You are on page 1of 9

Send Orders for Reprints to reprints@benthamscience.

net
Current Molecular Pharmacology, 2020, 13, 1-0 1
RESEARCH ARTICLE

Epigenetic Evaluation of N-(2-hydroxyphenyl)-2-Propylpentanamide, a


Valproic Acid Aryl Derivative with Activity Against HeLa Cells
GR Luna-Palencia1, J Correa-Basurto2, J Trujillo-Ferrara3, MA Meraz-Ríos4 and
I Vásquez-Moctezuma5,*

1
Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados del Instituto Pol-
itécnico Nacional, Av. IPN 2508, San Pedro Zacatenco, CDMX, 07360, México; 2Laboratorio de Diseño y Desarrollo
de Nuevos Fármacos e Innovación Biotecnológica (Laboratory for the Design and Development of New Drugs and
Biotechnological Innovation), Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz
Mirón s/n, Casco de Santo Tomás, CDMX, 11340, México; 3Laboratorio de Bioquímica, Laboratorio de Modelado
Molecular, Bioinformática y Diseño de Fármacos de la Escuela Superior de Medicina, Instituto Politécnico Nacional,
Plan de San Luis y Díaz Mirón s/n, Casco de Santo Tomás, CDMX, 11340, México; 4Departamento de Biomedicina
Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional; 5Laboratorio de Mor-
fología de la Maestría en Ciencias de la Salud, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de
San Luis y Díaz Mirón s/n, Casco de Santo Tomás, CDMX, 11340, México

Abstract: Background: Valproic acid (VPA) is an HDAC inhibitor (HDACI) with an anticancer
activity, but it is hepatotoxic. N-(2-hydroxyphenyl)-2-propylpentanamide (o-OH-VPA) is a VPA
aryl derivative designed in silico as a selective inhibitor of HDAC8 with biological properties
against HeLa, rhabdomyosarcoma and breast cancer cell cultures.

Objective: Objective: We studied the epigenetic mechanism of o-OH-VPA as an HDACI and we


evaluated whether it was toxic to normal cells.

Methods: HeLa cells and primary human fibroblasts were used for this study as carcinogenic and
ARTICLE HISTORY normal cells, respectively. Cell survival was evaluated by MTT assay, whereas viability and dou-
bling time were determined by the Trypan-blue method. HDAC activity was tested using the colori-
Received: February 06, 2020 metric HDAC activity assay. The expression of p21 was analyzed by PCR and HDAC8 expression
Revised: May 14, 2020
Accepted: May 25, 2020 was also evaluated by real-time PCR. Cell cycle and caspase-3 activity were analyzed by flow cy-
tometry and caspase-3 colorimetric assay, respectively.
DOI:
10.2174/1874467213666200730113828
Results: o-OH-VPA (IC50 = 0.1 mM) was fifty-eight times more effective than VPA (IC50 = 5.8
mM) to reduce HeLa cell survival. Furthermore, o-OH-VPA increased the doubling time of HeLa
cells by 33% with respect to the control. o-OH-VPA acted as HDACI in HeLa cells without affect-
ing the HDAC8 expression, arresting the cell cycle of HeLa cells in the G0/G1 phase due to the in-
crease in p21 expression with the inhibition of caspase-3 activity without exhibiting toxicity to-
ward normal cells.

Conclusion: Our results revealed that o-OH-VPA is an HDACI with a selective effect against
HeLa cells but without the known toxicity exerted by most pan-HDACIs on normal cells.

Keywords: Anticancer agents, histone deacetylase 8 inhibitor, valproic acid, aryl valproic acid derivative, HeLa cells, epigenet-
ic drugs.

1. INTRODUCTION E-mail: g17isma65@gmail.com

Reversible acetylation of histones is an epigenetic modi- teraction of positively charged histones with negatively
fication which is involved in the regulation of gene expres- charged DNA, and this regulates chromatin conformation
sion that is carried out by acetyltransferase and histone and transcriptional activity [1]. This control occurs because
deacetylase (HDACs) enzymes. These modifications con- HDACs remove the acetyl group of lysine residues located
trol the in- at the N-terminal tails of the histones, leading to a more
closed chromatin conformation that hinders the accessibility
* Address correspondence to this author at the Morfolog, Escuela Superior of transcription factors to DNA [2]. In addition, HDACs con-
de Medicina del IPN, Salvador D, Mexico; Tel: 5557296000; trol the acetylation status and function of numerous non-his-
1874-4672/20 $65.00+.00 © 2020 Bentham Science Publishers
2 Current Molecular Pharmacology, 2020, Vol. 13, No. 0 Luna-Palencia et al.

tone proteins and transcription factors [3]. Since epigenetic cultured in Dulbecco-Modified Eagle’s Medium (DMEM
alterations have been associated with cancer, new treatments Glutamax, Gibco, Life-technologies, Invitrogen, USA) sup-
are being under developement based on inhibitors of histone plemented with 10% (v/v) heat-inactivated fetal bovine
deacetylation [4-6]. HDACs are involved in regulating pro- serum (FBS, Biowest, Kansas City, MO, USA) and an antibi-
cesses such as the cell cycle, differentiation, apoptosis, mi- otic-antimycotic reagent (Gibco). The cell cultures, both un-
gration, invasion and angiogenesis of cancer cells [7]. At treated and treated with VPA or o-OH-VPA, were main-
this time, there are 18 HDAC family members that are tained at 37°C in a humidified atmosphere containing 5%
grouped into four classes: class I (1, 2, 3 and 8); class II,
CO2. DMSO was used as a drug vehicle in all the cell cul-
which is further divided into classes IIa (4, 5, 7 and 9) and
IIb (6 and 10); class III (12-18) and class IV (11). Classes I, ture experiments.
II, and IV are also called “classical” HDACs, and all these
2.2. MTT Assay
have Zn 2+ cofactor, whereas class III includes HDACs that
3
are nicotinamide adenine dinucleotide (NAD+) dependent, re- HeLa cells (1 x 10 cells per well) or human fibroblast (F-
ferred to as sirtuins [8, 9]. Since there are differences in the 3
B) cells (3 x 10 cells per well) were seeded in 96-well cul-
activity of histone deacetylases inhibitors (HDACIs) against tured plates in 100 µL DMEM containing 10% heat-inacti-
specific classes of HDACs, these may contribute to their tu- vated FBS. After 24 h, HeLa cells were treated with fresh
mor cell selectivity observed. The fatty acid valproic acid in- medium containing VPA (2, 4, 6 and 8 mM) or o-OH-VPA
hibits class I HDACs 1, 2, 3 and 8 in the mM scale, but it is (0.05, 0.1, 0.2, 0.4 and 0.5 mM); On the other hand, FB cells
ineffective against HDACs 6, 7 and 9 [10]; The very high were treated with fresh medium containing 5 mM VPA or
concentration required for its anti-tumor activity is one of 0.1 mM o-OH-VPA (IC50 for HeLa cells), so as to evaluate
the major drawbacks [7], for instance, sodium valproate in- their cytotoxicity in non-tumor cells. The plates were incu-
duces mitochondrial respiration dysfunction in HepG2 cells bated for 48 h and were analyzed for cell survival using the
at 2 mM [11]. colorimetric 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetra-
HDAC8 is a class I HDAC related to carcinogenesis and zolium bromide (MTT) dye reduction assay (Sigma-Aldrich,
cancer progression, and its expression levels are elevated ap- St. Louis, MO, USA) [14].
proximately 10-fold in HL-60, HeLa S3, K-562, MOLT-4,
SW480, A549 and G361 cancer cell lines compared to nor- 2.3. Trypan Blue Exclusion Method
mal tissues [12]. Moreover, HDAC8 is associated with the HeLa cells (10 x 103 cells per well) were seeded in 24-
regulation of the proliferation, clonogenic growth, and neuro- well cultured plates in 500 µL DMEM containing 10% FBS.
nal differentiation of neuroblastoma cells and its high expres- After 24 h, HeLa cells were treated with fresh medium con-
sion correlates with neuroblastoma progression and poor sur- taining VPA (2, 4, 6, 8 and 10 mM) or o-OH-VPA (0.1, 0.2,
vival prognoses [13]. HDAC 8 can be targeted using com- 0.3, 0.4 and 0.5 mM). Plates with different concentrations of
pounds that have a chemical scaffold with metal chelators or the compounds were incubated for 48 h, and those with the
hydrophobic properties. Our research group has been fo- same concentration were incubated for 24, 48 and 72 h. The
cused on drug design for some of the ligands with hydropho- viability, or live cells per well, was determined by the Try-
bic moieties (from valproic acid) and amino acids [14, 15] pan-blue exclusion method. For this, adherent and floating
or aryl derivatives [16, 17]. N-(2-hydrox- cells were collected, and 0.4% Trypan-blue solution (Sig-
yphenyl)-2-propylpentamide (o-OH-VPA) is a valproic acid ma-Aldrich, St. Louis, MO, USA) was added 1:1 (v/v) and
(VPA) aryl derivative (Fig. 1) designed in silico by mixing counted in a TC10 Automated Cell Counter (BIO-RAD, Lab-
VPA with the arylamine core of suberoylanilide hydroxamic oratories, CA, USA). The doubling time was calculated with
acid (SAHA) that shows biological properties against human 0, 24 and 48 h data [18].
cancer [16, 17]. However, the pharmacological mechanism
of o-OH-VPA remains unknown. 2.4. HDAC Activity Assay
This work aimed to evaluate the effect of o-OH-VPA on Assays for HDAC activity were performed using the col-
the proliferation of HeLa cells by MTT and trypan blue as- orimetric HDAC activity assay (BioVision Research Prod-
says, as well as its action as HDACI by determining the total ucts, Mountain View, CA, USA), which eventually detects
enzymatic activity of HDACs, expression of HDAC8, analy- the activity of all HDACs that can deacetylate histone. HeLa
sis of the cell cycle and caspase-3 activity. Moreover, we al- nuclear protein (50 µg) was obtained as described in a previ-
so tested the toxicity to normal cells (primary human fi- ous study [14] and was diluted with 83 µL of ddH2O and 2
broblasts). µL of both 250 mM VPA or 5 mM o-OH-VPA as appropri-
ate, followed by 10 µL of the 10X HDAC assay buffer, and
2. MATERIALS AND METHODS then 5 µL of the HDAC substrate. After incubating the plate
for 1 h at 37°C, the reaction was ended with 10 µL of Lysine
2.1. Cell Cultures
Developer, subsequently, the solution was incubated for 30
HeLa cell line derived from cervical cancer and human min at 37°C. Finally, the absorbance at 405 nm was read us-
primary dermal fibroblast ATCC PCS-201-012 (FB) were ing a microplate reader.
Epigenetic Evaluation of N-(2-hydroxyphenyl)-2-Propylpentanamide Current Molecular Pharmacology, 2020, Vol. 13, No. 0 3

2.5. Total RNA Extraction, Reverse Transcription and H-VPA were diluted with 50 µL cell lysis buffer and 50 µL
Polymerase Chain Reaction to Evaluate the Expression of 2X Reaction Buffer (containing 10 mM DTT). DEVD-p-
of HDAC8 and p21 NA substrate was added to achieve a final concentration of
Total RNA was isolated with TRIzol (Invitrogen CA, 200 µM, and this mixture incubated at 37°C for 2 h. In addi-
USA) according to the manufacturer’s instructions. RNA in- tion, for each sample, a blank was prepared equal to its corre-
tegrity was electrophoretically verified with 1% agarose gel sponding sample, but without adding the substrate, for sub-
by ethidium bromide staining, and RNA purity was verified tracting background. Samples were read at 405 nm in a mi-
by an OD260/OD280 nm absorption ratio >1.8. The total RNA crotiter plate reader.
(0.5 µg) of HeLa cells, untreated or treated with 5 mM VPA
2.9. Statistical Analysis
or 0.1 mM o-OH-VPA for 24 h, was reverse transcribed
with the Maxima First Strand cDNA Synthesis kit (Thermos- Data were expressed as the mean ± SD. One or two-way
cientific Pittsburgh, USA), and a polymerase chain reaction ANOVA followed by Dunnett’s multiple comparisons test
was conducted using the PCR Master Mix (2X) (Thermosci- was performed, as would be appropriate, using GraphPad
entific Pittsburgh, USA). Primer design was performed us- Prism version 6.00 for Windows, GraphPad Software, La Jol-
ing the Oligo Explorer software 1.1.2, hence the primer se- la California USA (www.graphpad.com).
quences are shown in Table 1, including GAPDH primers as
housekeeping gene control. The thermal cycling conditions 3. RESULTS
for each primer set were 95°C for 3 min followed by 35 cy- The concentration-response curve (Fig. 2A) showed that
cles which are as follows: denaturation step at 95°C for 30 s; either o-OH-VPA or VPA reduced the HeLa cell survival in
annealing at 50°C for 30 s; and extension at 72°C for 30 s. a concentration-dependent manner, but o-OH-VPA was fif-
Finally, a final extension cycle was run at 72°C for 5 min. ty-eight times more effective than VPA (0.1 mM vs 5.8
PCR products were visualized on a 3% agarose gel by ethidi- mM). Moreover, the membrane integrity, and consequently
um bromide staining. the viability of the HeLa cells, was reduced to 80.3 ± 8.6%
by o-OH-VPA compared to control cells near the IC50 ob-
2.6. Real-Time Quantitative PCR tained by MTT at 48 h (Fig. 2B).
Real-time qPCR was performed using the Power SY- The number of living HeLa cells per well, evaluated by
BR® Green PCR Master Mix (2X), purchased from Applied the trypan blue assay decreased 48% after 48 h of treatment
Biosystems. Reactions (10 µL) were conducted using a Ste- with 0.1 mM o-OH-VPA, was similar to the cells treated
pOne Real-Time PCR System, in which the concentrations with 5 mM VPA, compared with 0.5% DMSO (vehicle);
of primers were each 150 nM and 1 µL of cDNA diluted to that is, from 89 X 103 to 43 X 103 cells. At 72 h of treatment
1/10. The thermal cycling conditions for each primer set with o-OH-VPA treatment, the number of living HeLa cells
were 95°C for 10 min followed by 40 cycles as follows: de- per well decreased by 75% and 51% with respect to the con-
naturation step at 95°C for 15 s and annealing at 60°C for 60 trol and VPA. The doubling time of HeLa cells treated with
s. Finally, the melt curve was obtained. The experiment was 0.1 mM o-OH-VPA (15.4 h) was 33% and 7% higher than
carried out in triplicate, and each sample was also run in tri- that observed in HeLa cells treated with 0.5% DMSO (11.6
plicate. The relative expression to GAPDH was determined h) or 5 mM VPA (14.5 h), respectively (Fig. 3).
using the Pfaffl method [19]. The total activity of HDACs in HeLa nuclear protein ex-
tract treated with 0.1 mM o-VPA-OH was reduced only by
2.7. Cell Cycle and Apoptosis Analysis 23% compared to approximately 90% caused by 5 mM VPA
treatment (Fig. 4). In contrast, the overexpression of
Unsynchronized HeLa cell cultures were treated with 5 HDAC8 observed in the HeLa cells treated with 5 mM VPA
mM VPA or 0.1 mM o-OH-VPA for 48 h. Adherent and was not observed in the cells treated with 0.1 mM o-OH-V-
floating cells were collected by centrifugation and were PA (Fig. 5).
fixed in cold 70% ethanol. Fixed cells were washed twice
with cold PBS and treated with 0.2 mg/mL RNAse A and lat- The decrease in the number of viable HeLa cells caused
er stained with 40 µg/mL propidium iodide. The samples by 0.1 mM o-OH-VPA was associated with the relative in-
were analyzed using an LSR Fortessa flow cytometer (BD crease in p21 expression (Fig. 6A), as well as an increase
Biosciences). Cell cycle and apoptosis analysis were deter- (7.4%) in the numbers of HeLa cells in the G0/G1 phase
mined by the ModFit LT program. (58.6 ± 1.4%) compared to the cells treated with DMSO
(51.2 ± 1.2%), and the decrease in the cell cycle S phase
(Fig. 6B). Finally, it was observed that the activity of cas-
2.8. Caspase-3 Assay pase-3 (Fig. 6C), as well as apoptosis induction (Fig. 6B) in
Assays were performed using the Caspase-3/CPP32 col- HeLa cells treated with 0.1 mM o-OH-VPA, were reduced
orimetric assay kit (BioVision Research Products, Mountain by almost 50% with respect to 0.5% DMSO-treated HeLa
View, CA, USA) according to the instructions by the manu- cells.
factures. Cytosolic protein (100 µg) of HeLa cells treated Despite its toxic effect on HeLa cells (cancer cells), o-O-
with 0.5% DMSO (D, vehicle), 5 mM VPA or 0.1 mM o-O- H-VPA did not show toxicity towards normal cells, such as
4 Current Molecular Pharmacology, 2020, Vol. 13, No. 0 Luna-Palencia et al.

primary cultures of human fibroblasts, when they were chal- total activity of HDACs in HeLa nuclear protein extract treat-
lenged with the IC50 obtained in HeLa cells. In contrast, ed with 0.1 mM o-OH-VPA since only a decrease of 23%
VPA 5 mM reduced the fibroblast cell survival to 66.5 ± was observed.
3.7% (Fig. 7), demonstrating a selective toxic effect of o-O- o-OH-VPA at 0.1 mM did not increase HDAC8 gene ex-
H-VPA toward HeLa cancer cells. pression, as was the case for the 5 mM VPA, which is inter-
esting because HDAC8 is important for human tumor cell
4. DISCUSSION lines growth since the proliferation of lung, colon and cervi-
Most currently used HDACIs act rather non-selectively cal cancer cell lines is reduced after HDAC8 knockdown
and inhibit either all or at least several members of the [24], This reduction indicated that the inhibition of HDAC8
HDAC family [7]. Our research group is focused on the ra- is implicated in the decrease of proliferation.
tional design of selective HDACIs by using particularly crys- o-OH-VPA induced the overexpression of p21, which is
tal structures of HDAC8 as non-HDAC selective inhibitors a potent inhibitor of most cyclin/CDK complexes that result
approved by the FDA caused a variety of side-effects such in an accumulation of cells in the G0/G1 phase, preventing
as bone marrow depression, diarrhea, weight loss, taste dis- cells from shifting to the S-phase [25] and inhibiting cas-
turbances, electrolyte changes, disordered clotting, fatigue, pase-dependent apoptosis in HeLa cells. Reduced growth
and cardiac arrhythmias [7]. will protect the cell against long-term genome damage
Our results showed that o-OH-VPA had anti-prolifera- caused by replicative stress through the S-phase and chromo-
tive properties against HeLa cells and was fifty-eight times some missegregation during mitosis [26]. Previous studies
more effective than VPA, as demonstrated by its IC50 that have demonstrated that the cell cycle inhibitor p21 is com-
was determined using the MTT assay (Fig. 2). The o-OH-V- monly induced in response to HDACI [27, 28] and plays a
PA affected the membrane integrity more than VPA, decreas- key role in the G1 cell cycle arrest of many cancer cells
ing the viability of HeLa cells as a cytotoxic effect in a con- [29]. The progression through the G1-S checkpoint is neces-
centration-dependent manner. Furthermore, 0.1 mM o-O- sary for the induction of apoptosis by HDACIs, such as SA-
H-VPA decreased the proliferation of HeLa cells by increas- HA, oxamflatin and depsipeptide, because cells arrested in
ing the doubling time in more than 30% compared to the the G1 phase are resistant to HDACI-induced death [30].
cells that were treated with 0.5% DMSO as the control, with- The above was observed with o-OH-VPA, where apoptosis
out affecting the viability after 48 h of treatment. o-OH-V- and caspase-3 activity were decreased as a result of G0/G1
PA was designed in silico as a selective inhibitor of HDAC8 arrest. It is important to determine whether cell cycle arrest
[17], and it has a similar effect as an HDACI in HeLa cells; is a permanent state due to DNA damage caused by o-OH-V-
o-OH-VPA was recently reported to have an affect on HMG- PA because cellular senescence, which is a potent tumor sup-
B1, an essential enzyme with high expression in tumor cells pressive mechanism, can be activated [31].
which is dependent on its acetylation state [20]. VPA inhibit-
ed approximately 90% of HDACs at 5 mM treatment, o-OH-VPA did not show toxicity for normal cells, as ob-
whereas for o-OH-VPA at 0.1 mM, it was only 20%. In con- served with the human fibroblasts used in this work. The
trast to VPA, which has been reported to be a class I mechanism by which HDACIs exhibit selective toxicity for
HDACs (HDAC1, 2, 3, and 8) inhibitor that induces the cancer cells has not been defined in its entirety, Still there is
acetylation of histone H4 and slightly inhibits the acetyla- a close relationship between histone acetylation and the tran-
tion of histone H3 [10], o-OH-VPA could be a specific scriptional activity of specific genes that are involved in pro-
HDAC8 inhibitor. HDAC8 is primarily localized in the nu- liferation and/or apoptosis [32]. For example, it has been de-
cleus [21] but appeared to be more concentrated in the sub- monstrated that HDACs 8 and 6 are overexpressed in HeLa
nuclear regions rather than be distributed evenly throughout cells [33]. In contrast, it had previously been demonstrated
the entire nucleus. This observation suggests that HDAC8 that fibroblasts could suffer extensive damage in the pres-
may act on other proteins in addition to histones and may ence of HDACIs [34]. HDACIs cause rapid and extensive hi-
have only limited deacetylation activity on histone proteins; stone hyperacetylation, and consequently, changes in gene
The activity of HDAC8 is low compared to that of HDAC1, expression that can occur in a small proportion of genes
and its precise target of deacetylation has not yet been identi- (10% or less) [35-38] or in more than 40% of genes [32].
fied [12]. According to recent results, however, o-OH-VPA However, selective HDAC8 inhibitors showed a unique pat-
could also inhibit other HDACs, according to recent results, tern of hyperacetylated proteins compared to the pan-HDAC
because o-OH-VPA helps HMGB1 migration from the nu- inhibitor Trichostatin A in human cells [39]. The “epigenetic
cleus to the cytoplasm [20]. Waltregny and coworkers found vulnerability” of certain cancer cells was proposed by Daw-
that HDAC8 is predominantly cytosolic, both in human tis- son and Kouzarides in 2012 as a cause of the relative speci-
sues and in vitro-grown human vascular smooth muscle ficity of HDACIs for cancer cells. The hypothesis proposed
cells, where it displays a cytoskeleton-like pattern of expres- that normal cells have alternative compensating pathways,
sion and may colocalize with α-actin [22]. Some selective whereas cancer cells have a particular set of “essential”
HDAC8 inhibitors have been developed, such as PCI-34051, genes that are regulated by epigenetic components, allowing
but when compared to other broad-spectrum HDAC inhibi- epigenetic inhibitors to disrupt the expression of a few key
tors, they do not cause detectable acetylation of histones or target genes, leading to catastrophic effects in malignant
tubulin [23], This has been consistent with our results of the cells [40].
Epigenetic Evaluation of N-(2-hydroxyphenyl)-2-Propylpentanamide Current Molecular Pharmacology, 2020, Vol. 13, No. 0 5

Table 1. Oligonucleotides used to evaluate the expression of some genes associated with the arrest of the cell cycle and with epigenet-
ic mechanisms.

Gene Forward Reverse Size (bp)


1 p21 5’-AgACTCTCAgggTCgAAAAC-3’ 5’-ATTAgggCTTCCTCTTggAg-3’ 92
2 HDAC8 5’-gTCCCgAgTATgTCAgTATg -3’ 5’-CTATCCTCATCTgCTTATgC-3’ 108
3 GAPDH 5’-gTATgACAACAgCCTCAAgAT-3’ 5’-gTCCTTCCACgATACCAAAg-3’ 104

Fig. (1). VPA and VPA aryl derivative: N-(2-hydroxyphenyl)-2-propylpentamide (o-OH-VPA). (A higher resolution / colour version of this
figure is available in the electronic copy of the article).

Fig. (2). Effects of VPA and o-OH-VPA on the survival (A) and viability (B) of HeLa cells. Cells were incubated for 48 h at different concen-
trations of the compounds; the cell survival and viability were analyzed by MTT assay and the Trypan-blue exclusion method, respectively.
Data are expressed as the mean ± SD for n ≥ 6 wells from at least two independent experiments for cell survival and n = 4 for viability. (A
higher resolution / colour version of this figure is available in the electronic copy of the article).
6 Current Molecular Pharmacology, 2020, Vol. 13, No. 0 Luna-Palencia et al.

Fig. (3). Live cells per well, their doubling time (Dt) (A) and viability (B) of HeLa cells after being treated with 5 mM VPA and 0.1 mM o-O-
H-VPA for 24, 48 and 72 h as determined by the Trypan-blue exclusion method. Data are expressed as the mean ± SD for n = 3. (A higher re-
solution / colour version of this figure is available in the electronic copy of the article).

Fig. (4). Effects of 5 mM VPA and 0.1 mM o-OH-VPA on HDAC activity. Nuclear extracts from HeLa cells with HDAC activity were incu-
bated with 0.5% DMSO as a control and with the compounds. The percentage of HDAC activity is expressed as the mean ± SD for n = 3
from three independent experiments. (A higher resolution / colour version of this figure is available in the electronic copy of the article).

Fig. (5). Expression of the HDAC8 gene by PCR (A) and qPCR (B) of HeLa cells treated with 5 mM VPA and 0.1 mM o-OH-VPA for 24 h.
The relative fold expression is expressed as the mean ± SD for n = 3 from three independent experiments. (A higher resolution / colour ver-
sion of this figure is available in the electronic copy of the article).
Epigenetic Evaluation of N-(2-hydroxyphenyl)-2-Propylpentanamide Current Molecular Pharmacology, 2020, Vol. 13, No. 0 7

Fig. (6). Expression of the p21 gene related to the arrest of the cell cycle (A), as well as the flow cytometry analysis of the cell cycle and
apoptosis (B) and the activity of caspase-3, where 1 µM Camptothecin was used as a positive control (C). Data are expressed as the mean ±
SD for n = 3 from three independent experiments. (A higher resolution / colour version of this figure is available in the electronic copy of the
article).

Fig. (7). Effect of 5 mM VPA and 0.1 mM o-OH-VPA on human fibroblast (FB) cell survival. Fibroblasts were incubated for 48 h with 0.5%
DMSO as a control and with the compounds at a concentration close to the IC50 obtained from assays with HeLa cells. The percentage of cell
survival was determined by the MTT assay and was expressed as the mean ± SD for n = 8 from two independent experiments. (A higher reso-
lution / colour version of this figure is available in the electronic copy of the article).

CONCLUSION effective than VPA. Furthermore, o-OH-VPA is more selec-


This contribution aimed to describe the action mech- tive than VPA and the results suggested in this study, as it
anism and selectivity of o-OH-VPA toward cancer cells. targets HDAC8. Similar to other HDACIs, o-OH-VPA has
The novel HDAC inhibitor o-OH-VPA exhibits HDAC in- anti-proliferative properties toward cancer cells but shows
hibitory effects at the µM scale and is fifty-eight times more no toxicity toward normal human fibroblasts. The inhibited
8 Current Molecular Pharmacology, 2020, Vol. 13, No. 0 Luna-Palencia et al.

proliferation of HeLa cell cultures by o-OH-VPA functions [8] de Ruijter, A.J.; van Gennip, A.H.; Caron, H.N.; Kemp, S.; van
via arresting the cell cycle in the G0/G1 phase, which occurs Kuilenburg, A.B.P. Histone deacetylases (HDACs): characteriza-
tion of the classical HDAC family. Biochem. J., 2003, 370(Pt 3),
as a result of the overexpression of p21 without caspase-3 ac- 737-749.[http://dx.doi.org/10.1042/bj20021321] [PMID:
tivation. 12429021]
[9] Gregoretti, I.V.; Lee, Y.M.; Goodson, H.V. Molecular evolution
ETHICS APPROVAL AND CONSENT TO PARTICI- of the histone deacetylase family: functional implications of phylo-
genetic analysis. J. Mol. Biol., 2004, 338(1),
PATE 17-31.[http://dx.doi.org/10.1016/j.jmb.2004.02.006] [PMID:
Not applicable. 15050820]
[10] Khan, N.; Jeffers, M.; Kumar, S.; Hackett, C.; Boldog, F.;
Khramtsov, N.; Qian, X.; Mills, E.; Berghs, S.C.; Carey, N.; Finn,
HUMAN AND ANIMAL RIGHTS P.W.; Collins, L.S.; Tumber, A.; Ritchie, J.W.; Jensen, P.B.;
No animals/humans were used for studies that are the ba- Lichenstein, H.S.; Sehested, M. Determination of the class and iso-
form selectivity of small-molecule histone deacetylase inhibitors.
sis of this research. Biochem. J., 2008, 409(2),
581-589.[http://dx.doi.org/10.1042/BJ20070779] [PMID:
CONSENT FOR PUBLICATION 17868033]
[11] Komulainen, T.; Lodge, T.; Hinttala, R.; Bolszak, M.; Pietilä, M.;
Not applicable. Koivunen, P.; Hakkola, J.; Poulton, J.; Morten, K.J.; Uusimaa, J.
Sodium valproate induces mitochondrial respiration dysfunction
AVAILABILITY OF DATA AND MATERIALS in HepG2 in vitro cell model. Toxicology, 2015, 331,
47-56.[http://dx.doi.org/10.1016/j.tox.2015.03.001] [PMID:
Not applicable. 25745980]
[12] Van den Wyngaert, I.; de Vries, W.; Kremer, A.; Neefs, J.; Verhas-
FUNDING selt, P.; Luyten, W.H.; Kass, S.U. Cloning and characterization of
human histone deacetylase 8. FEBS Lett., 2000, 478(1-2),
We gratefully acknowledge CONACYT (Grants: 77-83.[http://dx.doi.org/10.1016/S0014-5793(00)01813-5] [P-
CB-254600 and APN-782), Instituto Politécnico Nacional MID: 10922473]
[13] Oehme, I.; Deubzer, H.E.; Wegener, D.; Pickert, D.; Linke, J.P.;
(Grant: Proyectos Insignia IPN-2015), and CO- Hero, B.; Kopp-Schneider, A.; Westermann, F.; Ulrich, S.M.; von
FAA-SIP/IPN. Deimling, A.; Fischer, M.; Witt, O. Histone deacetylase 8 in neu-
roblastoma tumorigenesis. Clin. Cancer Res., 2009, 15(1),
CONFLICT OF INTEREST 91-99.[http://dx.doi.org/10.1158/1078-0432.CCR-08-0684] [P-
MID: 19118036]
The authors have no conflicts of interest, financial or [14] Luna-Palencia, G.R.; Martínez-Ramos, F.; Vásquez-Moctezuma,
otherwise. I.; Fragoso-Vazquez, M.J.; Mendieta-Wejebe, J.E.; Padilla--
Martínez, I.I.; Sixto-López, Y.; Méndez-Luna, D.; Trujillo-Fer-
rara, J.; Meraz-Ríos, M.A.; Fonseca-Sabater, Y.; Correa-Basurto,
ACKNOWLEDGEMENTS J. Three amino acid derivatives of valproic acid: design, synthesis,
The authors wish to thank M. Sc. Rosales-García V. H. theoretical and experimental evaluation as anticancer agents. Anti-
cancer. Agents Med. Chem., 2014, 14(7),
from CINVESTAV-IPN for his technical support in the cell 984-993.[http://dx.doi.org/10.2174/187152061466614012711321
cycle analysis and to Dr. Efraín Garrido, CINVESTAV-IPN 8] [PMID: 24476311]
for providing the HeLa cells. [15] Martínez-Ramos, F.; Luna-Palencia, G.R.; Vásquez-Moctezuma,
I.; Méndez-Luna, D.; Fragoso-Vázquez, M.J.; Trujillo-Ferrara, J.;
REFERENCES Meraz-Ríos, M.A.; Mendieta-Wejebe, J.E.; Correa-Basurto, J.
Docking Studies of Glutamine Valproic Acid Derivative (S)-5-
[1] Chen, H.P.; Zhao, Y.T.; Zhao, T.C. Histone deacetylases and amino-2-(heptan-4-ylamino)-5-oxopentanoic Acid (Gln-VPA) on
mechanisms of regulation of gene expression. Crit. Rev. Oncog., HDAC8 with Biological Evaluation in HeLa Cells. Anticancer.
2015, 20(1-2), Agents Med. Chem., 2016, 16(11),
35-47.[http://dx.doi.org/10.1615/CritRevOncog.2015012997] [P- 1485-1490.[http://dx.doi.org/10.2174/1871520616666160204111
MID: 25746103] 158] [PMID: 26845132]
[2] Wolffe, A.P. Chromatin: Structure and Function., (3rd ed. ), 3rd [16] Luna-Palencia, G.R. Efecto de los inhibidores de las desacetilasas
ed. 1998, de histonas sobre la expresión de genes de resistencia a drogas en
[3] Li, T.; Song, B.; Wu, Z.; Lu, M.; Zhu, W.G. Systematic identifica- la línea celular HeLa., 2015.
tion of Class I HDAC substrates. Brief. Bioinform., 2014, 15(6), [17] Prestegui-Martel, B.; Bermúdez-Lugo, J.A.; Chávez-Blanco, A.;
963-972.[http://dx.doi.org/10.1093/bib/bbt060] [PMID: Dueñas-González, A.; García-Sánchez, J.R.; Pérez-González,
23975722] O.A.; Padilla-Martínez, I.I.; Fragoso-Vázquez, M.J.; Mendie-
[4] Hellebrekers, D.M.; Griffioen, A.W.; van Engeland, M. Dual tar- ta-Wejebe, J.E.; Correa-Basurto, A.M.; Méndez-Luna, D.; Trujil-
geting of epigenetic therapy in cancer. Biochim. Biophys. Acta, lo-Ferrara, J.; Correa-Basurto, J. N-(2-hydrox-
2007, 1775(1), 76-91.[PMID: 16930846] yphenyl)-2-propylpentanamide, a valproic acid aryl derivative de-
[5] Yoo, C.B.; Jones, P.A. Epigenetic therapy of cancer: past, present signed in silico with improved anti-proliferative activity in HeLa,
and future. Nat. Rev. Drug Discov., 2006, 5(1), rhabdomyosarcoma and breast cancer cells. J. Enzyme Inhib. Med.
37-50.[http://dx.doi.org/10.1038/nrd1930] [PMID: 16485345] Chem., 2016, 31(sup3),
[6] Minucci, S.; Pelicci, P.G. Histone deacetylase inhibitors and the 140-149.[http://dx.doi.org/10.1080/14756366.2016.1210138] [P-
promise of epigenetic (and more) treatments for cancer. Nat. Rev. MID: 27483122]
Cancer, 2006, 6(1), 38-51.[http://dx.doi.org/10.1038/nrc1779] [P- [18] Roth, V. 2006.Doubling Time Computing,
MID: 16397526] http://www.doubling-time.com/compute.php
[7] Witt, O.; Deubzer, H.E.; Milde, T.; Oehme, I. HDAC family: [19] Pfaffl, M.W. A new mathematical model for relative quantifica-
What are the cancer relevant targets? Cancer Lett., 2009, 277(1), tion in real-time RT-PCR. Nucleic Acids Res., 2001,
8-21.[http://dx.doi.org/10.1016/j.canlet.2008.08.016] [PMID: 29(9)e45[http://dx.doi.org/10.1093/nar/29.9.e45] [PMID:
18824292] 11328886]
Epigenetic Evaluation of N-(2-hydroxyphenyl)-2-Propylpentanamide Current Molecular Pharmacology, 2020, Vol. 13, No. 0 9

[20] Contis-Montes de Oca, A.; Rodarte-Valle, E.; Rosales-Hernández, 1191-1200.[http://dx.doi.org/10.7164/antibiotics.53.1191] [PMID:


M.C.; Abarca-Rojano, E.; Rojas-Hernández, S.; Fragoso-Vázquez, 11132966]
M.J.; Mendieta-Wejebe, J.E.; Correa-Basurto, A.M.; Vázquez-- [30] Peart, M.J.; Tainton, K.M.; Ruefli, A.A.; Dear, A.E.; Sedelies,
Moctezuma, I.; Correa-Basurto, J. N-(2′-Hydrox- K.A.; O’Reilly, L.A.; Waterhouse, N.J.; Trapani, J.A.; Johnstone,
yphenyl)-2-propylpentanamide (OH-VPA), a histone deacetylase R.W. Novel mechanisms of apoptosis induced by histone deacety-
inhibitor, induces the release of nuclear HMGB1 and modifies lase inhibitors. Cancer Res., 2003, 63(15), 4460-4471.[PMID:
ROS levels in HeLa cells. Oncotarget, 2018, 9(70), 12907619]
33368-33381.[http://dx.doi.org/10.18632/oncotarget.26077] [P- [31] Childs, B.G.; Baker, D.J.; Kirkland, J.L.; Campisi, J.; van
MID: 30279967] Deursen, J.M. Senescence and apoptosis: dueling or complemen-
[21] Hu, E.; Chen, Z.; Fredrickson, T.; Zhu, Y.; Kirkpatrick, R.; tary cell fates? EMBO Rep., 2014, 15(11),
Zhang, G.F.; Johanson, K.; Sung, C.M.; Liu, R.; Winkler, J. Clon- 1139-1153.[http://dx.doi.org/10.15252/embr.201439245] [PMID:
ing and characterization of a novel human class I histone deacety- 25312810]
lase that functions as a transcription repressor. J. Biol. Chem., [32] Peart, M.J.; Smyth, G.K.; van Laar, R.K.; Bowtell, D.D.; Richon,
2000, 275(20), V.M.; Marks, P.A.; Holloway, A.J.; Johnstone, R.W. Identifica-
15254-15264.[http://dx.doi.org/10.1074/jbc.M908988199] [P- tion and functional significance of genes regulated by structurally
MID: 10748112] different histone deacetylase inhibitors. Proc. Natl. Acad. Sci.
[22] Waltregny, D.; De Leval, L.; Glénisson, W.; Ly Tran, S.; North, USA, 2005, 102(10),
B.J.; Bellahcène, A.; Weidle, U.; Verdin, E.; Castronovo, V. Ex- 3697-3702.[http://dx.doi.org/10.1073/pnas.0500369102] [PMID:
pression of histone deacetylase 8, a class I histone deacetylase, is 15738394]
restricted to cells showing smooth muscle differentiation in nor- [33] Vanaja, G.R.; Ramulu, H.G.; Kalle, A.M. Overexpressed HDAC8
mal human tissues. Am. J. Pathol., 2004, 165(2), in cervical cancer cells shows functional redundancy of tubulin
553-564.[http://dx.doi.org/10.1016/S0002-9440(10)63320-2] [P- deacetylation with HDAC6. Cell Commun. Signal., 2018, 16(1),
MID: 15277229] 20.[http://dx.doi.org/10.1186/s12964-018-0231-4] [PMID:
[23] Balasubramanian, S.; Ramos, J.; Luo, W.; Sirisawad, M.; Verner, 29716651]
E.; Buggy, J.J. A novel histone deacetylase 8 (HDAC8)-specific [34] Korfei, M.; Stelmaszek, D.; MacKenzie, B.; Skwarna, S.; Chillap-
inhibitor PCI-34051 induces apoptosis in T-cell lymphomas. pagari, S.; Bach, A.C.; Ruppert, C.; Saito, S.; Mahavadi, P.;
Leukemia, 2008, 22(5), Klepetko, W.; Fink, L.; Seeger, W.; Lasky, J.A.; Pullamsetti, S.S.;
1026-1034.[http://dx.doi.org/10.1038/leu.2008.9] [PMID: Krämer, O.H.; Guenther, A. Comparison of the antifibrotic effects
of the pan-histone deacetylase-inhibitor panobinostat versus the
18256683]
IPF-drug pirfenidone in fibroblasts from patients with idiopathic
[24] Vannini, A.; Volpari, C.; Filocamo, G.; Casavola, E.C.; Brunetti,
pulmonary fibrosis. PLoS One, 2018,
M.; Renzoni, D.; Chakravarty, P.; Paolini, C.; De Francesco, R.;
13(11)e0207915[http://dx.doi.org/10.1371/journal.pone.0207915]
Gallinari, P.; Steinkühler, C.; Di Marco, S. Crystal structure of a
[PMID: 30481203]
eukaryotic zinc-dependent histone deacetylase, human HDAC8,
[35] Van Lint, C.; Emiliani, S.; Verdin, E. The expression of a small
complexed with a hydroxamic acid inhibitor. Proc. Natl. Acad. fraction of cellular genes is changed in response to histone hyper-
Sci. USA, 2004, 101(42), acetylation. Gene Expr., 1996, 5(4-5), 245-253.[PMID: 8723390]
15064-15069.[http://dx.doi.org/10.1073/pnas.0404603101] [P- [36] Glaser, K.B.; Staver, M.J.; Waring, J.F.; Stender, J.; Ulrich, R.G.;
MID: 15477595] Davidsen, S.K. Gene expression profiling of multiple histone
[25] Yang, Z.Y.; Perkins, N.D.; Ohno, T.; Nabel, E.G.; Nabel, G.J. The deacetylase (HDAC) inhibitors: defining a common gene set pro-
p21 cyclin-dependent kinase inhibitor suppresses tumorigenicity duced by HDAC inhibition in T24 and MDA carcinoma cell lines.
in vivo. Nat. Med., 1995, 1(10), Mol. Cancer Ther., 2003, 2(2), 151-163.[PMID: 12589032]
1052-1056.[http://dx.doi.org/10.1038/nm1095-1052] [PMID: [37] Mitsiades, C.S.; Mitsiades, N.S.; McMullan, C.J.; Poulaki, V.;
7489362] Shringarpure, R.; Hideshima, T.; Akiyama, M.; Chauhan, D.; Mun-
[26] Halsall, J.A.; Turan, N.; Wiersma, M.; Turner, B.M. Cells adapt to shi, N.; Gu, X.; Bailey, C.; Joseph, M.; Libermann, T.A.; Richon,
the epigenomic disruption caused by histone deacetylase inhibi- V.M.; Marks, P.A.; Anderson, K.C. Transcriptional signature of hi-
tors through a coordinated, chromatin-mediated transcriptional re- stone deacetylase inhibition in multiple myeloma: biological and
sponse. Epigenetics Chromatin, 2015, 8, clinical implications. Proc. Natl. Acad. Sci. USA, 2004, 101(2),
29.[http://dx.doi.org/10.1186/s13072-015-0021-9] [PMID: 540-545.[http://dx.doi.org/10.1073/pnas.2536759100] [PMID:
26380582] 14695887]
[27] Archer, S.Y.; Meng, S.; Shei, A.; Hodin, R.A. p21(WAF1) is re- [38] Dejligbjerg, M.; Grauslund, M.; Litman, T.; Collins, L.; Qian, X.;
quired for butyrate-mediated growth inhibition of human colon Jeffers, M.; Lichenstein, H.; Jensen, P.B.; Sehested, M. Differen-
cancer cells. Proc. Natl. Acad. Sci. USA, 1998, 95(12), tial effects of class I isoform histone deacetylase depletion and en-
6791-6796.[http://dx.doi.org/10.1073/pnas.95.12.6791] [PMID: zymatic inhibition by belinostat or valproic acid in HeLa cells.
9618491] Mol. Cancer, 2008, 7,
[28] Richon, V.M.; Sandhoff, T.W.; Rifkind, R.A.; Marks, P.A. His- 70.[http://dx.doi.org/10.1186/1476-4598-7-70] [PMID: 18789133]
tone deacetylase inhibitor selectively induces p21WAF1 expres- [39] K.; Marshall, B.L.; Hedglin, M.; Verdin, E.; Ulrich, S.M. Design
sion and gene-associated histone acetylation. Proc. Natl. Acad. and evaluation of ʻLinkerlessʼ hydroxamic acids as selective
Sci. USA, 2000, 97(18), HDAC8 inhibitors. Bioorg. Med. Chem. Lett., 2007, 17,
10014-10019.[http://dx.doi.org/10.1073/pnas.180316197] [PMID: 2874-2878.[http://dx.doi.org/10.1016/j.bmcl.2007.02.064]
10954755] [40] Dawson, M.A.; Kouzarides, T. Cancer epigenetics: from mech-
[29] Kim, Y.B.; Ki, S.W.; Yoshida, M.; Horinouchi, S. Mechanism of anism to therapy. Cell, 2012, 150(1),
cell cycle arrest caused by histone deacetylase inhibitors in human 12-27.[http://dx.doi.org/10.1016/j.cell.2012.06.013] [PMID:
carcinoma cells. J. Antibiot. (Tokyo), 2000, 53(10), 22770212]

You might also like