Histone Deacetylase Inhibitors: Clinical Implications For Hematological .

1y ago
5 Views
3 Downloads
573.90 KB
20 Pages
Last View : 1m ago
Last Download : 3m ago
Upload by : Jerry Bolanos
Transcription

Clin Epigenet (2010) 1:25–44DOI 10.1007/s13148-010-0006-2REVIEWHistone deacetylase inhibitors: clinical implicationsfor hematological malignanciesFrancesco Paolo Tambaro & Carmela Dell’Aversana &Vincenzo Carafa & Angela Nebbioso & Branka Radic &Felicetto Ferrara & Lucia AltucciReceived: 9 April 2010 / Accepted: 12 July 2010 / Published online: 28 July 2010# Springer-Verlag 2010Abstract Histone modifications have widely been implicated in cancer development and progression and arepotentially reversible by drug treatments. The N-terminaltails of each histone extend outward through the DNAstrand containing amino acid residues modified by posttranslational acetylation, methylation, and phosphorylation.These modifications change the secondary structure of thehistone protein tails in relation to the DNA strands,increasing the distance between DNA and histones, andthus allowing accessibility of transcription factors to genepromoter regions. A large number of HDAC inhibitors havebeen synthesized in the last few years, most being effectivein vitro, inducing cancer cells differentiation or cell death.The majority of the inhibitors are in clinical trials, unlikethe suberoylanilide hydroxamic acid, a pan-HDACi, andRomidepsin (FK 228), a class I-selective HDACi, whichF. P. Tambaro : C. Dell’Aversana : V. Carafa : A. Nebbioso :B. Radic : L. AltucciDipartimento di Patologia generale,Seconda università degli Studi di Napoli,Vico L. De Crecchio 7,80138 Naples, ItalyL. Altucci (*)CNR-IGB, via P. Castellino,Naples, Italye-mail: lucia.altucci@unina2.itF. FerraraEmatologia con Trapianto di Cellule Staminali,Ospedale Cardarelli,via Cardarelli 9,80131 Naples, ItalyC. Dell’AversanaUniversità di Messina,Messina, Italyare only approved in the second line treatment of refractory,persistent or relapsed cutaneous T-cell lymphoma, andactive in approximately 150 clinical trials, in monotherapyor in association. Preclinical studies investigated the use ofthese drugs in clinical practice, as single agents and incombination with chemotherapy, hypomethylating agents,proteasome inhibitors, and MTOR inhibitors, showing asignificant effect mostly in hematological malignancies.The aim of this review is to focus on the biological featuresof these drugs, analyzing the possible mechanism(s) ofaction and outline an overview on the current use in theclinical practice.Keywords HDAC inhibitors . Hematologicalmalignancies . Clinical trialsHistones: structure and functionsHistones are alkaline proteins found in eukaryotic cellnuclei, which package and order the DNA into structuralunits called nucleosomes. They can be grouped into fivemajor classes: H1/H5, H2A, H2B, H3, and H4. These areorganized into two super-classes: core histones, H2A, H2B,H3, and H4 and linker histones, H1 and H5. Two corehistones assemble to form one octameric nucleosome coreparticle by wrapping DNA around the protein spool in1.65 Å left-handed super-helical coil. The linker histone H1binds the nucleosome and the entry and exit sites of theDNA, thus locking the DNA into place and allowing theformation of higher order structure. A typical nucleosome iscomposed of an octamer of the four pairs of core histonesH2A, H2B, H3, and H4 and 146 base pairs of DNAwrapped around. Histones undergo posttranslational modifications altering their interaction with DNA and nuclear

26proteins and which remodel the chromatin condensationstatus and gene expression.Epigenetic modifications of the histone proteinsThe H3 and H4 histones have long tails protruding from thenucleosome, which can be covalently modified. The core ofH2A and H3 is susceptible as well. These modificationsinclude methylation, phosphorylation, sumoylation, citrullination, acetylation, and ubiquitination. Combinations ofmodifications are thought to constitute the so-called“histone code.” Diverse biological processes such as generegulation, DNA repair, and chromosome condensationseem to be regulated by this code.MethylationThe addition of one, two, or three methyl groups is generallyassociated with transcriptional repression. However, methylation of some lysine and arginine by residues of histones,results in transcriptional activation. Methylation is operatedby histone methyltransferases enzymes, histone-lysine Nmethyltransferase and histone-arginine N-methyltransferase:both catalyze the transfer of one to three methyl groups fromthe co-factor S-adenosyl methionine to lysine and arginineresidues of histone proteins. Methylation of histone 3 (H3) atlysine 4 (K4) is also associated with transcriptional activation,whereas methylation of H3 at K9 or 27 and of H4 at K20 isassociated with transcriptional repression (Cameron et al.1999; Esteller 2008; Kondo et al. 2008).Clin Epigenet (2010) 1:25–44Serine 139 has been identified as the site for thismodification, and its phosphorylation in response todamage is dependent on the phosphatidylinositol-3-OHkinase, and again serine 10 appears to play a key role(Cheung et al. 2000). Mec1, in yeast, Mec1-dependentserine 139 phosphorylation is apparently required forefficient non-homologous end-joining repair of DNA(Rogakou et al. 2000). This suggests that phosphorylationmediates an alteration of chromatin structure, which in turnfacilitates repair.SumoylationHistone H4 is modified by small ubiquitin-related modifier(SUMO) family proteins both in vivo and in vitro. H4binds to the SUMO-conjugating enzyme and mediates genesilencing through recruitment of histone deacetylase andheterochromatin proteins (Shiio and Eisenman 2003).CitrullinationPeptidylarginine deiminase (PAD) enzymes catalyze theconversion of arginine residues in proteins to citrullineresidues. Citrulline is a non-standard amino acid that is notincorporated into proteins during translation, but can begenerated posttranslationally by the PAD enzymes. Oneprotein isozyme responsible for this modification, proteinarginine deiminase 4 (PAD4), has also been proposed to“reverse” epigenetic histone modifications made by theprotein arginine methyltransferases (Denis et al. 2009).PhosphorylationAcetylationRelatively little is known about the enzymes that generatehistone modifications. Phosphorylation of serine 10 inhistone H3 has been shown to correlate with geneactivation in mammalian cells (Nowak and Corces 2000).The mechanism by which phosphorylation contributes totranscriptional activation is not well understood. Theaddition of negatively charged phosphate groups to histonetails neutralizes their basic charge reducing their affinity forDNA. Furthermore, it has been found that several acetyltransferases have increased HAT activity on serine 10phosphorylated substrates. Thus, phosphorylation maycontribute to transcriptional activation through the stimulationof HAT activity on the same histone tail. Phosphorylationof histone H3 is also known to occur after activation ofDNA damage signaling pathways. For example, aconserved motif found in the carboxyl terminus of yeastH2A and the mammalian H2A variant H2AX is rapidlyphosphorylated upon exposure to DNA-damaging agents(Rogakou et al. 1999).The core histone N-terminal domains are rich in positivelycharged basic amino acids, which can actively interact withDNA. Acetylation neutralizes the positive charges onhistones and disrupts the electrostatic interactions betweenDNA and histone proteins, promoting chromatin unfoldingwhich has been associated with gene expression (Gregoryet al. 2001). Under physiological conditions, chromatinacetylation is regulated by the balanced action of histoneacetyltransferases (HATs) and deacetylases (HDACs). TheHATs transfer acetyl groups from acetyl coenzyme Aonto the amino groups of conserved lysine residueswithin the core histones. Acetylation can neutralize thepositive charge of histones, loosening their interactionswith the negatively charged DNA backbone, and leadingto a more “open” active chromatin structure favoring thebinding of transcription factors for active gene transcription (Gregory et al. 2001).In contrast, the re-establishment of the positive charge inthe amino terminal tails of core histones catalyzed by

Clin Epigenet (2010) 1:25–44HDACs is thought to tighten the interaction betweenhistones and DNA, blocking the binding sites on promoter,thus inhibiting gene transcription. Obviously, a subtlyorchestrated balance between the actions of HATs andHDACs is essential to the maintenance of normal cellularfunctions, shifting this balance in both senses might havedramatic consequences on the cell phenotypes such ascarcinogenesis.UbiquitinationUbiquitination represents another important histone modification (Jason et al. 2002). Histone H2A, the first proteinidentified to be ubiquitinated at the highly conserved residue,Lys 119 (Nickel and Davie 1989), can be monoubiquitinated,in the majority of the cases and polyubiquitinated, lessfrequently, in many tissues and cell types (Nickel and Davie1989). In addition to H2A, H2B is also ubiquitinated (Westand Bonner 1980). Only monoubiquitinated H2B has beenreported, but like H2A, the ubiquitinated site has also beenmapped to Lys 120 residue located at the C terminus of H2Bin human. In addition to H2A and H2B, ubiquitination onH3 and H1 have also been reported (Chen et al. 1998; Phamand Sauer 2000).Addition of a ubiquitin moiety to a protein involves thesequential action of E1, E2, and E3 enzymes. Removingthe ubiquitin moiety, on the other hand, is achieved throughthe action of enzymes called isopeptidases (Wilkinson 2000).Accumulating evidence indicates that ubiquitin plays animportant role in regulating transcription either throughproteasome-dependent destruction of transcription factors orproteasome-independent mechanisms (Conaway et al. 2002).Several studies suggest that not only histone ubiquitination(Krogan et al. 2003), but also deubiquitination (Henry et al.2003), may participate in gene activation, linked to histoneacetylation and methylation (Dover et al. 2002).Histone deacetylasesIn the 1970s, the Friend erythroleukemia cell line wasfound to differentiate in the presence of dimethyl sulfoxideor butyrate (Sato et al. 1971). Many compounds with theability to promote differentiation of tumor cell lines,particularly those with a planar-polar configuration, inducedaccumulation of hyperacetylated histones.This histone modification increased the spatial separation of DNA from histone and enhanced binding oftranscription factor complexes to DNA (Lee et al. 1993).The first mammalian HDACs were cloned on the basis oftheir binding to known small molecule inhibitors of histonedeacetylation. These genes were homologous to yeast27transcriptional repressors, strengthening the evidence thathistone deacetylation suppresses gene expression.HDACs are a class of enzymes that remove acetyl groupsfrom a ε-N-acetyl lysine amino acid on a histone. Contrarilyto HAT, HDAC proteins are now also being referred to aslysine deacetylases (KDAC), as to more precisely describetheir function rather than their target, which also includesnumerous non-histone proteins. Eighteen human HDACshave been identified each with varying function, localization,and substrates (Table 2; Lane and Chabner 2009).Classes of HDACsHDACs can be divided in four different classes, based onthe homologies between human and yeast reduced potassiumdependency 3 (RPD3) enzymes.Class I HDACs (HDACs 1, 2, 3, and 8) are related tothe yeast RPD3 deacetylase and are primarily found inthe nucleus with the exception of HDAC 3, which can belocated both in the nucleus and the cytoplasm.Class II HDACs are divided into two subclasses, classIIa (HDACs 4, 5, 7, and 9) and class IIb (HDACs 6and 10), both homologous to the yeast Hda1 deacetylase.This class of HDACs is able to shuttle in and out of thenucleus responding to different signals.Class III HDACs consists of seven HDACs (SIRT1 toSIRT7) and shows several homologies with the yeast silentinformation regulator 2. This class of HDACs has a uniquecatalytic mechanism requiring the co-factor NAD foractivity.Class IV of HDACs has only one member (Butler andKozikowski 2008), HDAC 11, which shares similarities toboth class I and class II HDACs.Classes I, II, and IV require Zn2 for activity.Catalytic site and deacetylation mechanismHistone proteins catalytically remove an acetyl group froma ε-N-acetyl lysine amino acid on a histone. The active siteof HDACs consists of a cylindrical pocket, covered byhydrophobic and aromatic amino acids, where the lysineresidue fits when deacetylation takes place; a zinc ion islocated near the bottom of the cylindrical pocket, which iscoordinated by amino acids and a single water molecule(Butler and Kozikowski 2008). During deacetylation, thewater molecule acts as the nucleophile attacking thecarbonyl, assisted by the zinc ion (Fig. 1). Prior to attackingthe carbonyl group of the N-acetylated lysine, the watermolecule is activated by an Asp–His charge relay system.The residues forming the cylindrical pocket and theadjacent cavity of classes I and II HDACs are highlyconserved, although the residues lining the entrance of the

28Fig. 1 Water molecule acting as the nucleophile prior and during theattack of carbonyl, assisted by the zinc ionClin Epigenet (2010) 1:25–44gastric, colon, pancreas, breast, ovary, and thyroid cancers(Nakagawa et al. 2007).High level class I HDAC expression was found in 75%esophageal, gastric, colon, and prostate cancers, as well ascorresponding adjacent “normal” tissues. Esophageal andprostate cancers tended to exhibit more consistent overexpression of class I HDACs. Further studies suggest acorrelation between advanced stage of lung carcinoma andhigh expression of class 1 HDAC 1 (Sasaki et al. 2004), aswell as aggressive tumor histology, advanced stage ofdisease, and poor prognosis in patients with pancreaticcarcinoma (Miyake et al. 2008).Several retrospective studies, conducted on colon cancerprimary cells, showed high level expression of HDAC 1and 2 in gastric cancer; the higher expression correlatedwith nodes metastasis, undifferentiated histology, and poorprognosis (Weichert et al. 2008). High level expression ofHDAC 1, 2, and 3 was observed in 70%, 74%, and 95% of192 prostate cancers in a related study, where overexpression of HDAC 1 and/or HDAC 2 correlated with poorlydifferentiated tumors. Co-expression of elevated levels ofthe three HDACs was associated to an increased proliferation index, while overexpression of HDAC 2 alone was anindependent index of poor prognosis.Overexpression of HDAC 2 also correlates with advancedstage of disease and diminished survival of oropharyngealcarcinoma patients (Chang et al. 2009). In contrast, HDAC 1expression in breast cancer is associated with estrogenreceptor/progesterone (PR) expression at the earlier stage ofdisease (T as well as N classifications) and improved patientsurvival response to tamoxifen (Bicaku et al. 2008), andHDAC 6 expression is associated to improved patientsurvival (Zhang et al. 2004).HDAC expression in hematological malignanciespocket are not as conserved as the residues inside thepocket. Therefore, it is possible to design HDAC inhibitors(HDACis) that are isoform selective.HDAC expression in cancer tissuesEvidence has shown an implication of HDACs in cancer, asthe high expression of HDAC isoenzymes correspond tohypoacetylation of histones in cancer cells compared tonormal tissues (Nakata et al. 2004; Yoo and Jones 2006):HDAC 1 enzyme expression is higher in colon adenocarcinoma cells than in normal, while HDAC 2 expression iselevated in colon cancer, possibly as a result of adenomatosis poliposis coli gene deficiency.A study conducted by Nakagawa showed a hyperexpression of HDACs in a variety of cultured cancer linesand a broad panel of primary human lung, esophageal,In hematological malignancies, increased HDAC activitymay lead to transcriptional repression of genes essential forhematopoietic differentiation, playing a critical role in thepathogenesis of several leukemias. In the case of acutepromyelocytic leukemia (APL), the PML-RARα proteinproduct of the t(15–17) translocation, and in core bindingfactor leukemia gene products AML1–ETO, product of thet(8–21) translocation and CBF–MYH 11, cause a transcriptional repression, aberrantly recruiting HDACs to genespromoter regions (Minucci and Pelicci 1999); (Minucciet al. 2001). CTCL cells undergo higher rates of apoptosisthan normal lymphocytes in response to HDAC inhibitortreatment (Zhang et al. 2005). Sodium butyrate andtricostatin A induced apoptosis of acute myeloid leukemia(AML; HL60) and CML (K562) cell line, downregulatingthe expression of Daxx, without affecting the expression ofBcl-2 or Bcl-XL. A recent study reports anti-leukemic

Clin Epigenet (2010) 1:25–4429activity of valproic acid through induction of apoptosis, inCLL primary cells obtained from 14 patients (Stamatopouloset al. 2009).These findings supported drug development of HDACithat interacting with the catalytic region of HDACs, blockHDAC activity, induce differentiation, cell growth arrest, orcell death in tumors (Mai et al. 2005; Nebbioso et al. 2005).Histone deacetylase inhibitorsHDACis are compounds that are able to interact with thecatalytic domain of histone deacetylases to block the substraterecognition ability of these enzymes, thus resulting inrestoration of relevant gene expression (Finnin et al. 1999).The vast majority of HDACis shares a commonmechanism of action that consists with binding the catalyticdomain of HDAC enzyme, thereby blocking substraterecognition and inducing gene expression. Most of thedescribed HDACis primarily affect class I and class IIHDACs, which are zinc dependent.HDACis can be divided into four classes based ondifferent chemical properties: short-chain fatty acids,hydroxamic acids, cyclic peptides, and benzamides (Table 1).Short-chain fatty acidsThis group includes Na butyrate, 4-phenylbutyrate, valproicacid, and phenyl acetate. The mechanism of action has notbeen well clarified yet, although a strong hypothesis existsin which the carboxylic function acts as zinc-binding groupor competes with the acetate released in the deacetylationreaction by occupying the acetate escaping tunnel asdescribed by Wang (Mai and Altucci 2009).The butyrates (Na butyrate and 4 phenylbutyrate) inhibitthe growth of several cancers, such as colon prostate andendometrial, but just at high concentration; although theyboth show effects on phosphorylation and methylation ofhistone and other nuclear proteins.Sodium valproate (valproic acid, VPA), an “old” drugused in neurology as anticonvulsive and mood stabilizing,has been incidentally identified as HDACi and has shownanticancer effects in carcinoma cells, differentiation inhematopoietic cell lines, and also in clinical trials forhematological malignancies, such as leukemia, myelodysplastic syndrome (MDS), and lymphoma. VPA inhibitsclass I/IIa HDACs at very low concentrations compared tobutyrates (Mai and Altucci 2009).Hydroxamic acidsThis class includes the majority of HDACis presently in usein clinical trials in hematological malignancies. They have acommon structure characterized by a hydrophobic CAPgroup, able to interact with the rim of the catalytic tunnel ofthe enzyme, a polar connection unit (CU). Present in mostof the HDACis, the CU can interact with the amino acids inthe tunnel, and a 4- or 6-carbon unit hydrophobic spacer(linker), allowing the following zinc-binding group (ZBG)Table 1 Histone deacetylasesClass HDACHDACs yeastHDACs mammalianLengthMechanism of deacetylationTissue expressionClass IRPD3UbiquitousHDA1Zn2 dependentRestrictedClass IIISIR2HST1HST2HST3HST4NAD dependentNDClass 14310355400347Zn2 dependentClass C10SIRT1SIRT2SIRT3SIRT4SIRT5SIRT6SIRT7HDAC11Zn2 dependentUbiquitous

30to reach and to complex the zinc ion and thus inhibiting theenzyme (Mai et al. 2005).Past reports of the hydroxamic acid tricostatin A, firstisolated as antifungal antibiotic (Tsuji et al. 1976), indicatedthe capacity of this drug to differentiate the Frienderythroleukemia cells. Further experiments have shownthat the compound caused hyperacetylation due to hystonedeacetylation inhibition.The hybrid polar compounds (HPCs) are potent inducersof differentiation of murine erythroleukemia cells and avarious other cancer cells (Andreeff et al. 1992; Mai et al.2005). The progenitor of these compounds was hexamethylene bisacetamide, a drug that is able to induceremission in patients with myelodysplastic syndrome andacute myeloid leukemia, but cannot be used in clinical trialsfor the high dosage required and adverse side effects. Thesecond generation of HPCs has shown a strong induction ofapoptosis or cell differentiation at low doses (Mai andAltucci 2009).The prototype of this class is the suberoylanilidehydroxamic acid (SAHA or Vorinostat). The chemicalstructure of SAHA is similar to VPA and the others, HPCs,with a CAP, a CU, and a ZBG. It has shown to induceacetylation in a vast variety of cell line and apoptosis, cellcycle arrest, and differentiation. SAHA is selective forHDAC 1, 2, 3, 4, 6, 7, and 9 and shows lower potencyagainst HDAC 8.In October 2006 the US FDA, approved SAHA in thetherapy of refractory, relapsed cutaneous T-cell lymphoma(CTCL) and is at the present involved in a vast variety ofclinical trials both in hematological malignancies, such asleukemia, MDS, lymphoma and myeloma, and solidtumors. The mechanism of action is still unclear becauseof the involvement of many pathways, including apoptosis,autophagy, and induction of ROS and DNA damage repair,all of them subsequent to re-expression of genes thatbecome accessible to transcription factors, when hystoneproteins are in an acetylated status.The clinical efficacy of SAHA inspired the development ofnew analogues of the same class, as the indolyethylaminomethylcinnamyl hydro-amides LAQ-824 and LBH 589(panobinostat; Arts et al. 2007).Panobinostat, as SAHA, is in a vast variety of phase II/IIIclinical trials, both in solid tumors and hematologicalmalignancies, such as lymphomas, multiple myeloma,MDS, acute myeloid leukemia, and CML. Its HDACinhibition is strong against class I HDACs and lesspotent against class IIa.Belinostat (PXD101) is a hydroxamic acid derivative,which has been administered as an infusion on days 1 to5 of a 21-day cycle in a phase I study in patients withadvanced B-cell malignancies refractory to standardtherapy.Clin Epigenet (2010) 1:25–44Cyclic peptidesThe cyclic peptide Romidepsin, also known as FK-228, hasbeen reported to induce cell cycle arrest and apoptosis in avariety of human cancer cells. In vitro it has shown a strongactivity against HDAC 1 and 2, but also against HDAC 6and HDAC 4, although it results to be weaker (Mai andAltucci 2009). The drug has been in clinical trials for CMLand AML (Byrd et al. 2005; Piekarz et al. 2004) and hasbeen approved in November 2009 for the treatment ofrefractory–relapsed CTCL (Kim et al. 2008).BenzamidesThis class of HDACis presents a different structure fromthe other classes because of the 2′-aminoanilide moiety,which can likely function as a weak zinc-chelatinggroup into the tube-like active site of the deacetylasecore of the enzyme as suggested by molecular modelingstudies (Wang et al. 2005), or may contact key aminoacids in the active site without Zn ion coordination.MS-275 (entinostat) inhibits preferentially HDAC 1, 2,and 3 and is inactive (IC50s 10 μM) against HDAC 4,6, 7, and 8 (Khan and La Thangue 2008). In clinicaltrials, it has been used in solid tumors, such as lungand breast cancer and metastatic melanoma, and inhematological malignancies, as CML, AML, CMML, andHodgkin disease.MGCD 0103 is a more recent benzamide that showsselectivity for HDAC I/II. It has been used in clinical trialsmostly for hematological malignancies, such as AML,CML, non-Hodgkin lymphomas and refractory Hodgkindisease, where it has shown very encouraging results.SIRT inhibitorsThe compounds of this class of HDACis (class III) aremuch less validated as anticancer agents compared toHDAC I and II. This is also due to the fact that a lownumber of preclinical in vivo data exist presently and asmall number of inhibitors are on the market.Sirtuins play an important role in many cellularprocesses such as gene silencing and regulation oftranscription factors. Members of the sirtuin family ofNAD -dependent protein deacetylases SIRT1 controls celldifferentiation, metabolism, circadian rhythms, stressresponses, and cellular survival. When tested in the humanleukemia U937 cell line, SIRT inhibitors displayed highlevels of apoptosis and cyto-differentiation. SIRT1 bindsand deacetylates the histone acetyltransferase p300, whichinhibits p300 enzymatic activity, which has the potential topromote hypoacetylation of nucleosomal histones andaffect gene expression outcomes (Zhang et al. 2009).

Clin Epigenet (2010) 1:25–44Cambinol is a SIRTi (against SIRT1 and -2, respectively)that induces apoptosis in BCL-6 expressing Burkit lymphoma cells, through acetylation of both BCL-6 and p53(Heltweg et al. 2006). Finally, the anticancer action of SIRTinhibitors has been also reported to act via reactivation ofmethylated genes without evident DNA demethylation atthe promoters level (Pruitt et al. 2006).Mechanism of actionThe clinical manifestation of aberrant HDAC expression ishistology dependent. Each of the HDACs have differenttargets, not only interacting directly with chromatin throughthe acetylation of core histones (Ozdag et al. 2006) or“marking” chromatin for further recruitment of chromatinremodeling complexes (Choi and Howe 2009), such ashormone receptors, DNA repair enzymes, and signaltransduction mediators chaperone proteins and cytoskeletonproteins, regulating cell proliferation and cell death(Dokmanovic and Marks 2005). Acetylation can eitherincrease or decrease the function or stability of the proteinsor protein–protein interaction (Glozak et al. 2005).Gene expressionAcetylation generally results in an increase of the negativecharge on histone core leading to an uncoiled chromatinstructure, and accessibility to transcription factors ofdifferent genes, but the final gene expression is the resultof the interaction of other alterations in chromatin structuremediated by DNA, such histone methylation, and thesummation of activators and repressors recruited to therespective promoters (Ellis et al. 2008). It is important tostress that the number of genes detected with alteredtranscription is strongly influenced by the time of cultureand concentration of HDACi. A short exposition and a lowconcentration of HDACi are associated with fewer changes,compared to consisting doses of the drugs and longer timepoints (Ellis et al. 2008).The gene that seems to act on of the most induced byHDACis is a cyclin-dependent kinase, p21, which isregulated via p53 dependent as well as p53 independent;its expression correlates with an increase in the acetylationof histones associated with the p21 promoter region(Richon et al. 2000) and coincides with acetylation ofhistones H3 and H4.HDACis are also able to increase the activity of5-azacytidine and 5-aza-2′-deoxycytidine of the expressionof suppressor gene promoters, silenced by aberrant methylation (Cameron et al. 1999). This mechanism of potentiation resulted to be more complex because recent studiessuggest that TSA decreases the stability of DNMT3b31mRNA, resulting in decrease of de novo methylation inendometrial cancer cells (Xiong et al. 2005).Cell cycle regulationHDACis have shown to impact the cell cycle by inducing,at low concentrations, predominantly G1 arrest (Markset al. 1996) and G1 and G2/M arrest, at high concentration(Richon et al. 1997).Cell cycle arrest is associated with decreased expressionof cyclins A, B, and D, as well as their respective cyclindependent kinases, and induction of p21 and p27 (Emanueleet al. 2008). TSA, FK 228, deplete levels of kinethocoreproteins and decreased phosphorylation of histone H3 inpericentrometric chromatin during G2 phase, while SAHAinhibits the transcription of aurora kinase A and B, resultingin apparent mitotic catastrophe (Dowling et al. 2005; Parket al. 2008; Robbins et al. 2005).Regulation of the apoptotic pathwaysExtrinsic pathway of apoptosis is initiated by the binding ofdeath receptors to their ligands leading to activation ofcaspase-8 and caspase-10. HDACis upregulate the expression of the death receptors, Fas (Apo-1 or CD95), tumornecrosis factor (TNF) receptor-1 (TNFR-1), in transformedcells, but not in normal cells (Nakata et al. 2004). TRAILand its receptor DR-5 were induced in the mouse model ofAPL by VPA (Insinga et al. 2005); TNF-related apoptosisinducing ligand death receptors (DR-4 and -5) wereinduced in the mouse model of APL by VPA.The intrinsic apoptosis pathway is mediated by mitochondria with the release of mitochondrial inter-membraneproteins, such as cytochrome c, apoptosis-inducing factorSmac, and the consequent activation of caspases. It isregulated, in part, by pro- and anti-apoptotic proteins ofbcl-2 family (Insinga et al. 2005). Activation of the intrinsicapoptotic pathway is a major pathway for HDACis toinduce cell death utilizing mechanisms that are still not wellunderstood. HDACis facilitate the release of cytochrome cfrom mitochondrial inter-membrane space and activation ofcaspase-9 (Marks et al. 1996; Bolden et al. 2006), overexpression of Bcl-2 or Bcl-XL, while protecting themitochondria, inhibit HDACi-induced apoptosis.HDACis and autophagyAutophagy is a mechanism of cellular proteins selfdigestion by complexes called auto-phagosomes rich inlysosome, activated in cells during stress conditions, suchas ATP deprivation, infections, and glucose deprivation.This mechanism of death is caspase independent andinduced by nuclear, and p73 upregulation in response to

32cellular stress, and is p53 independent. HDACis regulateautophagy by inhibiting

nuclei, which package and order the DNA into structural units called nucleosomes. They can be grouped into five major classes: H1/H5, H2A, H2B, H3, and H4. These are organized into two super-classes: core histones, H2A, H2B, H3, and H4 and linker histones, H1 and H5. Two core histones assemble to form one octameric nucleosome core

Related Documents:

The occurrence of histone marks is catalyzed by histone modifying enzymes, which can be classified into three major groups. Histone writers are the enzymes that catalyze the covalent attachment of the aforementioned small molecules, such as histone methyltransferases (HMTs), histone acetyltransferases (HATs). The second group are

Histones, the structural unit of chromatin, must be assembled/dissembled to preserve or change . Histone chaperones can be illustrated on the basis of their histone binding selectivity and thus deposition, us- . which interacts with histone s H3/H4 and H1 [17]. This raises the question of how histone chaperones interact with histones. Many .

reported ndings, the combination of MEK inhibitors with chemotherapy, immune checkpoint inhibitors, epidermal growth factor receptor-tyrosine kinase inhibitors or BRAF inhibitors is highly signicant for improving clinical ecacy and causing delay in the occurrence of drug resistance. This review summarized the existing experimental results and

fications, specifically reversible histone methylation. A nucleosome, the repeating unit of chromatin, is composed of a histone octamer core, which consists of two copies of each histone H2A, H2B, H3, and H4 pro-teins, and a short segment of DNA, between 145 and 147 base pairs, which is wrapped around it (Fig. 1). The

essential function of FLA SH in Drosophila is histone mRNA 3′ end formation as a component of the HLB (see Fig. 4). To understand the molecular basis for the FLASH mutant phenotype, we examined the histone mRNA species produced in various FLA SH mutant genotypes (Fig. 1 E). When histone pre-mRNA

Structural Basis for the Recognition of Methylated Histone H3K36 by the Eaf3 Subunit of Histone . sible for histone H4 acetylation at lysines 5, 8, and 12 (Allard et al., 1999; Clarke et al., 1999; Loewith et al., 2000; Reid et al., 2000), and may be recruited to nucleosomes by interaction with

core particle consists of four histone proteins, Histone H2A, H2B, H3, and H4, and has an octa-meric structure, formed by a central heterotetramer of histones H3 and H4, and two heterodimers of histones H2A and H2B.1-5 Additionally a linker pro-tein, Histone H1/H5, locks the DNA onto the core domain. All together, they constitute a spool-like

ASME BPV CODE, EDITION 2019 Construction Code requirements Section VIII, Div. 1, 2 a 3 ; Section IX ASME BPV Section V, Article 1, T-120(f) ASME BPV Section V, Article 1, Mandatory Appendix III ASME BPV Section V, Article 1, Mandatory Appendix II (for UT-PA, UT-TOFD, RT-DR, RT-CR only ) SNT-TC-1A:2016; ASNT CP-189:2016 ASME B31.1* Section I Section XII ASME BPV Section V, Article 1, Mandatory .