thailand
- Natural Products Research Unit; Khon Kaen University; Khon Kaen 40002 Thailand
- Food and Products Chemical Analysis Research Group; Faculty of Science; Khon Kaen University; Khon Kaen 40002 Thailand
| |
Collapse
|
45
|
Zhou J, Li M, Chen N, Wang S, Luo HB, Zhang Y, Wu R. Computational design of a time-dependent histone deacetylase 2 selective inhibitor. ACS Chem Biol 2015; 10:687-92. [PMID: 25546141 PMCID: PMC4372102 DOI: 10.1021/cb500767c] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
![]()
Development of isoform-selective
histone deacetylase (HDAC) inhibitors is of great biological and medical
interest. Among 11 zinc-dependent HDAC isoforms, it is particularly
challenging to achieve isoform inhibition selectivity between HDAC1
and HDAC2 due to their very high structural similarities. In this
work, by developing and applying a novel de novo reaction-mechanism-based
inhibitor design strategy to exploit the reactivity difference, we
have discovered the first HDAC2-selective inhibitor, β-hydroxymethyl
chalcone. Our bioassay experiments show that this new compound has
a unique time-dependent selective inhibition on HDAC2, leading to
about 20-fold isoform-selectivity against HDAC1. Furthermore, our
ab initio QM/MM molecular dynamics simulations, a state-of-the-art
approach to study reactions in biological systems, have elucidated
how the β-hydroxymethyl chalcone can achieve the distinct time-dependent
inhibition toward HDAC2.
Collapse
Affiliation(s)
- Jingwei Zhou
- School of Pharmaceutical
Sciences, Sun Yat-sen University, Guangzhou 510006, P.R. China
| | - Min Li
- School of Pharmaceutical
Sciences, Sun Yat-sen University, Guangzhou 510006, P.R. China
| | - Nanhao Chen
- School of Pharmaceutical
Sciences, Sun Yat-sen University, Guangzhou 510006, P.R. China
| | - Shenglong Wang
- Department of Chemistry, New York University, New York, New York 10003, United States
| | - Hai-Bin Luo
- School of Pharmaceutical
Sciences, Sun Yat-sen University, Guangzhou 510006, P.R. China
| | - Yingkai Zhang
- Department of Chemistry, New York University, New York, New York 10003, United States
- NYU-ECNU Center for Computational Chemistry at NYU Shanghai, Shanghai 200062, P.R. China
| | - Ruibo Wu
- School of Pharmaceutical
Sciences, Sun Yat-sen University, Guangzhou 510006, P.R. China
| |
Collapse
|
46
|
Chessum N, Jones K, Pasqua E, Tucker M. Recent advances in cancer therapeutics. PROGRESS IN MEDICINAL CHEMISTRY 2015; 54:1-63. [PMID: 25727702 DOI: 10.1016/bs.pmch.2014.11.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In the past 20 years, cancer therapeutics has undergone a paradigm shift away from the traditional cytotoxic drugs towards the targeting of proteins intimately involved in driving the cancer phenotype. The poster child for this alternative approach to the treatment of cancer is imatinib, a small-molecule kinase inhibitor designed to target chronic myeloid leukaemia driven by the BCR-ABL translocation in a defined patient population. The improvement in survival achieved by treatment of this patient cohort with imatinib is impressive. Thus, the aim is to provide efficacy but with low toxicity. The role of the medicinal chemist in oncology drug discovery is now closely aligned with the role in most other therapeutic areas with high-throughput and/or fragment-based screening, structure-based design, selectivity, pharmacokinetic optimisation and pharmacodynamic biomarker modulation, all playing a familiar part in the process. In this chapter, we selected four areas in which compounds are either approved drugs or in clinical trials. These are chaperone inhibitors, kinase inhibitors, histone deacetylase inhibitors and inhibitors of protein-protein interactions. Even within these areas, we have been selective, particularly for kinase inhibitors, and our aim has been to exemplify newer approaches and novel aspects of medicinal chemistry.
Collapse
Affiliation(s)
- Nicola Chessum
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Keith Jones
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Elisa Pasqua
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Michael Tucker
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| |
Collapse
|
47
|
Vandekerckhove S, D'hooghe M. Quinoline-based antimalarial hybrid compounds. Bioorg Med Chem 2014; 23:5098-119. [PMID: 25593097 DOI: 10.1016/j.bmc.2014.12.018] [Citation(s) in RCA: 150] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 12/03/2014] [Accepted: 12/11/2014] [Indexed: 10/24/2022]
Abstract
Quinoline-containing compounds, such as quinine and chloroquine, have a long-standing history as potent antimalarial agents. However, the increasing resistance of the Plasmodium parasite against these drugs and the lack of licensed malaria vaccines have forced chemists to develop synthetic strategies toward novel biologically active molecules. A strategy that has attracted considerable attention in current medicinal chemistry is based on the conjugation of two biologically active molecules into one hybrid compound. Since quinolines are considered to be privileged antimalarial building blocks, the synthesis of quinoline-containing antimalarial hybrids has been elaborated extensively in recent years. This review provides a literature overview of antimalarial hybrid molecules containing a quinoline core, covering publications between 2009 and 2014.
Collapse
Affiliation(s)
- Stéphanie Vandekerckhove
- SynBioC Research Group, Department of Sustainable Organic Chemistry and Technology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, B-9000 Ghent, Belgium
| | - Matthias D'hooghe
- SynBioC Research Group, Department of Sustainable Organic Chemistry and Technology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, B-9000 Ghent, Belgium.
| |
Collapse
|
48
|
Ma X, Duan H, Liu J, Mo Q, Sun C, Ma D, Wang J. Effect of LIV1 on the sensitivity of ovarian cancer cells to trichostatin A. Oncol Rep 2014; 33:893-8. [PMID: 25420545 DOI: 10.3892/or.2014.3622] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2014] [Accepted: 10/31/2014] [Indexed: 11/05/2022] Open
Abstract
In a previous study, we used a functional gene screen approach to identify the key genes responsible for the tumor-selective action of trichostatin A (TSA), of which LIV1, a novel zinc transporter, was isolated by its marked ability to confer resistance against TSA-induced apoptosis. The aim of the present study was to investigate the effect of LIV1 expression on the sensitivity of ovarian cancer cells to TSA. We tested the induction of LIV1 in ovarian cancer cells and clinical samples after TSA treatment by real-time PCR and western blot analysis. We investigated the effect of LIV1 expression on the sensitivity of ovarian cancer cells to TSA by MTT assay, flow cytometry and colony forming assays. Finally, we analyzed the mechanism of LIV1 in ovarian cancer cells by western blot analysis. We found that the levels of LIV1 mRNA and protein were significantly upregulated after TSA treatment. The viability and colony forming rates of the ovarian cancer cells transfected with AS-LIV1 (pCEP4 carrying antisense LIV1 cDNA) were obviously higher than the rates of the control as detected by MTT and colony forming assays, which could be reversed by FL-LIV1 (pCEP4 carrying full-length LIV1 cDNA). The apoptotic rate of the AS-LIV1 cells was markedly lower than the rate of the control as determined FACS. Using western blot analysis, we demostrated that the inhibition of TSA-induced apoptosis by knockdown of LIV1 might be associated with decreased endogenous levels of Bcl-2, enhanced levels of Bax and cleavage of procaspase-3. The present study suggests that the drug resistance of ovarian cancer cells to TSA may be related to expression of the LIV1 gene, and targeting LIV1 could be exploited as a novel strategy to more effectively kill ovarian cancer cells.
Collapse
Affiliation(s)
- Xiaoli Ma
- Gynecological Minimal Invasive Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100006, P.R. China
| | - Hua Duan
- Gynecological Minimal Invasive Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100006, P.R. China
| | - Jia Liu
- Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Qingqing Mo
- Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Chengjuan Sun
- Gynecological Minimal Invasive Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100006, P.R. China
| | - Ding Ma
- Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Jiandong Wang
- Gynecological Minimal Invasive Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100006, P.R. China
| |
Collapse
|
49
|
Hao ZF, Su YM, Wang CM, Yang RY. Activating transcription factor 3 interferes with p21 activation in histone deacetylase inhibitor-induced growth inhibition of epidermoid carcinoma cells. Tumour Biol 2014; 36:1471-6. [PMID: 25371069 DOI: 10.1007/s13277-014-2618-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 09/09/2014] [Indexed: 12/31/2022] Open
Abstract
Inhibition of histone deacetylase (HDAC) activity by HDAC inhibitors (HDACis) results in cancer cell growth inhibition, and HDACis have been revealed as potential anti-skin cancer agents. p21 is a cyclin-dependent kinase inhibitor and an essential regulator of growth inhibition. Recently, we reported that activating transcription factor 3 (ATF3) could significantly promote skin cancer cell growth. This study explored the relationship between ATF3 and HDACi-induced growth inhibition of epidermoid carcinoma cells. We found that trichostatin A (TSA) treatment inhibited cell growth in A431 epidermoid carcinoma cells in a dose-dependent manner. Simultaneously, p21 and ATF3 expression levels were upregulated and downregulated upon TSA stimulation, respectively. ATF3 overexpression promoted cell growth and downregulated p21 expression. In contrast, ATF3 depletion resulted in cell growth reduction and p21 transcriptional upregulation. More importantly, ATF3 overexpression partially antagonized TSA-induced growth inhibition and p21 activation. Collectively, these data demonstrate that ATF3 acts as an essential negative regulator of TSA-induced cell growth inhibition through interfering with TSA-induced p21 activation.
Collapse
Affiliation(s)
- Zhen-Feng Hao
- Institute of Skin Damage and Repair, General Hospital of Beijing Region of PLA, Beijing, 100700, China
| | | | | | | |
Collapse
|
50
|
Zohre S, Kazem NK, Abolfazl A, Mohammad RY, Aliakbar M, Effat A, Zahra D, Hassan D, Nosratollah Z. Trichostatin A-induced Apoptosis is Mediated by Krüppel-like Factor 4 in Ovarian and Lung Cancer. Asian Pac J Cancer Prev 2014; 15:6581-6. [DOI: 10.7314/apjcp.2014.15.16.6581] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|