You are on page 1of 11

Biomedicine & Pharmacotherapy 103 (2018) 18–28

Contents lists available at ScienceDirect

Biomedicine & Pharmacotherapy


journal homepage: www.elsevier.com/locate/biopha

Phosphoethanolamine induces caspase-independent cell death by reducing T


the expression of C-RAF and inhibits tumor growth in human melanoma
model

Lisley I. Mambellia, , Sarah F. Teixeirab, Salomão D. Jorgea,c, Bárbara Kawamuraa,d,

Renato Menegueloe, José A.M. Barbutoa, Ricardo A. de Azevedoa,c, Adilson K. Ferreiraa,c,d,
a
Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
b
Department of Pharmacology, Institute of Biomedical Science, University of Sao Paulo, Sao Paulo, Brazil
c
Alchemy, Innovation, Research & Development, Department of Oncology, CIETEC/IPEN, University of Sao Paulo, Sao Paulo, Brazil
d
Medical Science, University of Sao Paulo Medical School, Sao Paulo, Brazil
e
Instituto Tecnológico da Aeronáutica, Sao Jose dos Campos, Sao Paulo, Brazil

A R T I C LE I N FO A B S T R A C T

Keywords: Phosphoethanolamine (PEA) is a fundamental precursor during the biosynthesis of cell membranes phospholi-
Phosphoethanolamine pids. In the past few years, it has been described as a potential antitumor agent. In previous studies, we de-
Caspase-independent cell death monstrated that PEA showed antitumor properties in vitro and in vivo in a wide range of tumor cell lines. Herein,
Antitumor properties we showed that PEA possesses cytotoxic properties and notably revealed to induce caspase-independent cell
Melanoma
death. Of interest, we provided evidence that PEA inhibits melanoma cells proliferation through the reduction of
C-RAF. Molecular docking of PEA evidenced that this compound indeed fits satisfactory in the binding site
located between the dimers of C-RAF protein with 107,01 Å and score of −29,62. Also, PEA arrested A2058 cells
at G2/M phase in the cell cycle. Moreover, cell proliferation, migration and adhesion capacities of A2058 cells
were also inhibited by PEA. Most importantly, PEA inhibited tumor growth of melanoma tumors and prolonged
survival rate of mice. Also, PEA induced a significant immune response in a syngeneic metastatic melanoma
model. Taken together, these data indicate that PEA is a promising candidate for future developments in cancer
field.

1. Introduction in the bone marrow. Accordantly, Pho-s induced in vivo cell death of
these immature cells in the spleen and liver, which is a notable effect.
The primary amine phosphoethanolamine is the central precursor of Recently, we demonstrated that Pho-s has a putative anti-metastatic
ethanolamine branch in the Kennedy pathway to produce phosphati- effect which was validated in a renal carcinoma model. Potentially,
dylethanolamine-phospholipids, which besides many functions, are also Pho-s inhibited lung metastasis in nude mice and blocked endothelial
related to the turnover of cell membranes [1,2]. These phospholipids cell proliferation, migration and tube formation, besides provoked cell
take part in the lipid signaling pathways and when hydrolyzed result in cycle arrest at the G2/M phase [6].
the formation of the second messengers [3]. Consequently, these mo- Metastatic malignant melanoma (MM) is a form of skin cancer
lecules can regulate the cell proliferation and survival through lipids which is highly aggressive. According to American Cancer Society,
signalling [4]. Recently, the therapeutic potential of Synthetic Phos- clinical treatments for this tumor type are based on systemic che-
phoethanolamine (Pho-s) was demonstrated in the acute promyelocytic motherapy by using chemotherapeutic drugs such as dacarbazine, pa-
leukemia (APL) using a transgenic mouse model harboring the PML/ clitaxel and cisplatin as standard therapy. Such strategies showed a
RARα fusion [5]. Pho-s showed anti-proliferative effects in APL by poor prognosis for patients with a response rate of only 10–20% [7,8].
blocking the dissemination of malignant clones and reducing the More recently, strategies based on melanoma biology understanding
number of CD117+ cells and Gr-1+ immature myeloid cells in the bone have been focused on blocking the often-activating mutation in the
marrow, spleen and liver. Importantly, Pho-s attenuated the expansion RAS-RAF-MEK-ERK signaling as a possible target for therapy [9–11].
of malignant clones CD34+/CD117+, as well as CD34+ and Gr-1+ cells The use of chemotherapy agents for the MM treatment and either their


Corresponding authors at: Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil.
E-mail addresses: lis_mambelli@yahoo.com.br (L.I. Mambelli), ferreira-kleber@usp.br (A.K. Ferreira).

https://doi.org/10.1016/j.biopha.2018.03.135
Received 28 November 2017; Received in revised form 22 March 2018; Accepted 22 March 2018
0753-3322/ © 2018 Elsevier Masson SAS. All rights reserved.
L.I. Mambelli et al. Biomedicine & Pharmacotherapy 103 (2018) 18–28

Fig. 1. Cytotoxic effect of PEA in A2058 cell line. (A) Chemical structure of PEA. MTT assay demonstrated that PEA is cytotoxic to A2058 cell line with IC50 value of
160 mM. (B) Colony forming units assay was used to analyze the capacity of PEA on reducing cell proliferation and diminish the formation of new colonies. In
accordance to MTT assay, IC50 values of PEA significantly reduced the capacity of A2058 cells to proliferate and form colonies. Paclitaxel, used as positive control,
showed to be more efficient on reducing the viability and the capacity of these cells to proliferate. (C) Panel of apoptotic and anti-apoptotic proteins analyzed by
western blotting. We demonstrated that PEA did not induce cleavage of caspases 3 and 8 and also PARP. Although, it is notable the alteration in the expression of
other important proteins, such as cytochrome c, BAD, and BCL-XL. (* p < 0.05; ** p < 0.001; *** p < 0.0001, relative to control). β-actin was used as total protein
loading control. Proteins quantification graph representing the values of each band normalized by the corresponding β-actin expression.

combination with radiotherapy has not demonstrated an impressed 2. Results


impact on patient’s survival. Thus, the development and the optimiza-
tion of new compounds, which could be new candidates for MM 2.1. PEA induces cytotoxic effect and caspase-independent cell death in
treatment are urgently required. Therefore, the main purpose of the human melanoma tumor cells
present work was to investigate the potential anti-melanoma effects of
O-Phosphorylethanolamine (PEA) in a human melanoma model. The cytotoxicity of PEA (Fig. 1A) was evaluated at different con-
centrations in comparison to paclitaxel, used as positive control. MTT

19
L.I. Mambelli et al. Biomedicine & Pharmacotherapy 103 (2018) 18–28

assay was performed to evaluate such cytotoxic effect. Human mela- A2058 cells. It was possible to note that when cultures were treated
noma cell line (A2058 cells) was incubated for 24 h with increasing with IC50 values of PEA (160 mM) or paclitaxel (10μM; positive con-
concentrations of PEA and paclitaxel. MTT reduction shows that PEA is trol) for 24 h, the capacity of cells of adhering to the matrix and to the
cytotoxic to A2058 (Fig. 1A) with IC50 value of 160 mM. In accordance plastic decreased significantly, when compared to control (non-treated
to MTT assay, we observed that PEA at IC50 values, reduced the ca- cells; Fig. 3A; *** p < 0.0001 and ** p < 0.001, respectively). In
pacity of A2058 cells to form colonies, when compared to paclitaxel contrast, when we used the same conditions previously mentioned,
used as control (Fig. 1B; ** p < 0.001). As expected, paclitaxel was both compounds significantly increased cell-cell interaction ability of
cytotoxic to A2058 cells with IC50 value of 10μM and also inhibited the A2058 cells (Fig. 3A). Notably, PEA was capable of significantly in-
capacity of A2058 cells of proliferating and forming new colonies crease cell-cell interaction of A2058, when compared to paclitaxel
(Fig. 1B; *** p < 0.0001). Of note, for colony-forming units experi- (positive control; Fig. 3A; *** p < 0.0001).
ment, we observed a slight increase of new colonies formed using The inhibitory effect of PEA on tumor cell migration, another crucial
40 mM of PEA (Fig. 1B; * p < 0.05), when compared to paclitaxel. step of melanoma skin cancer progression, was examined by the wound-
To further evaluate the molecular mechanisms underlying cell death healing assay. Approximately 80% of decreasing of A2058 cell migra-
by PEA, we investigated proteins related to apoptosis by western blot- tion was observed after 24 h of tumor cells incubation with IC50 value
ting. A2058 cells were treated for 24 h with 10μM of paclitaxel (positive of PEA, when compared to control (non-treated cells; Fig. 3B; *
control) and different concentrations of PEA (40, 80 and 160 mM). Our p < 0.05). The treatment with 10μM of paclitaxel for 24 h showed
results showed that PEA did not induce the cleavage of caspases 3 and similar result as observed for PEA (Fig. 3B; ** p < 0.001). However,
8. In accordance, PEA also did not cleavage the Poly (ADP-ribose) after 48 h of treatment with PEA, the “scratch” was completely closed
polymerase (PARP), a molecular sensor of DNA repair and programmed (Fig. 3B), while in the treatment with paclitaxel (positive control) the
cell death (Fig. 1C). Based on these findings, we propose that the cy- “scratch” remained opened.
totoxic effects of PEA are based on a caspase-independent manner. In- Of interest, we observed a significant decrease in the expression of
terestingly, PEA at the concentration of 80 mM down-regulated BCL-XL, p.C-RAF and p.ERK when cells were treated with IC50 value of PEA
an antiapoptotic protein of BCL-2 family. In agreement to the reduction (Fig. 3C). C-RAF is a well-known proto-oncogene that in combination
of BCL-XL, PEA upregulated the expression of BAD, an apoptotic with ERK forms a complex signaling pathway (RAF-MEK-ERK) re-
member of BCL-2 family. BAD is not capable of promoting apoptosis by sponsible for regulating proliferative, differentiation, and survival
itself, so it needs to be dimerized with different BCL-2 family members pathways of many tumors including melanoma. Also, it was possible to
into the conserved BH1 and BH2 domains. Thus, our data suggests that note an increase in the expression of A-RAF, when compared to control
the cell death induced by PEA in A2058 cells can be dependent on the (non-treated cells; Fig. 3C). We did not observe differences in the ex-
depletion of BCL-XL expression (Fig. 1C). pression of p.B-RAF as well as non-phosphorylated C-RAF in A2058 cell
line treated with different concentrations of PEA and with IC50 value of
2.2. Cell cycle arrest of A2058 cells in response to PEA treatment paclitaxel (Fig. 3C).
Having observed a significant decrease in the expression of C-RAF
To further investigate whether PEA exerts anti-proliferative effects protein by western blotting, we investigated whether PEA could sa-
on A2058 cells flow cytometry analysis was performed. Initially, A2058 tisfactory fits into C-RAF protein. Docking simulations were used to
cells were synchronized by deprivation of serum for 12 h. To reenter in investigate the possible binding site of PEA in C-RAF kinase (Figure 5D,
the cell cycle, serum was added and at this moment cells were treated 5E), the orientation and accommodation of such compound in the
with 10μM of paclitaxel (positive control) or IC50 value (160 mM) of mapped binding sites, as well as to calculate the affinity of PEA for each
PEA for 24 h. Cell cycle results revealed that the treatment with PEA at one (Gibbs interaction free energy, ΔG) with the formation of the en-
the concentration of 160 mM reduced the population of A2058 cells in zyme-ligand complex. The intermolecular interactions established
G0/G1 phase and increased the population of cells in G2/M phase, among PEA and amino acids of the interaction site were determined in
when compared to cells treated with IC50 value of paclitaxel the best model of dock simulation according to the ΔG (Fig. 5F).
(Fig. 2A,B). Our data indicates that PEA induces cell cycle arrest in G2/ The simulations showed that PEA fits well in the binding site located
M phase in A2058 cell line. The population of cells at Sub-G0/G1 phase between the dimers, in a binding site with 107,01 Å and score of
was not significant for this analysis. Such data was confirmed by wes- −29,62. The scores values varied from −7,57 to −29,62. PEA estab-
tern blotting when analyzed proteins associated to cell cycle. It was lished interactions with the amino acid residues of both dimers into the
observed a slight decrease in the expression of proteins associated to binding pocket through four hydrogen bonding interactions, via three
cell proliferation and expansion, when A2058 cell line was treated for oxygen atoms to the PEA side chain of the catalytic Arg401(a) residue
24 h with IC50 value of paclitaxel (10μM; positive control) and PEA and Trp342 residue of both dimers (a and b), and via protonated ni-
(160 mM). Proteins as p.RB, which besides many properties is re- trogen of amine on the oxygen of Arg398(a) residue.
sponsible for G1 checkpoint, blocking S-phase entry and consequently
cell growth; and p.CDC2, mainly responsible for the entry of eukaryotic 2.4. Up-regulation of p.STAT-3 induced by PEA leads to the decrease of
cells into mitosis were also downregulated when compared to control MEK1/2 expression
(non-treated cells). It was noted a minimal increase in the expression of
p.P38, which can be in response to stress stimuli. Importantly, we ob- We demonstrated that PEA did not induce caspase-dependent cell
served a significant reduction in the expression of cyclin proteins, such death in A2058 cell and fits well into C-RAF binding site. Then, we
as cyclin D1 and D3 and cyclin-dependent kinases, CDK 4 and 6 investigated if PEA could induce cell death trough RAS/RAF/MEK/ERK
(Fig. 2C). Taken together, our data indicates that PEA impair A2058 cell signaling/pathway. It is important to note that this pathway plays a role
division and proliferation by arresting cells at G2/M phase. in melanoma initiation, maintenance and progression [10,11]. It has
been previously described that the inhibition of STAT-3 can be directly
2.3. PEA reduces A2058 cell mobility and fits in silico into the C-RAF related to MEK regulation [12], as well as that STAT-1 activation can
binding site modulate MEK1 [13], then we focused on evaluating such proteins.
First, it was examined, by flow cytometry, the expression of p.STAT-
In order to evaluate the effect of PEA on interfering in A2058 cells 3 in A2058 cell line with or without the treatment with PEA and pa-
capacity of adhering to plastic, or to a matrix or interfering in cell-cell clitaxel. Further, we analyzed the expression of such protein while
interaction, adhesion assay was performed. Indeed, PEA demonstrated using an inhibitor of STAT-3 and its association with both compounds.
similar effects of paclitaxel (positive control) on adhesion ability of Cucurbitacin (JSI-124) a well-known inhibitor of JAK/STAT-3 pathway

20
L.I. Mambelli et al. Biomedicine & Pharmacotherapy 103 (2018) 18–28

Fig. 2. PEA arrested A2058 cells at G2/M phase of the cell cycle. (A) Representative histograms are indicating selected gates of flow cytometry analysis of A2058 cell
line treated with paclitaxel or PEA. The first histogram represents the control (non-treated cells). The second one represents the paclitaxel treatment, used as positive
control. We can note that A2058 cells were arrested in G0/G1 phase. The last histogram is representative of PEA treatment, indicating the accumulation of cells in
G2/M phase. (B) Cell cycle analysis demonstrated that PEA significantly induced cell cycle arrest of A2058 cells in G2/M phase, while paclitaxel provoked the
accumulation of cells in G0/G1 phase. (C) Panel of proteins analyzed by western blotting indicates the upregulation of p.RB, when A2058 cells were treated with 40
and 80 mM of PEA, but no significative expression alteration was observed when these cells were treated with IC50 value of this compound. Also, we note a
significant increase in the expression of p.CDC2, when cells were treated with the same concentrations above. The increase in the expression of p.P38 was also
observed, when cells were treated with different concentrations of PEA, including IC50 value. β-actin was used as total protein loading control. Proteins quantifi-
cation graph representing the values of each band normalized by the corresponding β-actin expression. (* p < 0.05; ** p < 0.001; *** p < 0.0001, relative to
control).

was used. After the pre-treatment for 12 h with cucurbitacin, it was serum-free solution. When tumors reached 100 mm3, the animals
noted a considerable amount of non-viable cells in culture, even when started to be treated according to their experimental group: control,
treated with only this inhibitor. Notably, when IC50 values of paclitaxel PEA or vemurafenib. These animals were treated daily for 10 days and
(10μM) and PEA (160 mM) were added to the pre-treated culture, more then they were euthanized when tumors reached a maximum of 2 cm3.
cells were observed (data not shown). Interestingly and not expected, Such experiment allowed us to validate in vivo therapeutic effec-
flow cytometry analyses demonstrated that JSI-124 significantly in- tiveness of PEA as a potential new candidate antitumor agent. The
creased the expression of p.STAT-3, when compared to control (non- antitumor effect of PEA was compared to vemurafenib (used as positive
treated cells; Fig. 4A; *** p < 0.0001). Moreover, the combination of control), a well-known chemotherapeutic agent used for non-treated
cucurbitacin with paclitaxel or PEA considerably increased the ex- melanoma. Analyzing the survival rate, we noted that both compounds
pression of p.STAT-3 (Fig. 4A; *** p < 0.0001) even when compared PEA and vemurafenib were efficient on increasing lifespan on the two
with both compounds without the inhibitor (Fig. 4A, * p < 0.05 (pa- studied models (Fig. 5A). PEA showed to be more efficient on enhan-
clitaxel) and ** p < 0.001 (PEA)). Of note, the basal expression of cing survival rate in human model, than in murine model (Fig. 5A) but
p.STAT-3 in A2058 cells is not high (Fig. 4A), indicating that STAT-3 is only vemurafenib was capable of significantly inhibiting tumor growth
not constitutively activated in this cell line. along the treatment in human model injected with A2058 cells (Fig. 5B,
Finally, we evaluated by western blotting the expression of STAT-1 * p < 0.05). However, PEA significantly inhibited tumor growth in
and MEK1/2 and also their phosphorylated forms on A2058 cells C57BL/6 injected with B16F10 cells when compared to control (ani-
treated with IC50 value of paclitaxel or with PEA (40, 80, 160 mM). We mals treated with placebo) and vemurafenib (positive control; Fig. 5B; *
showed that IC50 value of PEA (160 mM) reduced the expression of p < 0.05). Taken together, our data demonstrates that PEA is capable
STAT-1 and p.STAT-1, as well as MEK1/2 and its phosphorylated form, to inhibit the tumor growth in B16F10 model and prolong survival rate
when compared to control (non-treated cells; Fig. 4B). of mice in both human and murine melanoma models.

2.5. PEA inhibits tumor growth and increases survival rate in melanoma 2.6. Antitumor activity of PEA in B16F10 animal model is immune-system
models dependent

To evaluate the antitumor effect of PEA in vivo, we used two mel- Since we obtained a promising data on increasing survival rate and
anoma models: first, a human melanoma model in which female BALB/ reducing tumor growth, we studied whether PEA could affect the im-
c nu/nu mice were injected with 5 × 106 of A2058 cells diluted in 50% mune system in vivo. Herein, we propose a therapeutic protocol using
matrigel solution and; second, a model performed in female C57BL/6 bone marrow-derived dendritic cells previously described by Figueiredo
mice injected with the same amount of B16F10 cells in RPMI-1640 and colleagues [14]. The dendritic cells were first incubated ex vivo

21
L.I. Mambelli et al. Biomedicine & Pharmacotherapy 103 (2018) 18–28

(caption on next page)

with PEA in combination with a melanoma cell lysate and then adop- 3. Discussion
tively transferred into C57Bl/6 mice with lung metastases of B16F10
cells. We observed that such combination of treatment significantly Herein, we demonstrated that PEA is capable of inducing caspase-
protected the animals (Fig. 6A; ** p < 0.001) and lead us to suggest independent cell death in A2058 human melanoma cell line. According
that PEA stimulated DCs on decreasing the number of metastatic no- to our data, PEA was efficient on significantly decrease the expression
dules in this model (Fig. 6B). of C-RAF and ERK proteins when cells were treated with IC50 value of
this compound. Moreover, we showed in silico that PEA indeed fits into

22
L.I. Mambelli et al. Biomedicine & Pharmacotherapy 103 (2018) 18–28

Fig. 3. Effects of PEA on cell mobility of A2058 and in silico molecular docking. (A) Cell adhesion assay was used to evaluate the capacity of PEA on interfering at
metastatic ability of A2058 cells. PEA significantly decreased the capacity of A2058 cells of adhering to the matrix and to the plastic, but increased cell-cell
interaction ability when treated with IC50 values. (B) Migration area examined by phase contrast microscopy at 0, 24 and 48 h (magnification, ×100). PEA
significantly delayed the repopulation of “scratch” area, demonstrating similar effects of paclitaxel, used as positive control, in 24 h of treatment. (C) Panel of proteins
analyzed by western blotting demonstrated a significant decrease of p.C-RAF and p.ERK. Other important proteins, as p.BRAF, C-RAF and ARAF were also analyzed,
but no significant alteration concerning PEA treatment was observed. β-actin was used as total protein loading control. Proteins quantification graph representing the
values of each band normalized by the corresponding β-actin expression. (* p < 0.05; ** p < 0.001; *** p < 0.0001, relative to control). Having demonstrated that
PEA could modulate the expression of phosphorylated C-RAF, the main component of RAS/RAF/MEK/ERK signaling/pathway, we performed in silico docking of PEA
and observed that this compound fits well in the C-RAF protein domain. (D) View of crystal structure of C-RAF homodimer. (E) Mapped binding sites (green bubbles)
on the molecular surface of C-RAF homodimer, considering regions larger than 20 Å. Circle represents the binding site with the best score in docking simulations; (F)
Schematic representation for the molecular docking of PEA in the mapped binding site of C-RAF using the CLC Drug Discovery Workbench program. PEA is displayed
as stick model where the carbon atoms are represented in gray, nitrogen in blue, oxygen in red, hydrogen in white and phosphorus in yellow. The amino acid residues
(Arg398(a), Arg401(a), Trp 342(a), Trp342(b)) in the binding site are also shown as stick model (For interpretation of the references to colour in this figure legend,
the reader is referred to the web version of this article).

Fig. 4. Regulation of p.STAT-3 induced by PEA on MEK1/2 signaling pathway. STAT-3 inhibitors have been described as potential targets to promote resistance
phenotypes in melanoma cells and other tumor types, by modulating RAS/RAF/MEK/ERK pathway. (A) A2058 cells treated with JSI-124, paclitaxel or PEA were
analyzed by flow cytometry. Such analysis demonstrated that the basal expression of p.STAT-3 protein in A2058 cells is not high, but after the pre-treatment for 12 h
with cucurbitacin, this protein expression significantly increased. Also, the combination of cucurbitacin with paclitaxel or PEA considerably increased the expression
of p.STAT-3, when compared with both compounds without the inhibitor (B) Panel of proteins analyzed by western blotting indicated the reduction in the expression
of STAT-1 also. Corroborating with previous data, the expression of MEK1/2 was considerably reduced when cells were treated with IC50 value of PEA (* p < 0.05;
** p < 0.001; *** p < 0.0001, relative to control). β-actin was used as total protein loading control.

23
L.I. Mambelli et al. Biomedicine & Pharmacotherapy 103 (2018) 18–28

Fig. 5. Antitumor activity in vivo of PEA. We evaluated the antitumor effect of PEA in vivo in two melanoma models: a human melanoma model performed in
immunocompromised mice injected with A2058 cells; and a murine model that was performed in female C57BL/6 mice injected with B16F10 cells. (A) The analysis
of survival rate showed that both PEA and vemurafenib (used as positive control) were efficient on prolonging lifespan in human and murine melanoma models.
Mostly important, PEA showed to be more efficient than vemurafenib on improving survival rate in human model. (B) Vemurafenib was capable of significantly
inhibiting tumor growth in human model, while PEA significantly inhibited tumor growth in murine model. (* p < 0.05, relative to control).

Fig. 6. PEA can act in combination with the immune system to promote metastasis regression in syngeneic murine melanoma model. Bone marrow-derived dendritic
cells were used in combination with PEA in order to test their capacity in reducing lung metastases provoked by B16F10 cells in C57Bl/6 mice. (A) Therapeutic
activity of dendritic cells stimulated by PEA and tumor cells lysate significantly protected the animals and decreased metastatic sites. (B) Significant regression of
metastatic lung nodules is shown demonstrating that PEA may act in combination with the immune system. (** p < 0.001, relative to control).

C-RAF protein binding site with a score of -29,62 kcal/mol. Taken to- cell proliferation and regulating differentiation and survival of A2058
gether, our main results lead us to suggest that the mechanisms in- cells. Of note, lots of efforts have been done to develop antitumor
volved in this cell death process could be related to RAS/RAF/MEK/ agents that could target key constituents of RAS/RAF/MEK/ERK
ERK signaling/pathway. Most importantly, we showed that PEA pro- pathway. Surely, PEA represents an important drug candidate to target
longs the lifespan of mice-developed human and murine melanomas, this pathway.
besides of inhibiting tumor growth in murine model. Synthetic phosphoethanolamine (Pho-s) has been considered a po-
Mitogen-activated protein kinase (MAPK) pathway is appointed as tential antitumor agent, and it has been shown that its cytotoxic effect
RAS/RAF/MEK/ERK cascade and can be considered a well-known and acts selectivity against tumor cells and could be used for the treatment
deeply studied cell signaling/pathway. This pathway is commonly of a wide range of tumors [1,2,5,6,15,16]. At current study, we tested
dysregulated not only in melanoma but also in a wide variety of tumor the effect of a commercial molecule of PEA in melanoma A2058 cells. In
types [10]. Importantly, PEA can impair the activation of this pathway accordance to what was previously demonstrated for different cell lines
and lead as to assume that this compound is mediating the reduction of using Phos-s, the commercial compound (PEA) also exhibited cytotoxic

24
L.I. Mambelli et al. Biomedicine & Pharmacotherapy 103 (2018) 18–28

effect on A2058 cells. PEA not only reduced viability but also blocked responsible for preventing decontrolled cell proliferation, by the in-
the capacity of A2058 cells of forming new clones, which in turn is hibition of cell cycle progression. When this protein is phosphorylated,
considered an important step for tumor development. The in vitro ef- it becomes inactive and allows cells to divide. Herein, we observed a
fects observed by PEA treatment may suggest that this compound can significant increase in the expression of such phosphorylated protein,
reduce tumor cells invasion toward different sites [17]. In accordance when A2058 cells were treated with 40 and 80 mM of PEA, but inter-
to this, we previously demonstrated that Pho-s is capable of reducing esting this expression pattern was not confirmed when the IC50 value of
the proliferative capacity of cells in an acute promyelocytic leukemia PEA was used. More interestingly, it was possible to note a significant
(APL) model [5]. alteration in the expression of p.CDC2 protein when these cells were
Interestingly, at low concentrations PEA provoked the opposite ef- treated with the same concentration of this compound. CDC2, also
fect on A2058 cells proliferation and capacity of forming new colonies. known as cyclin-dependent kinase 1 (CDK1) is a cell cycle key protein
Compared to control, the concentration of 40 mM of PEA significantly responsible for regulating the progression of cell cycle. When phos-
increased the capacity of these cells to divide. We reasonably start out phorylated, this protein forms complexes responsible to phosphorylate
from the assumption that there is no cytotoxicity at low concentration important substrates, which will allow the entry of cell in S phase of cell
of a compound, then as the concentration increases, the cell kill is cycle, thus permitting cell division. According to the literature, the
proportional to drug concentration until it reaches a plateau at high increase of p.CDC2 indicates that cells were accumulated in G1/S phase
concentrations [18]. of cell cycle [21]. Considering our data which showed that PEA arrested
Recently, it was proposed a hormesis model which claims that is A2058 cells at G2/M, we suggest that p.CDC2 increased significantly as
necessary to consider the mechanisms acquired by organisms to de- a reverse mechanism of the cell as an unsure to stimulate cell division
toxify chemicals and then being resistant to drugs [19]. In respect to and avoid cells death. Of interest, A2058 cells were not able to reenter
such data, it has been described that breast cancer cell lines exposed to the cell cycle and were arrested in G2/M phase. We also note an in-
low concentrations of an experimental compound also showed opposite crease in the expression of p.P38, which is a kinase from the class of
effects [20]. On the other hands, PEA at sub-toxic concentration may mitogen-activated protein kinases (MAPK) that are responsive to stress
enter in a growth-stimulating zone and induces cell proliferation. stimuli, in cell treated with IC50 value of PEA. Such data could be
Contrary to expectations of what we showed in previous studies considered a response to such stress stimuli induced by PEA in A2058
using Pho-s [15,16], here, for the first time, we demonstrated that PEA cell line.
did not induce caspase-dependent cell death. Notably, we observed Interestingly, we demonstrated that PEA exhibits similar effect of
alterations in the expression of important apoptotic proteins, such as paclitaxel when considering the inhibition of A2058 cells of adhering to
cytochrome c and BAD, as well as BCL-XL an anti-apoptotic protein. a matrix or a substrate (plastic). The most important and relevant data
Although the expression of BAD was similar to the control (non-treated of this finding is that PEA can reduce the capacity of A2058 cells to pass
cells) when IC50 of PEA was used, it is important to highlight that this through the extracellular matrix. The cell adhesion assay is often used
protein increased significantly when cells were treated with low values to assess the metastatic ability of a specific tumor cell population, as
of PEA. In accordance to such data, the expression of anti-apoptotic well as, to test whether a compound may interfere in such ability [22].
protein BCL-XL significantly decreased compared to control (non- Our finding suggests that PEA can prevent metastasis, which is one of
treated cells), including IC50 value. Both BAD and BCL-XL are im- the reasons for the resultant mortality of patients with cancer [23,24].
portant proteins of BCL-2 family, which are in mitochondria and are In this study, PEA was found to trigger the increase of cell-cell inter-
responsible for controlling programmed cell death, such as apoptosis action properties of A2058 cells. Of note, PEA was more efficient on this
and autophagy. An increase of BAD, a pro-apoptotic member of BCL-2 role than paclitaxel. Regarding melanoma and metastasis progression,
family induced by PEA can be responsible for initiating cell death in such feature exerted by PEA may be valuable if it is considered that the
A2058 cells. The increase of BAD can blocks BCL-XL activity and pre- initially main property to be lost for cell invasion is the ability of a cell
vent the release of cytochrome c, then avoiding cell death. Thus, BAD to adhere to its neighbored cells [17].
increase in response to PEA treatment indicates that PEA provokes the It was also analyzed whether PEA would be able to interfere at
formation of a mitochondrial membrane pore which in turn initiate the migration capacity of A2058 cells. Notably, PEA significantly delayed
apoptotic cascade in A2058 cells. the repopulation of “scratch” area, corroborating with previous data of
Notably, PEA did not induce the cleavage of proteins that are re- adhesion analyses. Moreover, proteins that are known to be important
presentative indicators of caspase-dependent cell death, even following for melanoma tumor cells division, maintenance and progression were
the treatment with different concentrations, including IC50 value of also investigated. Most importantly, there was an expressive reduction
PEA. Herein, we did not observe the cleavage of caspase 3 or 8. Indeed, in the expression of p.C-RAF protein, and consequently of p.MEK1/2
it was not possible to detect the presence of cleaved PARP. The com- and p.ERK.
bination of such data, the deregulation of BCL-XL and the increase of Herein, we observed that PEA significantly decreased p.C-RAF ex-
BAD lead us to suggest that PEA induced cell death in a caspase-in- pression, the main component of MAPK pathway. Thus, the inhibition
dependent manner in A2058 cells. of C-RAF phosphorylation by PEA leads to the decrease of ERK1/2
One interesting finding is that PEA showed to possess an inhibitory phosphorylation in A2058 cells. Importantly, when p.C-RAF is activated
effect on A2058 cells proliferation by inducing cell cycle arrest in G2/M it is capable of modulating different substrates than those directly in-
phase. Some of our data on the expression pattern of proteins related to volved in MAPK cascade, notably one of these such important targets
cell cycle, such as cyclin proteins and cyclin-dependent kinases besides that was increased by PEA was the pro-apoptotic protein BAD [25].
of p.RB and p38, corroborated with such result. Cyclins are the main Indeed, cell cycle arrest of A2058 cells in response to PEA treatment can
proteins responsible for controlling the progression of cells division. also be related to the inhibition of C-RAF phosphorylation and the re-
These proteins act by triggering cyclin-dependent kinases and forming duction of MEK activation, which is capable to regulate the cell cycle
complexes which are capable to control the progression of specific progression and consequently cell division. One unanticipated finding
phases of the cell cycle. Herein, we observed the reduction of the ex- was that PEA has induced cell death in A2058 cells by the reduction of
pression of cyclins D1 and D3 and consequently the cyclin-dependent C-RAF activation, and leads to the increase of BAD expression in a
kinases 4 and 6 which are activated by the cyclin D family. This family mechanism independent of caspase cleavage.
of cyclins are known to be related to the progression of G1 to S phase, Additionally, as previously mentioned, the pro-apoptotic protein
then the reduced signaling to the progression of G1 to S phase can BAD also showed to be downregulated when the same cell line was
explain the arrest in G2/M phase of A2058 cells treated by PEA. treated with IC50 value of paclitaxel and PEA. Notably, when the mo-
Retinoblastoma protein (p.RB) is a tumor suppressor protein, which is lecular docking of PEA was performed in silico, we noted that this

25
L.I. Mambelli et al. Biomedicine & Pharmacotherapy 103 (2018) 18–28

compound established intermolecular interactions with the amino acids demonstrating that this compound may act in combination with the
of the interaction site of C-RAF protein. This analysis showed that PEA immune system.
fits well in the binding site located between the dimers of the protein, in Herein, we presented for the first time, new data that PEA is capable
a binding site with 29,62 kcal/mol of score. All these data together, lead of inducing caspase-independent cell death in A2058 cells by reducing
us to suggest that PEA could induce cell death in A2058 cell line by C-RAF expression and modulating RAF/MEK/ERK signaling/pathway.
modulating RAS/RAF/MEK/ERK pathway. Nevertheless, further analyses are required to clarify the antitumor
Having demonstrated that PEA could lead cell death by the mod- mechanisms of action in more details. Most importantly, PEA showed
ulation of RAS/RAF/MEK/ERK cascade, we attempt to investigate if therapeutic effects in animal melanoma models. Thus, PEA represents a
important proteins indirectly related to such signaling/pathway could potential prototype as a new chemotherapeutic agent for the treatment
be altered. It has been previously described that STAT-3 inhibitors of untreated melanoma.
could mediate resistance phenotypes not only in melanoma cells but
also in other tumor types, by modulating RAS/RAF/MEK/ERK pathway 4. Methods
[12,26]. STAT-3 is a transcription factor that although plays an im-
portant role during normal cell development, usually is constitutively 4.1. Chemical and reagents
activated in almost 70% of solid and hematological tumors, thus in-
hibitors of this protein appeared as a novel target for cancer drug dis- O-Phosphorylethanolamine (cat.27640) was purchased from Sigma-
covery [27,28]. We showed that A2058 cells possess low expression Aldrich (St. Louis, MO, USA) and stock solution was diluted in phos-
levels of p.STAT-3, indicating that STAT-3 is not constitutively acti- phate buffered saline (PBS) at a concentration of 0.6 M. The pH of the
vated in this cell line. But interestingly, when cells are treated with solution was adjusted to 7.2 with sodium hydroxide (NaOH) and then
cucurbitacin, a natural selective inhibitor of p.STAT-3, there is a sig- stored at 4 °C. For in vivo and in vitro tests PEA was again diluted in PBS-
nificant increase of this protein expression. Although this inhibitor vehicle or directly in the basal medium, respectively. Paclitaxel (cat.
alone provoked cell death on A2058 population, the combination with T7191) was purchased from Sigma-Aldrich and diluted in dimethyl
PEA significantly increased the expression of p.STAT-3. Cichowski and sulfoxide (DMSO) according to manufacture’s instructions.
Jänne mentioned that some inhibitors, such as some of RAF enzymes, Cucurbitacin I (JSI-124) was purchased from Sigma-Aldrich, as well as
could either suppress or activate the same signaling pathway [29]. The Thiazolyl Blue Tetrazolium Bromide (3-(4,5-dimethylthiazol-2-yl)-2,5-
precise mechanism by which some inhibitors could contrarily act is diphenyltetrazolium bromide; MTT), Propidium Iodide (PI) and RNase.
unknown, but provides a warning note for targeting anticancer drug
discovery. 4.2. Cell culture
In previous studies developed by our group, Pho-s showed in vivo
anti-melanoma properties [3] and anti-metastatic effect in a renal The A2058 (human melanoma; CRL-11147) and B16F10 (murine
carcinoma model6. In this model, Pho-s inhibited lung metastasis in melanoma; CRL-6475) cell lines were obtained from American Type
nude mice and blocked endothelial cells proliferation, migration and Culture Collection (ATCC). All cell lines were routinely maintained in a
tube formation, besides provoked cell cycle arrest at the G2/M phase. basal medium composed by RPMI-1640 (Sigma-Aldrich) supplemented
Herein, we evaluated the antitumor effect of PEA in vivo in two with 10% (v/v) fetal bovine serum (FBS) (Gibco, Karlsruhe, Germany),
melanoma models: a human melanoma model in which female BALB/c 100U/ml penicillin and 100 μg/ml streptomycin at 37 °C under a hu-
nu/nu mice were injected with A2058 cells; and a second model that midified atmosphere of 5% CO2.
was performed in female C57BL/6 mice injected with B16F10 cells. We
showed that PEA triggered an important therapeutic effect in both 4.3. Cell viability assay
melanoma models in vivo. Considering the survival rate, it was observed
that both compounds PEA and vemurafenib (used here as positive The MTT cell viability assay was conducted according to manu-
control) were efficient on increasing lifespan in human and murine facturer’s instructions (Sigma-Aldrich). A2058 cells were seeded in
melanoma models. Most important, PEA showed to be more efficient triplicate into 96-well plates at a density of 1 × 104 cells/100 μL/well
than vemurafenib on improving survival rate in human model. On the and allowed to attach overnight. Cells were then treated with different
other hand, only vemurafenib was capable of significantly inhibiting concentrations of PEA (from 4.69 to 300 mM) or paclitaxel and in-
tumor growth in such model. It is important to highlight that PEA cubated for 24 h. Cell viability was monitored by the MTT solution at a
significantly inhibited tumor growth in murine model, while vemur- final concentration of 5 mg/mL. Then, the medium was discarded,
afenib was not efficient on inhibiting tumor growth. Vemurafenib is a DMSO was added and absorbance was measured in a plate reader
well-known BRAF enzyme inhibitor used for non-treated melanoma and (Thermoplate Q4 TP Reader, Tokay Hit, Japan) at 540 nm. The data
part of our results, such as the fact that this chemotherapeutic was ef- obtained were further analyzed using Prism nonlinear regression soft-
ficient on inhibiting tumor growth in human model, could be explained ware (GraphPad Software, Inc.) for curve fitting and the determination
by this feature. As previously mentioned, the human model consisted in of absolute IC50 values.
immunocompromised mice injected with A2058 cells. This cell popu-
lation is known to be a BRAF-mutant cell line and then is expected that 4.4. Cell cycle analysis
vemurafenib would be the most efficient on regretting tumor growth.
Taking into consideration that PEA was capable of prolonging sur- The A2058 cell line was seeded into 6-well plates at a density of
vival rate in both animal models proposed here, but only in the murine 1 × 104 cells/well, synchronized by deprivation of serum for 24 h and
model showed to be efficient on delaying tumor growth, while in im- induced to reenter the cell cycle by the subsequent addition of serum.
munocompromised animals had no effect, we decided to explore whe- Then, cells were treated for 24 h with PEA or paclitaxel at concentra-
ther the immune system could be associated to this effect. Herein, we tions of 160 mM and 10μM, respectively. Next, cells were collected and
used bone marrow-derived dendritic cells, previously incubated ex vivo fixed with cold 70% ethanol and stored at -20 °C for 24 h. Cells were
with PEA and melanoma cell lysate, to test their adoptive transfer into washed and resuspended in PBS for being incubated at 37 °C for 15 min
C57Bl/6 mice with lung metastases provoked by B16F10 cells. with 10 mg/ml RNase and 1 mg/ml PI. After, they were washed and
Interestingly, we observed that this combination of treatment sig- resuspended in PBS for analysis. A total of 10.000 cells per analysis
nificantly protected the animals and lead us to suggest that PEA was were examined by flow cytometry (FACSCalibur) and analyzed using
capable to stimulate the dendritic cells to decrease the number of me- Modfit LT software (Verity Software House, Topsham, ME). Data re-
tastatic nodules in this syngeneic metastatic melanoma model, present the mean ± SD of three independent experiments.

26
L.I. Mambelli et al. Biomedicine & Pharmacotherapy 103 (2018) 18–28

4.5. Colony forming units assay 4.10. In vivo preclinical studies

To analyze the capacity of A2058 cell line to form new colonies All animal experiments were conducted in agreement with ethical
when treated with 160 mM PEA or 10μM paclitaxel, 102 cells were principles for animal research and approved by the ethics committee for
plated in triplicate in 6-well culture plates. The cultures, both experi- animal research of the University of Sao Paulo, Sao Paulo, SP, Brazil
mental and control were maintained until day 15 and the medium was (process number 31/2014). Females BALB/c nu/nu, 6- to 8-week old
changed every two or three days. After 15 days, cells were fixed with were injected subcutaneously with a suspension of A2058 cells
4% paraformaldehyde for 2 h and stained by crystal violet. (5 × 106/cells per animal) in a 50% growth factor-reduced Matrigel
(BD Biosciences, Franklin Lakes, NJ) solution. These experiments were
also conducted using female C57BL/6 mice, 6- to 8-week old, which
4.6. Cell lysate and Western Blot analysis
were injected subcutaneously with a suspension of B16F10 cells
(1 × 106/cells per animal) in RPMI-1640 serum-free solution.
Lysates were prepared from A2058 cell line treated with different
Melanoma tumors were allowed to reach 100mm3 and randomized
concentrations of PEA (40, 80 or 160 mM) or 10μM paclitaxel with lysis
(n = 5 per group) into control (vehicle solution only), PEA 100 mg/kg
buffer containing protease and phosphatase inhibitors. Briefly, cells
or vemurafenib 5 mg/kg groups. All groups were treated daily for 10
debris were removed by centrifugation at 12.000 rpm for 5 min, and
days, tumors were measured with a manual caliper every 2 days and
after protein separation by gel electrophoresis, the proteins were
volumes were calculated using the formula: tumor weight
transferred to PVDF membranes. Membranes were then blocked with
(mg) = [length (mm) × width2 (mm2)]/2. The tumor doubling time,
5% bovine serum albumin (BSA, Sigma-Aldrich) in TBS-Tween 20 so-
i.e. the time for a tumor to double in volume and tumor growth delay,
lution for 1 h, followed by overnight agitating incubation with appro-
that was the difference between the time required for tumors in treated
priate primary antibodies obtained from cell signaling at 4 °C. Finally,
and untreated animals to reach at least 1cm3, was evaluated. Animals
membranes were incubated for 1 h with HRP-conjugated secondary
were euthanized when tumors reached a maximum of 2cm3. Then, the
antibodies. Detection was by WesternSure ECL Substrate (Li-Cor,
tumors were surgically excised and masses were recorded.
Lincoln, NE, USA). Images were acquired by ChemiDoc MP System (Bio-
Rad, California, USA) and the blots were normalized for β-actin in each
4.11. Bone marrow-derived dendritic cells (BMDCs) and adoptive transfer
lane.
therapy

4.7. Cell adhesion assay In order to evaluate whether PEA could act in combination with the
immune system to prolong survival rate in animal models, we per-
The binding capacity of A2058 cell line when treated with 160 mM formed adoptive transfer analysis. BMDCs were obtained from femurs
PEA and 10μM paclitaxel was tested. In order to analyze the interaction of C57Bl/6 mice after euthanasia. These femurs were flushed with
among A2058 cell line and the plate surface, the Geltrex Matrix RPMI-1640 medium and red blood cells were lysed for 5 min. The cells
(Thermo Fisher, Massachusetts, USA) and itself, 104 cells were seeded from each femur were suspended in 20 mL of basal medium supple-
in 96-well culture plate. First, Geltrex Matrix was prepared by in- mented with 50 ng/mL of GM-CSF and 25 ng/mL of IL-4 (BD
cubation for 30 min at 37 °C in 5% CO2 humidified chamber. At the Bioscience) and incubated in a Petri dish at 37 °C with 5% CO2. After
same time, a new culture plate was prepared with 105 cells, which were five days in culture, part of the medium was replaced by fresh medium
allowed to adhere for 2 h to test cell-cell interaction. Next, the same with GM-CSF. Non-adherent blood cells were discarded and adherent
quantity of cells (104 cells/well) was plated on top of Geltrex, or on top cells were used in the experiment. Dendritic cells were pulsed with the
of previously adhered cells, or directly onto the bottom of the plate. At lysate of a subcutaneous B16F10 tumor in the proportion of 10:1 (10 of
this moment, each treatment was added to the corresponding well. Cells dendritic cells and 1 of tumor cells lysate (L)) and with 100 mM of PEA.
were treated for 24 h, then the supernatants were individually cen- Then, mature dendritic cells (maDCs) were used for adoptive transfer in
trifuged and the number of viable cells was counted by using trypan C57BL/6 syngeneic mice, with previously developed-melanoma by the
blue solution. injection of 5 × 105 B16F10 cells. After seven days of the adoptive
transfer, animals were treated with 5 × 105 maDCs and separated in
two groups (n = 5 per group): Control (vehicle + L) and experimental
4.8. Migration (wound-healing) assay
(100 mM PEA + L). On the day 14, mice were euthanized and meta-
static lung nodules were quantified.
To evaluate the capacity of A2058 cell line to migrate in vitro under
specific treatment, 160 mM PEA and 10μM paclitaxel was used. For this
4.12. Molecular docking of PEA
experiment, 5 × 105 cells were plated in 24-well culture plates until
they reached total confluence. At this point, the “wound” was per-
A molecular docking approach was performed to predict a binding
formed and the corresponding treatment added. Pictures were obtained
mode for PEA on C-RAF kinase. The C-RAF co-crystallized structure
at the moment the wound was performed and after 24 and 48 h.
bounded to the (1E)-5-(1-piperidin-4-yl-3-pyridin-4-yl-1H-pyrazol-4-
yl)-2,3-dihydro-1H-inden-1-one oxime inhibitor (SM5), entry code
4.9. Expression of p.STAT-3 by flow cytometry 3OMV at 4.0 Å resolution [32] was retrieved from Brookhaven Protein
Data Bank (PDB) [33] and used as geometric reference for constructing
The A2058 cell line was seeded into 6-well culture plates and pre- the biomacromolecule model.
treated for 12 h with 5μM of Cucurbitacin I (JSI-124), a selective in- PEA was docked at the C-RAF kinase mapped binding sites. Docking
hibitor of JAK/STAT-3 [27,30,31]. After this period, it was added in the studies were developed with the CLC Drug Discovery Workbench pro-
culture medium 160 mM PEA or 10μM paclitaxel for 24. Next, cells gram (version 2.4, Qiagen Aarhus A/S, 2014). The protocol used was
were fixed and permeabilized with Triton X-100 0.02%, then stained 1000 interactions, considering rigid interactions, and the minimization
with p.STAT-3 antibody and the corresponding secondary antibody was calculated with the Nelder-Mead Simplex Minimization function
both obtained from cell signaling. The percentage of cells expressing the implemented in the software [34]. The classification or score function
protein was analyzed by flow cytometry (FACSCalibur). A total of considers the total potential energy of the complex generated (Gibbs
10.000 cells/sample were analyzed using FlowJo software (TreeStar, interaction free energy, ΔG) with the PLANTSPLP [35] algorithm and
Version X). also considers the square root mean square deviation (RMSD). Negative

27
L.I. Mambelli et al. Biomedicine & Pharmacotherapy 103 (2018) 18–28

values of score indicate the energetic favoring of formation of the en- parallelism from laptops to supercomputers, SoftwareX 1 (2015) 19–25.
zyme-ligand complex. The more negative value of ΔG, the more fa- [9] U.J. Mey, C. Renner, R. Moos, Vemurafenib in combination with cobimetinib in
relapsed and refractory extramedullary multiple myeloma harboring the BRAF
vorable is the formation of the enzyme-ligand complex. V600E mutation, Hematol. Oncol. (2016).
[10] L. Rosen, P. LoRusso, W.W. Ma, J. Goldman, A. Weise, A.D. Colevas, A. Adjei,
4.13. Statistical analysis S. Yazji, A. Shen, S. Johnston, A first-in-human phase 1 study to evaluate the MEK1/
2 inhibitor GDC-0973 administered daily in patients with advanced solid tumors,
AACR, (2011).
GraphPad Prism 5.0 (San Diego, CA) was utilized for all tests. All [11] A.M. Grimaldi, E. Simeone, P.A. Ascierto, The role of MEK inhibitors in the treat-
values were expressed as mean ± SD. Each value is the mean of at least ment of metastatic melanoma, Curr. Opin. Oncol. 26 (2) (2014) 196–203.
[12] A. Vultur, J. Villanueva, C. Krepler, G. Rajan, Q. Chen, M. Xiao, L. Li, P.A. Gimotty,
three independent experiments in each group. Two-way analysis of M. Wilson, J. Hayden, MEK inhibition affects STAT3 signaling and invasion in
variance (ANOVA) with Tukey Post-Hoc test or Student’s t-test was human melanoma cell lines, Oncogene 33 (14) (2014) 1850.
carried out to determine significant differences from untreated controls. [13] Y. Zhang, Y.-Y. Cho, B.L. Petersen, F. Zhu, Z. Dong, Evidence of STAT1 phosphor-
ylation modulated by MAPKs, MEK1 and MSK1, Carcinogenesis 25 (7) (2004)
The asterisk (*) indicates the values that are significantly different from
1165–1175.
control (* p < 0.05, ** p < 0.001 and *** p < 0.0001). The absence [14] C.R. Figueiredo, A.L. Matsuo, R.A. Azevedo, M.H. Massaoka, N. Girola, L. Polonelli,
of * indicates non-significant differences. L.R. Travassos, A novel microtubule de-stabilizing complementarity-determining
region C36L1 peptide displays antitumor activity against melanoma in vitro and in
vivo, Sci. Rep. 5 (2015).
Author contributions [15] A.K. Ferreira, R. Meneguelo, A. Pereira, O.M.R. Filho, G.O. Chierice, D.A. Maria,
Anticancer effects of synthetic phosphoethanolamine on Ehrlich ascites tumor: an
A.K.F designed the research. L.I.M, S.F.T, S.D.J, B.K, R.A.A and experimental study, Anticancer Res. 32 (1) (2012) 95–104.
[16] A. Ferreira, R. Meneguelo, S. Neto, G. Chierice, D. Maria, Synthetic phosphoetha-
A.K.F performed the experiments. L.I.M, S.D.J, J.A.B, R.A.A and A.K.F nolamine induces apoptosis through caspase-3 pathway by decreasing expression of
analyzed the data. L.I.M and A.K.F wrote the main text and prepared all Bax/Bad protein and changes cell cycle in melanoma, J. Cancer Sci. Ther. 3 (1)
figures. All authors reviewed and approved this manuscript. (2011) 53–59.
[17] D. Hanahan, R. Weinberg, The hallmarks of cancer, Cell 100 (2000) 57–70 Find this
article online.
Competing financial interests [18] F. Fu, M.A. Nowak, S. Bonhoeffer, Spatial heterogeneity in drug concentrations can
facilitate the emergence of resistance to cancer therapy, PLoS Comput. Biol. 11 (3)
(2015) e1004142.
The authors declare no competing financial interests. [19] E.J. Calabrese, Hormesis: a revolution in toxicology, risk assessment and medicine,
EMBO Rep. 5 (1S) (2004) S37–S40.
Acknowledgments [20] N. Mahmoodi, N. Motamed, S.H. Paylakhi, The comparsion of silybin and silybin-
phosphatidylcholine effects on viability, ESR1 and ESR2 expression in human breast
cancer T47D cell line, Cell. J. 16 (2015) 1–20.
The authors thank the financial support of FAPESP on this work [21] M. Castedo, J. Perfettini, T. Roumier, G. Kroemer, Cyclin-dependent kinase-1:
(grants 2015/18528-7, 2013/07273-2; post-doctoral fellowships 2016/ linking apoptosis to cell cycle and mitotic catastrophe, Cell Death Differ. 9 (12)
07519-0; 2014/14267-1 and 2017/01265-9; and doctoral fellowship (2002) 1287.
[22] Y. Chen, Cell Adhesion Assay, Bio-protocol Bio101: e98 (2012).
2016/09392-7). [23] T.A. Martin, L. Ye, A.J. Sanders, J. Lane, W.G. Jiang, Cancer Invasion and
Metastasis: Molecular and Cellular Perspective, (2013).
References [24] D. Hanahan, R.A. Weinberg, Hallmarks of cancer: the next generation, Cell 144 (5)
(2011) 646–674.
[25] Z.Y. Diana, S. Jin, Y. Zhuo, J. Field, p21-Activated kinase 1 (Pak1) phosphorylates
[1] A.C. de Lima Luna, G.K.V. Saraiva, O.M. Ribeiro Filho, G.O. Chierice, S.C. Neto, BAD directly at serine 111 in vitro and indirectly through Raf-1 at serine 112, PloS
I.M. Cuccovia, D.A. Maria, Potential antitumor activity of novel DODAC/PHO-S One 6 (11) (2011) e27637.
liposomes, Int. J. Nanomed. 11 (2016) 1577. [26] C.P. Lim, X. Cao, Regulation of Stat3 activation by MEK kinase 1, J. Biol. Chem. 276
[2] A.K. Ferreira, R. Meneguelo, A. Pereira, O.M.R. Filho, G.O. Chierice, D.A. Maria, (24) (2001) 21004–21011.
Synthetic phosphoethanolamine induces cell cycle arrest and apoptosis in human [27] R. Fagard, V. Metelev, I. Souissi, F. Baran-Marszak, STAT3 inhibitors for cancer
breast cancer MCF-7 cells through the mitochondrial pathway, Biomed. therapy: Have all roads been explored? Jak-Stat 2 (1) (2013) e22882.
Pharmacother. 67 (6) (2013) 481–487. [28] N. Jing, D.J. Tweardy, Targeting Stat3 in cancer therapy, Anti-Cancer Drugs 16 (6)
[3] A.K. Ferreira, R. Meneguelo, F.L.N. Marques, A. Radin, O.M.R. Filho, S.C. Neto, (2005) 601–607.
G.O. Chierice, D.A. Maria, Synthetic phosphoethanolamine a precursor of mem- [29] K. Cichowski, P.A. Jänne, Drug discovery: inhibitors that activate, Nature 464
brane phospholipids reduce tumor growth in mice bearing melanoma B16-F10 and (7287) (2010) 358–359.
in vitro induce apoptosis and arrest in G2/M phase, Biomed. Pharmacother. 66 (7) [30] M.S. Van Kester, J.J. Out-Luiting, A. Peter, R. Willemze, C.P. Tensen, M.H. Vermeer,
(2012) 541–548. Cucurbitacin I inhibits Stat3 and induces apoptosis in Sezary cells, J. Invest.
[4] A. Annibal, T. Riemer, O. Jovanovic, D. Westphal, E. Griesser, E.E. Pohl, J. Schiller, Dermatol. 128 (7) (2008) 1691–1695.
R. Hoffmann, M. Fedorova, Structural, biological and biophysical properties of [31] O. Molavi, Z. Ma, S. Hamdy, R. Lai, A. Lavasanifar, J. Samuel, Synergistic antitumor
glycated and glycoxidized phosphatidylethanolamines, Free Radical Biol. Med. 95 effects of CpG oligodeoxynucleotide and STAT3 inhibitory agent JSI-124 in a mouse
(2016) 293–307. melanoma tumor model, Immunol. Cell Biol. 86 (6) (2008) 506.
[5] A. Ferreira, B. Santana-Lemos, E. Rego, Synthetic phosphoethanolamine has in vitro [32] G. Hatzivassiliou, K. Song, I. Yen, B. Brandhuber, D. Anderson, R. Alvarado,
and in vivo anti-leukemia effects, Br. J. Cancer 109 (11) (2013) 2819. M. Ludlam, D. Stokoe, S. Gloor, G. Vigers, RAF inhibitors prime wild-type RAF to
[6] A.K. Ferreira, V.M. Freitas, D. Levy, J.L.M. Ruiz, S.P. Bydlowski, R.E.G. Rici, activate the MAPK pathway and enhance growth, Nature 464 (2010) 431–435.
O.M.R. Filho, G.O. Chierice, D.A. Maria, Anti-angiogenic and anti-metastatic ac- [33] H.M. Berman, J. Westbrook, Z. Feng, G. Gilliland, T. Bhat, H. Weissig,
tivity of synthetic phosphoethanolamine, PLoS One 8 (3) (2013) e57937. I.N. Shindyalov, P.E. Bourne, The protein data bank, Nucleic Acids Res. 28 (1)
[7] D.C. Lev, M. Ruiz, L. Mills, E.C. McGary, J.E. Price, M. Bar-Eli, Dacarbazine causes (2000) 235–242.
transcriptional up-regulation of interleukin 8 and vascular endothelial growth [34] J.A. Nelder, R. Mead, A simplex method for function minimization, Comput. J. 7 (4)
factor in melanoma cells: a possible escape mechanism from chemotherapy1, Mol. (1965) 308–313.
Cancer Ther. 2 (8) (2003) 753–763. [35] O. Korb, T. Stutzle, T.E. Exner, Empirical scoring functions for advanced protein-
[8] M.J. Abraham, T. Murtola, R. Schulz, S. Páll, J.C. Smith, B. Hess, E. Lindahl, ligand docking with plants, J. Chem. Inf. Model. 49 (1) (2009) 84–96.
GROMACS: High performance molecular simulations through multi-level

28

You might also like