ArticlePDF Available

Characterization of the in vitro cytotoxic effects of brachydins isolated from Fridericia platyphylla in a prostate cancer cell line

Authors:

Abstract and Figures

Brachydins (Br) A, B, and C are flavonoids extracted from Fridericia platyphylla (Cham.) L.G. Lohmann roots (synonym Arrabidaea brachypoda), whose extract previously exhibited cytotoxic and antitumor activity. In vitro cell culture of human prostate tumor cell line (PC-3) was used to determine cell viability as evidenced by MTT, neutral red, and LDH release using nine concentrations (0.24 to 30.72 µM) of each brachydin. A triple-fluorescent staining assay assessed the mechanism resulting in cell death. Genomic instability and protein expression were evaluated using comet assay and western blot analysis, respectively. The pro-oxidant status was analyzed using the5-(and-6)-chloromethyl-2′,7′-dichlorodihydrofluorescein diacetate (CM-H2DCFDA) probe. The IC50 values for brachydins BrA, BrB, and BrC were 23.41, 4.28, and 4.44 µM, respectively, and all compounds induced apoptosis and necrosis. BrB and BrC increased p21 levels indicating a possible G1 cell cycle arrest. BrA (6 µM) and BrB (3.84 µM) decreased phospho-AKT (AKT serine/threonine kinase) expression, which also influenced cell cycle and proliferation. BrA, BrB, and BrC elevated cleaved PARP (poly (ADP-ribose) polymerase), a protein related to DNA repair and induction of apoptotic processes. Therefore, this study determined the IC50 values of brachydins in the PC-3 cell line as well as the influence on cell proliferation and cell death processes, such as apoptosis and necrosis, indicating the proteins involved in these processes. Abbreviations ANOVA: Analysis of Variance; BrA: Brachydin A; BrB: Brachydin B; BrC: Brachydin C; CGEN: Genetic Heritage Management Council; CID: Compound identification number; CM-H2DCFDA, 5-(and-6)-chloromethyl-2′,7′-dichlorodihydrofluorescein diacetate, acetyl ester; CO2: Carbon dioxide; DMSO: Dimethyl sulfoxide; DNA: Deoxyribonucleic acid; DTT: Dithiothreitol; DXR: Doxorubicin; ECL: Chemiluminescence; EDTA: Ethylenediaminetetraacetic acid; FBS: Fetal bovine serum; H2O2: Hydrogen peroxide; HRMS: High-Resolution Mass Spectrometry; IC50: Half maximal inhibitory concentration; LDH: Lactate dehydrogenase; MTT, 3-[4,5-Dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide; Na3VO4: Sodium Orthovanadate; NaOH: Sodium hydroxide; NCBI: National Center for Biotechnology Information; NMR: Nuclear Magnetic Resonance; PBS: Phosphate buffer saline; PCR: Polymerase chain reaction; PSMF: Phenylmethylsulfonyl fluoride; RPMI: Roswell Park Memorial Institute Medium; SDS-PAGE: Sodium Dodecyl Sulfate-Polyacrylamide gel electrophoresis; STR: Short tandem repeat; TBS-T: Tris-buffered saline and Polysorbate 20; UPHLC: Ultra-Performance Liquid Chromatography
Content may be subject to copyright.
Characterization of the in vitro cytotoxic eects of brachydins isolated from
Fridericia platyphylla in a prostate cancer cell line
Higor Lopes Nunes
a
, Katiuska Tuttis
a
, Juliana Mara Serpeloni
a
, Jessyane Rodrigues do Nascimento
b
,
Claudia Quintino da Rocha
b
, Viviane Aline Oliveira Silva
c
, André van Helvoort Lengert
c
, Rui Manuel Reis
c,d,e
,
and Ilce Mara de Syllos Cólus
a
a
Departamento de Biologia Geral, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina, Paraná, Brasil;
b
Departamento
De Química, Centro de Ciências Exatas e Tecnologia, Universidade Federal do Maranhão, São Luís, Maranhão, Brasil;
c
Centro de Pesquisa em
Oncologia Molecular, Hospital de Câncer de Barretos, Barretos, São Paulo, Brasil;
d
Life and Health Sciences Research Institute (ICVS), School of
Medicine, University of Minho, Braga, Portugal;
e
ICVS/3B’s - PT Government Associate Laboratory, Braga/Guimarães, Portugal
ABSTRACT
Brachydins (Br) A, B, and C are avonoids extracted from Fridericia platyphylla (Cham.) L.G. Lohmann
roots (synonym Arrabidaea brachypoda), whose extract previously exhibited cytotoxic and anti-
tumor activity. In vitro cell culture of human prostate tumor cell line (PC-3) was used to determine
cell viability as evidenced by MTT, neutral red, and LDH release using nine concentrations (0.24 to
30.72 µM) of each brachydin. A triple-uorescent staining assay assessed the mechanism resulting
in cell death. Genomic instability and protein expression were evaluated using comet assay and
western blot analysis, respectively. The pro-oxidant status was analyzed using the5-(and-6)-
chloromethyl-2,7-dichlorodihydrouorescein diacetate (CM-H
2
DCFDA) probe. The IC
50
values for
brachydins BrA, BrB, and BrC were 23.41, 4.28, and 4.44 µM, respectively, and all compounds
induced apoptosis and necrosis. BrB and BrC increased p21 levels indicating a possible G1 cell
cycle arrest. BrA (6 µM) and BrB (3.84 µM) decreased phospho-AKT (AKT serine/threonine kinase)
expression, which also inuenced cell cycle and proliferation. BrA, BrB, and BrC elevated cleaved
PARP (poly (ADP-ribose) polymerase), a protein related to DNA repair and induction of apoptotic
processes. Therefore, this study determined the IC
50
values of brachydins in the PC-3 cell line as well
as the inuence on cell proliferation and cell death processes, such as apoptosis and necrosis,
indicating the proteins involved in these processes.
Abbreviations: ANOVA: Analysis of Variance; BrA: Brachydin A; BrB: Brachydin B; BrC: Brachydin C;
CGEN: Genetic Heritage Management Council; CID: Compound identication number; CM-H
2
DCFDA, 5-(and-6)-chloromethyl-2,7-dichlorodihydrouorescein diacetate, acetyl ester; CO
2
:
Carbon dioxide; DMSO: Dimethyl sulfoxide; DNA: Deoxyribonucleic acid; DTT: Dithiothreitol; DXR:
Doxorubicin; ECL: Chemiluminescence; EDTA: Ethylenediaminetetraacetic acid; FBS: Fetal bovine
serum; H
2
O
2
: Hydrogen peroxide; HRMS: High-Resolution Mass Spectrometry; IC50: Half maximal
inhibitory concentration; LDH: Lactate dehydrogenase; MTT, 3-[4,5-Dimethylthiazol-2-yl]-2,5-diphe-
nyl tetrazolium bromide; Na3VO4: Sodium Orthovanadate; NaOH: Sodium hydroxide; NCBI: National
Center for Biotechnology Information; NMR: Nuclear Magnetic Resonance; PBS: Phosphate buer
saline; PCR: Polymerase chain reaction; PSMF: Phenylmethylsulfonyl uoride; RPMI: Roswell Park
Memorial Institute Medium; SDS-PAGE: Sodium Dodecyl Sulfate-Polyacrylamide gel electrophoresis;
STR: Short tandem repeat; TBS-T: Tris-buered saline and Polysorbate 20; UPHLC: Ultra-Performance
Liquid Chromatography
KEYWORDS
Apoptosis; cytotoxicity;
flavonoids
Introduction
Cancer is one of the diseases responsible globally
for high morbidity and mortality rates. This disease
has been treated with several therapies, including
cytotoxic chemotherapy, using synthetic or natural
cytotoxic chemotherapeutics (Huang, Cai, and
Zhang 2009; Siegel, Miller, and Jemal 2016).
Prostate cancer is the type of non-cutaneous cancer
frequently diagnosed in men associated with a high
rate of morbidity and mortality (Pezaro, Woo, and
Davis 2014; Woodrum et al. 2017). Treatments for
this condition include surgery, chemotherapy,
immunotherapy, and radiotherapy. Although
these options are effective for different types of
cancers, there are limitations as adverse effects
occur in patients (Bonam et al. 2018). Considering
CONTACT Juliana Mara Serpeloni julianaserpeloni@yahoo.com.br Departamento de Biologia Geral, Centro de Ciências Biológicas, Universidade
Estadual de Londrina, Londrina, Paraná CEP 86057-970, Brasil
JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH, PART A
https://doi.org/10.1080/15287394.2020.1784339
© 2020 Taylor & Francis
factors such as (1) increased incidence of cancer,
(2) aging population, and (3) need to develop thera-
pies with fewer adverse effects, the search for new
therapeutic approaches becomes necessary.
Plant-derived natural compounds have become
indispensable for modern pharmacotherapy
(Corveloni et al. 2020; Gontijo et al. 2018; Rocha
et al. 2019a; Yesil-Celiktas et al. 2010). Medicinal
plants have historically been a valuable drug source
and are still explored regarding their modulation of
several molecular targets for the treatment of
chronic diseases, including cancer (Atanasov et al.
2015; González-Vallinas, Reglero, and Ramírez de
Molina 2015; Majolo et al. 2020).
The products of plant secondary metabolism are
the focus of several new research investigations.
These products need to be extracted, isolated, and
structurally verified before the evaluation of poten-
tial biological and pharmacological activities using
in vitro and in vivo models (Atanasov et al. 2015;
Maistro et al. 2019; Russo et al. 2010).
Brachydin A (BrA), B (BrB), and C (BrC) are
unusual previously described dimeric flavonoids
isolated from the dichloromethane fraction of
a hydroalcoholic extract of the roots of the plant
species Fridericia platyphylla (Cham.) L.G.
Lohmann, identified as Arrabidaea brachypoda
(DC) Bureau (da Rocha et al. 2014). In a recent
study, Serpeloni et al. (2020) demonstrated cyto-
toxic and anti-tumor activity in tumor gastric cells
incubated with Fridericia platyphylla extract mod-
ulating apoptosis- and cell cycle-related genes.
The traditional use of teas prepared from the
roots of this plant is already known for the treat-
ment of kidney stone and arthritis (Rodrigues and
Carlini 2005). De Sousa Andrade et al. (2020)
reported that brachydins BrA and BrB displayed
antimicrobial actions against Staphyloccocus aur-
eus, Escherichia coli, and Candida albicans.
Recently investigators focused predominantly on
the cytotoxic activity of isolated brachydins, mainly
as anti-parasites. da Rocha et al. (2014) examined
the toxicity of these three compounds against the
extracellular trypomastigotes of Trypanosoma
cruzi. In the same study, BrB and BrC exhibited
cytotoxicity against mammalian cells and intracel-
lular amastigotes of T. cruzi and were capable of
inhibiting parasite invasion. Further, in vivo tests
using female BALB/c mice demonstrated that BrB
was able to reduce blood parasitemia and mortality
when compared to controls. Antileishmanial activ-
ity was also observed for BrB, which was toxic to
intracellular amastigotes of Leishmania amazonen-
sis but not to host mammalian cells (Rocha et al.
2019b). It is emphasized that in the Ames test, no
mutagenicity of BrA, BrB, and BrC was noted
(Resende et al. 2017).
The dichloromethane fraction of the hydroetha-
nolic root extract, which contains a mixture of the
three compounds, was also examined using in vivo
and in vitro assays. This extract did not induce
estrogenic activity (Resende et al. 2017); however,
gastroprotective (da Rocha et al. 2017), antinoci-
ceptive, and anti-inflammatory effects (da Rocha
et al. 2011), as well as alleviation of acute pain in
mice, were reported (Rodrigues et al. 2017).
Considering the positive cytotoxic and antitu-
moral activity observations obtained for the crude
extract of Fridericia platyphylla in cancer cells
(Serpeloni et al. 2020), this study aimed to assess
in vitro the possible bioactivities of the three bra-
chydins, the major constituents of the extract, using
the human prostate cancer-derived cell line, PC-3
and determine the molecular mechanisms under-
lying potential medicinal uses, providing insights to
new cancer therapies.
Materials and methods
Plant material and compound obtainment
Fridericia platyphylla root samples were collected in
April 2016 at Sant’Ana da Serra farm João Pinheiro,
Minas Gerais, Brazil (Location: 17º44ʹ45”S,
46º10ʹ44”W). A voucher specimen (n°17935) was
deposited at the Herbarium of the Federal
University of Ouro Preto, Brazil. This procedure
was approved by CGEN Genetic Heritage
Management Council (A451DE4). Previously the
nomenclature Arrabidaea brachypoda (DC.)
Bureau (da Rocha et al. 2011) proposed by Prof.
Dr. Ana Maria Cristina Braga Messias from Federal
University of Ouro Preto was used, but its taxonomy
was later modified to Fridericia platyphylla (Cham.)
L.G. Lohmann by Kaehler (2020). The plant name
was verified with http://www.theplantlist.org.
All isolation processes and structure elucidation of
the three dimeric flavonoids identified as brachydin
2H. L. NUNES ET AL.
A (BrA), brachydin B (BrB), and brachydin C (BrC)
are described by da Rocha et al. (2014). The structure
elucidation was determined using nuclear magnetic
resonance (NMR) and high-resolution mass spectro-
metry (HRMS) techniques. There is only a single radi-
cal difference between them: a hydroxyl group is
found in BrA, a methoxyl group is present in BrB,
and a single hydrogen occurs in BrC (Figure 1). The
purities of the three compounds were determined to
employ ultra-performance liquid chromatography
(UHPLC- HMRS) analysis and were greater than 98%.
BrA, BrB, and BrC were diluted in dimethyl
sulfoxide (DMSO) purchased from Labsynth,
Diadema, SP, Brazil, and then phosphate buffer
saline (PBS) was added in equal quantity to obtain
a final solution of 50% DMSO, resulting in 0.5% in
cell culture.
Cell line and cell culture
The prostate tumor cell line PC-3 (American Type
Culture Collection – Manassas, VA, USA), which is
derived from a bone metastatic site, was cultivated
using Roswell Park Memorial Institute (RPMI
Gibco, Grand Island, NY, USA) 1640 culture medium
supplemented with 10% fetal bovine serum (FBS –
Gibco, Grand Island, NY, USA) and 1% antibiotic-
antimycotic solution (Gibco, Grand Island, NY, USA)
at 37°C in an atmosphere containing 5% CO
2
and
95% relative humidity. The assays were performed
using cells cultured between the third and tenth pas-
sages. In addition to the absence of mycoplasma
determined by PCR, the authentication of PC3 cancer
cell line was confirmed at Molecular Diagnostic
Center of Barretos Cancer Hospital by short tandem
repeat (STR) DNA typing and performed according
to the International Reference Standard for
Authentication of Human Cell Lines using a panel
of eight (D5S818, D13S317, D7S820, D16S539, vWA,
TH01, TPOX, and CSF1P0) STR loci and gender
determination amelogenin (AMEL) (Dirks et al.
2005).
Cell viability assays
To perform the MTT, neutral red, and LDH activ-
ity release viability assays, 1 × 10
4
cells were seeded
per well in 96-well culture plates and to adhere for
24 hr. Subsequently, the medium was removed,
and cells washed in PBS. Then, negative control
(PBS), solvent control (DMSO 0.5%), positive
control (Docetaxel 10 µM), and treatments were
added to the medium without FBS for 24 hr. Nine
concentrations of BrA, BrB, and BrC (0.24, 0.75,
0.96, 1.5, 3.84, 6, 15.36, 24, or 30.72 µM), were
selected based upon a previous study (da Rocha
et al. 2014). Three biological replicates and four
technical replicates were performed for each cell
viability assay. The neutral red and MTT results
were expressed as cell viability %, considering the
negative control as 100%. LDH activity release
findings were expressed in absorbance relative to
LDH activity.
MTT assay
The MTT assay was performed as described by
Mosmann (1983) using 3-[4,5-dimethylthiazol-
2-yl]-2,5-diphenyl tetrazolium bromide (CAS:
298–93-1, Sigma-Aldrich, St. Louis, MO, USA).
After treatments, cells were washed in PBS and
incubated in MTT solution (0.5 mg/ml) for 4 hr at
37°C. Then, the MTT solution was removed, and
Figure 1. The chemical structure of the brachydins.
JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH, PART A 3
200 µl DMSO was added to each well. The absor-
bance at 570 nm was read with a spectrophotometer
(Biotek Eon, Winooski, VT, USA).
Neutral red assay
The neutral red assay was conducted following the
protocol of Repetto, Del Peso, and Zurita (2008)
using neutral red (CAS: 553–24-2, Sigma-Aldrich,
St. Louis, MO, USA). After the treatments, cells
were washed with PBS and incubated in neutral
red solution (40 µg/ml) for 3 hr at 37°C. Then, the
neutral red solution was removed, and the cells
were washed with PBS. To enable the reading,
0.1 ml destaining solution (50% ethanol, 49% deio-
nized water, and 1% glacial acetic acid) was added
to each well. The absorbance was read with a Biotek
Eon spectrophotometer at 540 nm.
LDH activity release assay
The Pierce™ LDH Cytotoxicity Assay Kit (Thermo
Fisher Scientific, Waltham, MA, USA) was used to
measure membrane integrity according to the man-
ufacturer’s instructions. An aliquot of 50 µl cell
culture medium was withdrawn to a new plate
after treatment, and kit solutions added into the
new plate. The absorbance was measured with
a Biotek Eon spectrophotometer at 490 nm and
680 nm.
Cell death assay
To perform the cell death assay, 1.5 × 10
5
cells were
seeded per well in 12-well culture plates, stabilized
in culture medium for 24 hr and then treated in
triplicate with three concentrations of BrA, BrB, or
BrC (1.5, 3.84, or 6 µM) selected according to the
results of previous viability assays. A positive (dox-
orubicin – DXR at 0.345 µM) and negative (PBS)
controls were also used. After treatment, cells were
washed with PBS and trypsinized. To assess cell
death morphology, a triple-staining assay using
propidium iodide (1 µg/ml), Hoechst 33342 (4 µg/
ml) and fluorescein diacetate (7.5 µg/ml) was per-
formed (Martínez-López et al. 2010; Proietti De
Santis et al. 2003; Ryhänen et al. 2009). Two hun-
dred cells per treatment were counted using an
Olympus BX 43 fluorescence microscope
(Olympus Microscopy, Europe), recording the
number of viable, necrotic, and apoptotic cells.
Pro-oxidant evaluation
To quantify intracellular reactive species, 1 × 10
4
cells were seeded in 96-well black plates and
allowed to stabilize for 24 hr. Two biological repli-
cates and four technical replicates were performed
for each treatment. Subsequently, treatment in
quadruplicate with 4 concentrations of BrA, BrB,
or BrC was performed (0.96, 1.5, 3.84, or 6 µM) for
4 different treatment periods (1, 3, 12, or 24 hr).
Hydrogen peroxide (H
2
O
2
) 1 mM was used as
a positive control 20 min before the treatment end
point and PBS as a negative control. To measure
reactive species, the probe CM-H
2
DCFDA (Life
Technologies, Eugene, OR, USA) was employed at
5 µM, following the manufacturer’s instructions
and lab protocols (Serpeloni et al. 2015; Tuttis
et al. 2018). The obtained values represent the
fluorescence absorbance of the probe.
Comet assay
The comet assay was performed as described by
Tice et al. (2000) and Singh et al. (1988). Initially,
2 × 10
5
cells were seeded in 24-well plates and
stabilized for 24 hr. The treatments were performed
in two biological and three technical replicates with
three concentrations of BrA, BrB, or BrC (1.5, 3.84,
or 6 µM), PBS as negative control and DXR
(0.345 µM) as positive control. The treatment
time was 4 hr, preventing DNA repair (OECD TG
489, 2016; Sasaki, Nakamura, and Kawaguchi 2007;
Singh et al. 1988). Cell viability was verified (≥90%)
with trypan blue exclusion using the Countess®
Automated Cell Counter (Life Technologies,
Eugene, OR, USA). Then, cells were mixed in low
melting point agarose (0.5%) and placed on slides
coated with normal melting pointing agarose
(1.5%). After drying, the coverslips were removed,
and slides submerged in lysis solution (2.5 M NaCl,
100 mM EDTA and 10 mM Tris, pH 10) for 1.5 hr
at 4°C. For electrophoresis, the slides were allowed
to rest in alkaline solution (300 mM NaOH, 1 mM
EDTA, pH >13) for 20 min at 4°C. Then, electro-
phoresis was carried out for 20 min, 25 V, and
300 mA. After, the slides were submerged in neu-
tralization buffer (0.4 M Tris, pH 7.5) for 15 min,
then fixed in absolute ethanol (99%) for 5 min and
left to air dry. For analysis, slides were stained with
4H. L. NUNES ET AL.
GelRed™ Nucleic Acid Gel Stain (Biotium,
Fremont, CA, USA) and analyzed using fluores-
cence microscopy. A total of 300 cells were counted
for each treatment using the software Comet
Imager v 2.2 (MetaSystems, Germany), and tail
length was determined.
Western blot analysis
To assess cell protein expression, 1 × 10
6
PC-3 cells
were seeded in 6-well plates and allowed to stabilize
for 24 hr. The treatments were performed with two
biological and three technical replicates at two con-
centrations of BrA (3.84 or 6 µM) and one concen-
tration of BrB, and BrC (3.84 µM). The
concentration of 3.84 µM was chosen because it is
close to the IC
50
value. For BrA that presented
higher IC
50
values, the concentration of 6 µM was
also added. After 24 hr incubation, the cells were
harvested and lysed with lysis buffer containing
Tris buffer (50 mM, pH 7.6 ~ 8), sodium chloride –
NaCl (150 mM), EDTA (5 mM), sodium orthova-
nadate Na
3
VO
4
(1 mM), sodium fluoride NaF
(10 mM), Na pyrophosphate (10 mM), and NP-40
(1%), supplemented with inhibitor cocktail con-
taining protease inhibitor DTT (1 mM), leupeptin
(1 μg/ml), aprotinin (1 μg/ml), phenylmethylsulfo-
nyl fluoride – PSMF (1 mM) and EDTA (1 mM) for
1 hr. After centrifugation at 12,000 g for 15 min at
4°C, the soluble proteins were quantified by
a Bradford assay kit (Sigma-Aldrich, St. Louis,
MO, USA) following the manufacturer’s instruc-
tions. Proteins (20 µg) were separated through
SDS-PAGE (12% and 15%) and transferred into
a nitrocellulose membrane (Hybond-C,
Amersham Biosciences, Little Chalfont, UK) with
a mini Trans-blot® Turbo Transfer System (Bio-
Rad, CA, USA). The membranes were blocked
with milk (5%) in Tris-buffered saline (TBS) and
0.1% Tween 20 (TBS-T) for 1 hr. Then, membranes
were incubated with the corresponding primary
antibodies overnight. The antibodies used were
anti-total PARP (Poly(ADP-ribose) polymerase),
anti-phospho AKT (AKT serine/threonine kinase),
anti-p21 (Cyclin-dependent kinase inhibitor 1A)
and anti-p27 (Cyclin-Dependent Kinase Inhibitor
1B) (Cell Signaling Technology, MA, USA) diluted
1:1000. α-Tubulin (Cell Signaling) was utilized as
a loading control. The membranes were washed
with TBS-T and incubated with peroxidase-
conjugated secondary antibody (Cell Signaling
Technology, MA, USA). The detection was per-
formed through chemiluminescence (ECL) western
blot detection reagents (RPN2109, GE Healthcare,
IL, USA), and the signal was detected and imaged
using ImageQuant™ LAS 4000 mini (GE
Healthcare). The results were quantified into den-
sitometry data using the software ImageJ v. 1.41.
Comparison of similar molecules
The search for molecules with ≥90% structural simi-
larity to brachydins (compound identification num-
ber – CID: – BrA −102339051; BrB – 102339053;
BrC – 102339052) was conducted using a PubChem
Chemical Structure Search.
Statistical analysis
Normality of all results was obtained using the
Shapiro–Wilk test. With the null hypothesis
accepted, analysis of variance (ANOVA) was applied
followed by a Dunnett’s test or Tukey’s test. The
comet assay results were analyzed with a non-
parametric Kruskal–Wallis test followed by Dunn’s
test. All statistical analyses were performed using the
free software RStudio© v. 1.1.442 (RStudio, Inc.).
Results
Cell viability
The MTT test illustrated that BrA, BrB, and BrC
produced cytotoxicity in the PC-3 prostate tumor
cell line (Figure 2). BrA, BrB, and BrC decreased
cell viability starting at 15.36 µM, 3.84 µM, and
3.84 µM, with IC
50
values of 23.41 µM, 4.28 µM,
and 4.44 µM, respectively.
Figure 3 shows results obtained in the neutral red
assay for the PC-3 cell line. BrA, BrB and BrC
induced cytotoxicity starting at 15.36 µM, 6 and
and 3.84 µM, respectively.
Further, the LDH activity release assay showed
that BrA, BrB, and BrC induced the death of PC-3
cells at concentrations of 15.36 µM, 3.84 µM, and
6 µM, respectively (Figure 4). In order to assess cell
death induced by brachydins, a triple-staining cell
death assay was performed which demonstrated
JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH, PART A 5
that BrA (6 µM) induced necrosis, BrC (1.5, 3.84, or
6 µM) produced apoptosis and BrB (6 µM) initiated
both necrosis and apoptosis (Table 1).
Pro-oxidant eects
Due to the cell death observed in the triple-staining
assay, reactive intracellular species were quantified
to verify if the results observed were related to pro-
oxidant effects in PC-3 cells treated with brachy-
dins. Only one concentration of BrA (6 µM) at one
time point (24 hr) initiated a significant increase in
reactive intracellular species (Figure 5). Both BrB
and BrC, which were cytotoxic at lower concentra-
tions, failed to markedly affect reactive species
generation.
Figure 2. Cell viability (%) of the PC-3 cell line evaluated by MTT assay after 24 hr of treatment with different concentrations of BrA, BrB
and BrC. NC – negative control (PBS: phosphate buffer saline). SV solvent control (dimethyl sulfoxide 0.5%). PC – positive control
(Docetaxel 10 µM). Data are presented as the mean ± SD. *Statistically different from negative control (p < .05) by ANOVA followed by
Dunnett’s test.
Figure 3. Cell viability (%) of the PC-3 cell line evaluated by neutral red assay after 24 hr of treatment with different concentrations of
BrA, BrB and BrC. NC – negative control (PBS: Phosphate buffer saline). SV – solvent control (dimethyl sulfoxide 0.5%). PC – positive
control (Docetaxel 10 µM). Data are presented as the mean ± SD. *Statistically different from negative control (p < .05) by ANOVA
followed by Dunnett’s test.
Figure 4. LDH assay of PC-3 cells treated for 24 hr with different concentrations of BrA, BrB and BrC. NC – negative control (PBS:
Phosphate buffer saline). SV – solvent control (dimethyl sulfoxide 0.5%). PC – positive control (cells lysed by manufacturer’s reagent).
Data are presented as the mean ± SD. *Statistically different from negative control (p < .05) by ANOVA followed by Tukey’s test.
6H. L. NUNES ET AL.
Table 1. Percentage of PC-3 cells that were viable, apoptotic, and necrotic after treatment with three
different concentrations of BrA, BrB, and BrC and their respective controls.
Cells (%)
Treatment Viable Necrotic Apoptotic
NC (PBS) 96.00 ± 2.00 1.67 ± 1.53 2.33 ± 1.53
PC (DXR −0.345 µM) 80.33 ± 1.53* 9.00 ± 2.65* 10.67 ± 2.08*
SV (DMSO 0.5%) 91.67 ± 2.52 5.00 ± 1.73 2.00 ± 1.00
Brachydin A (µM)
1.5 96.00 ± 2.65 3.67 ± 2.08 0.33 ± 0.58
3.84 94.67 ± 0.58 4.33 ± 1.53 1.00 ± 1.00
6.00 91.67 ± 2.08 7.00 ± 2.65* 1.33 ± 0.58
Brachydin B (µM)
1.5 81.67 ± 8.98 14.83 ± 9.36 3.50 ± 0.50
3.84 85.50 ± 3.04 8.17 ± 2.02 6.33 ± 1.53
6.00 64.50 ± 3.50* 17.00 ± 2.18* 18.50 ± 3.90*
Brachydin C (µM)
1.5 90.17 ± 3.21 1.67 ± 0.29 8.17 ± 3.33*
3.84 89.67 ± 2.25 4.33 ± 2.02 6.00 ± 1.50*
6.00 90.00 ± 1.32 2.33 ± 0.58 7.67 ± 1.89*
NC – negative control (PBS: Phosphate buffer saline). SV – solvent control (DMSO 0.5%). PC – positive control (DXR
0.345 µM). Data are presented as the percentage (%) of cell morphology type found with triple staining.
*Statistically different from the negative control by ANOVA followed by Tukey’s test (p < 0.05).
Figure 5. Quantification assay of intracellular reactive species using the CM-H
2
DCFDA probe. NC – negative control (PBS: Phosphate
buffer saline). SV – solvent control (dimethyl sulfoxide 0.5%). PC – positive control (H
2
O
2
1 mM). Data are presented as the mean ± SD.
*Statistically different from negative control (p < .05) by ANOVA followed by Tukey’s test.
JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH, PART A 7
Genotoxic eects
None of the concentrations evaluated of brachydins
were genotoxic to PC-3 cells after 4 hr incubation
(data not shown).
Protein expression
The results obtained from western blot analysis are
presented in Figure 6. The protein p21 promotes
cell cycle arrest in response to a variety of stimuli
and was overexpressed in PC-3 cells treated with
3.84 µM of BrB or BrC but not BrA. However, the
expression of the protein p27, which also regulates
the cell cycle, was not markedly altered by any of
the treatments. pAKT expression was reduced in
cells treated with BrA at 6 µM and BrB at 3.84 µM,
indicating dysregulation of cellular survival pro-
cesses. The expression of cleaved PARP was higher
in all treatments examined, suggesting cell death by
apoptosis.
Similar molecules have similar eects
The similarity of brachydins with other molecules
was investigated using the public database PubChem
from the National Center for Biotechnology
Information (NCBI). Among 1330 molecules with
similarity (≥90%) to brachydins, 101 were already
evaluated in bioassays, and 44 of them were active.
Some similar molecules exhibited cytotoxicity in
several tumor cell lines (HepG2 liver; MCF-7 and
T-47D mammary epithelium; A549 lungs;
SV480 – colon) but did not induce mutagenic or
genotoxic effects (Table 2).
Discussion
The three classical prostate cancer cell lines,
namely, DU145, PC-3, and LNCaP, are most widely
used in prostate cancer research (Namekawa et al.
2019). Further, the PC-3 cell line was used in the
present study as an in vitro model to assess the
effects of brachydins.
Different methodologies may be employed for
analysis of cytotoxicity of compounds and the com-
bination between these is an important factor
(Specian et al. 2016). The biological effects observed
after the combination of tests may enable inference
regarding the mechanisms of action of the com-
pounds, which may result in the identification of
target molecules and/or organelles to determine
their biological activities (Atanasov et al. 2015). In
the present study, different assays were performed
to evaluate the effects of brachydins to different
cellular targets such as respiratory chain activity
(MTT), lysosomal activity (Neutral Red), and
membrane integrity (LDH activity release). It is
noteworthy that similar results were detected with
the brachydins.
Table 2. PubChem public database study summary and compar-
ison of brachydins and similar molecules.
Biological activities
Similar mole-
cules
(PubChem)
Brachydins
Reference
Cytotoxicity Present study
Activity against pathogens da Rocha et al.
(2014)
Genotoxicity Present study
Mutagenicity Resende et al.
(2017)
Induction of reactive intracellular
species
Present study
presence of effect, absence of effect.
Figure 6. Densitometry quantification of p21, p27, pAKT and
cleaved PARP of western blot with PC-3 cell lines treated with
different concentrations of BrA, BrB, and BrC after 24 hr. NC –
negative control (PBS: Phosphate buffer saline). PC – positive
control (DXR 0.345 µM). A – Brachydin A, B – Brachydin B, C –
Brachydin C. Data are presented as the mean ± SD. *Statistically
different from negative control (p < .05) by ANOVA followed by
Tukey’s test.
8H. L. NUNES ET AL.
The cytotoxic concentration of each brachydin was
determined to utilize the PC-3 cell line. The IC
50
values found were 23.41, 4.28, and 4.44 µM for BrA,
BrB, and BrC, respectively. These values are also
similar to those obtained in PC-3 cells in vitro with
the chemotherapy drug cisplatin (3.3 µM), employed
in the treatment of prostate cancer (Altaf et al. 2019).
Da Rocha et al. (2014) estimated the IC
50
values of
brachydins BrB and BrC in Trypanosoma cruzi (5.3
and 6.6 µM, respectively), intracellular amastigotes
(6 µM and 6.8 µM, respectively) and in macrophages
(15.6 µM and 17.3 µM), while BrA was inactive.
In addition to inducing cytotoxicity, chemother-
apeutic compounds might influence cell cycle pro-
gression and cell death mechanisms, which are
mechanisms that limit cancer progression and are
important parameters for the evaluation of potential
chemotherapy drugs (Sikes 2007). In order to under-
stand the influence of brachydins on cellular path-
ways, some of the intracellular pathways following
short-term exposure to these plant extracts were
examined in prostate cancer cells. The AKT protein,
which is involved in the PI3K/AKT/mTOR meta-
bolic pathway, regulates quiescence, proliferation,
and cell longevity (King, Yeomanson, and Bryant
2015) and its activity was significantly reduced in
PC-3 cells after treatment with BrA and BrB.
Further, BrB and BrC also decreased cell cycle pro-
gression through induction of p21 expression. Since
p27 expression is not markedly changed by brachy-
dins, p21 may have prevented cyclin-CDK4/6 com-
plex formation, inducing cell cycle arrest at G1
phase. This finding might be confirmed in further
studies with flow cytometry. These data indicate
a possible interference of dimeric flavonoids in the
cell cycle as well as in cell growth and metabolic
processes, which may favor and explain the observed
apoptotic cell death (Nitulescu et al. 2018).
With respect to cell death, LDH activity release
and cleaved PARP expression support the hypoth-
esis that brachydins are cytotoxic for human pros-
tate tumor cell line. The LDH assay data are
consistent with observations for cell death. The
PARP protein is involved in DNA repair and pro-
grammed cell death, and the cleaved version of
PARP, which occurs in apoptosis cell death, is fre-
quently used as a marker of this type of cell death
(Morales et al. 2014). The increase in cleaved PARP
expression after cell treatment with brachydins
suggests the induction of apoptosis processes by
these compounds.
The exact type of cell death induced could not be
identified with the assays performed because triple
staining demonstrated both apoptotic and necrotic
morphology. Nevertheless, overexpression of PARP
signaled apoptosis, and the results of the LDH
activity release assay suggested cell death by mem-
brane disruption such as necrosis. It is noteworthy
that western blot analysis revealed brachydins
influenced the expression of cell cycle and cell
death proteins, corroborating the viability assays,
MTT and neutral red, which measure death and
proliferation.
Oxidative stress induced by unbalanced control
between reactive species and antioxidants might be
related to necrotic cell death (Choi et al. 2009).
However, no marked alteration in intracellular reac-
tive species was noted after brachydin treatment
except for 6 µM of BrA, which elevated the reactive
species level with 24 hr treatment. This concentration
also induced necrosis in the triple-staining assay.
The identification of genotoxicity and mutagenicity
is crucial for the evaluation of new compounds, since
these processes are related to the capability of
a compound to damage genetic material (Eastmond
et al. 2009). Resende et al. (2017) used the Ames test to
demonstrate that the extracts obtained from leaves,
roots, and stalks of F. platyphylla extract were not
mutagenic. F. platyphylla root extract increased the
frequency of nuclear buds in tumor gastric cells
(Serpeloni et al. 2020). No genotoxicity and mutageni-
city were detected for brachydins isolated from
F. platyphylla in the present study (comet assay), nor
by Resende et al. (2017) in Ames test, indicating
noDNA strand breaks or mutations in genetic
material.
To compare brachydin A with other molecules,
a computational method was used based upon
Tanimoto’s coefficient (Maggiora et al. 2014) in
PubChem. This method is employed in chemical
informatics and drug discovery and enabled us to
verify our results in relation to similar molecules
from the literature. In the search for similar mole-
cules, some active molecules, such as licoricidin
(CID: 480865) and glyasperin D (CID: 480860),
were cytotoxic to HepG2, MCF-7, A459, and
SV480 tumor cells. In proliferation assays, two
molecules showed divergent activity: erycibenin
JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH, PART A 9
E (CID: 16093658) inhibiting proliferation in
tumor lymphocytes, and vestitol (CID: 182259) sti-
mulating proliferation in T-47D tumor cells. These
data demonstrated similarities to the cytotoxic pro-
files of brachydins in tumor cells, as well as activity
against pathogens (de Sousa Andrade et al. 2020;
Rocha et al. 2019b), and absence of genotoxicity
and mutagenicity (Resende et al. 2017).
Due to the cytotoxicity of BrB at lower doses than
others (reducing the necessity of raw material), BrB is
a more promising compound for further investiga-
tion. With only one radical difference in the three
brachydins evaluated in this study (BrA – hydroxyl,
BrB – methoxyl, and BrC hydrogen), it is still not
possible to conclude if these differences confer distinct
biological activities of these phytochemicals. Thus,
additional studies need to be performed in order to
understand the influence of brachydins on different
cellular pathways.
Conclusions
The viability/cytotoxicity assays performed indicate
that the flavonoids brachydins A, B, and C,
obtained from the root extract of Fridericia platy-
phylla (Cham.) L.G. Lohmann, induce cytotoxicity
in the human prostate tumor cell line PC-3.
Although the present study examined the effect of
brachydins only in PC-3 cells, it was possible to
define minimum toxicity thresholds, which might
be used in subsequent studies to assess efficacy
without adverse consequences. The results showed
that the cytotoxicity and lower proliferation rate are
sustained by the responses observed in western blot
assays with p21, pAKT, and PARP proteins and by
computational comparison between similar mole-
cules in a public database. These compounds need
to be further investigated to (1) assess whether
effects are selective on normal cells and (2) better
understand the underlying mechanisms of action
and molecular targets to be potentially considered
as phytopharmaceuticals for disease treatment,
especially for prostate cancer.
Authors’ contributions
Conceptualization and design: Serpeloni, J.M.; Cólus, I.M.S.;
Nunes, H.L. Provision of study materials and funding acquisi-
tion: Serpeloni, J.M.; Rocha, C.Q.; Reis, R.M.; Cólus, I.M.S.
Collection and assembly of data: Nunes, H.L.; Tuttis, K.;
Lengert, A.H.; Nascimento, J.R.; Silva; V.A.O. Data analysis
and interpretation: Nunes, H.L.; Serpeloni, J.M.; Cólus, I.M.S.
Writing – original draft and editing: Nunes, H.L.; Cólus, I.
M.S.; Serpeloni, J.M.;
Supervision and project administration: Cólus, I.M.S.;
Serpeloni, J.M.
Conicts of interest
No potential conflict of interest was reported by the authors.
Funding
The authors are thankful to CNPq (No. 401516/2016-4),
FAPEMA (No. 00849/18), CAPES (Financial code 001), and
the State University of Londrina and Barretos Cancer Hospital
(HCB), which made the development of this study possible.
References
Altaf, M., N. Casagrande, E. Mariotto, N. Baig, A. N. Kawde,
G. Corona, R. Larcher, C. Borghese, C. Pavan,
A. A. Seliman, et al. 2019. Potent in vitro and in vivo antic-
ancer activity of new bipyridine and bipyrimidine gold (III)
dithiocarbamate derivatives. Cancers 11:474. doi:10.3390/
cancers11040474.
Atanasov, A. G., B. Waltenberger, E.-M. Pferschy-Wenzig,
T. Linder, C. Wawrosch, P. Uhrin, V. Temml, L. Wang,
S. Schwaiger, E. H. Heiss, et al. 2015. Discovery and resup-
ply of pharmacologically active plant-derived natural pro-
ducts: A review. Biotechnol. Adv. 33:1582–614. doi:10.1016/
j.biotechadv.2015.08.001.
Bonam, S. R., Y. S. Wu, L. Tunki, R. Chellian,
M. S. K. Halmuthur, S. Muller, and V. Pandy. 2018. What
has come out from phytomedicines and herbal edibles for
the treatment of cancer? Chem. Med. Chem. 13:1854–72.
doi:10.1002/cmdc.201800343.
Choi, K., J. Kim, G. W. Kim, and C. Choi. 2009. Oxidative
stress-induced necrotic cell death via
mitochondira-dependent burst of reactive oxygen species.
Curr. Neurovasc. Res. 6:213–22. doi:10.2174/
156720209789630375.
Corveloni, A. C., S. C. Semprebon, A. Baranoski, B. I. Biazi,
T. A. Zanetti, and M. S. Mantovani. 2020. Carnosic acid
exhibits antiproliferative and proapoptotic effects in
tumoral NCI-H460 and nontumoral IMR-90 lung cells.
J. Toxicol. Environ. Health A 83:412–21. doi:10.1080/
15287394.2020.1767741.
da Rocha, C. Q., -F. M. D.-F. L. Marcourt, S. N. Ebrahimi,
B. T. Kitano, A. F. Ghilardi, A. L. Ferreira, A. C. A. de
Almeida, R. J. Dunder, A. R. M. Souza-Brito, et al. 2017.
Gastroprotective effects of hydroethanolic root extract of
Arrabidaea brachypoda: Evidences of cytoprotection and
10 H. L. NUNES ET AL.
isolation of unusual glycosylated polyphenols.
Phytochemistry 135:93–105. doi:10.1016/j.
phytochem.2016.12.002.
da Rocha, C. Q., E. F. Queiroz, C. S. Meira, D. R. M. Moreira,
M. B. P. Soares, L. Marcourt, W. Vilegas, and J.-
L. Wolfender. 2014. Dimeric flavonoids from Arrabidaea
brachypoda and assessment of their anti-Trypanosoma cruzi
activity. J. Nat. Prod. 77:1345–50. doi:10.1021/np401060j.
da Rocha, C. Q., F. C. Vilela, G. P. Cavalcante, F. V. Santa-Cecília,
L. Santos-e-Silva, M. H. Dos Santos, and A. Giusti-Paiva. 2011.
Anti-inflammatory and antinociceptive effects of Arrabidaea
brachypoda (DC.) bureau roots. J. Ethnopharmacol.
133:396–401. doi:10.1016/j.jep.2010.10.009.
de Sousa Andrade, L. M., A. B. M. de Oliveira, A. L. A. B. Leal,
F. A. de Alcântara Oliveira, A. L. Portela, J. de Sousa Lima
Neto, J. P. de Siqueira-júnior, G. W. Kaatz, C. Q. da Rocha,
and H. M. Barreto. 2020. Antimicrobial activity and inhibi-
tion of the NorA efflux pump of Staphylococcus aureus by
extract and isolated compounds from Arrabidaea
brachypoda. Microb. Pathog. 140:103935. doi:10.1016/j.
micpath.2019.103935.
Dirks, W. G., S. Faehnrich, I. A. J. Estella, and H. G. Drexler.
2005. Short tandem repeat DNA typing provides an inter-
national reference standard for authentication of human
cell lines. ALTEX 22:103–09.
Eastmond, D. A., A. Hartwig, D. Anderson, W. A. Anwar,
M. C. Cimino, I. Dobrev, G. R. Douglas, T. Nohmi,
D. H. Phillips, and C. Vickers. 2009. Mutagenicity testing
for chemical risk assessment: Update of the WHO/IPCS
harmonized scheme. Mutagenesis 24:341–49. doi:10.1093/
mutage/gep014.
Gontijo, D. C., M. A. N. Diaz, G. C. Brandão, P. C. Gontijo,
A. B. de Oliveira, L. G. Fietto, and J. P. V. Leite. 2018.
Phytochemical characterization and antioxidant, antibac-
terial and antimutagenic activities of aqueous extract from
leaves of Alchornea glandulosa. J. Toxicol. Environ. Health A
81:805–18. doi:10.1080/15287394.2018.1492479.
González-Vallinas, M., G. Reglero, and A. Ramírez de Molina.
2015. Rosemary (Rosmarinus ocinalis L.) extract as
a potential complementary agent in anticancer therapy. Nutr.
Cancer 67:1221–29. doi:10.1080/01635581.2015.1082110.
Huang, W.-Y., Y.-Z. Cai, and Y. Zhang. 2009. Natural pheno-
lic compounds from medicinal herbs and dietary plants:
Potential use for cancer prevention. Nutr. Cancer 62:1–20.
doi:10.1080/01635580903191585.
Kaehler, M. F. D. B. 2020. Accessed June 12, 2020. http://
reflora.jbrj.gov.br/reflora/floradobrasil/FB113396.
King, D., D. Yeomanson, and H. E. Bryant. 2015. PI3King
the lock: Targeting the PI3K/Akt/mTOR pathway as
a novel therapeutic strategy in neuroblastoma.
J. Pediatr. Hematol. Oncol. 37:245–51. doi:10.1097/
MPH.0000000000000329.
Maggiora, G., M. Vogt, D. Stumpfe, and J. Bajorath. 2014.
Molecular similarity in medicinal chemistry. J. Med.
Chem. 57:3186–204. doi:10.1021/jm401411z.
Maistro, E. L., P. M. Terrazzas, F. F. Perazzo, I. O. D. M. Gaivão,
A. C. H. F. Sawaya, and P. C. P. Rosa. 2019. Salix alba (white
willow) medicinal plant presents genotoxic effects in human
cultured leukocytes. J. Toxicol. Environ. Health A 82:1223–34.
doi:10.1080/15287394.2019.1711476.
Majolo, F., S. Bitencourt, B. Wissmann Monteiro, G. Viegas
Haute, C. Alves, J. Silva, S. Pinteus, R. C. V. Santos,
H. F. V. Torquato, E. J. Paredes-Gamero, et al. 2020.
Antimicrobial and antileukemic effects: In vitro activity of
Calyptranthes grandifolia aqueous leaf extract. J. Toxicol.
Environ. Health A 83:289–301. doi:10.1080/
15287394.2020.1753606.
Martínez-López, W., E. Marotta, M. V. Di Tomaso,
L. Méndez-Acuña, and F. Palitti. 2010. Distribution of
UVC-induced chromosome aberrations along the
X chromosome of TCR deficient and proficient Chinese
hamster cell lines. Mutat. Res. 701:98–102. doi:10.1016/j.
mrgentox.2010.02.008.
Morales, J., L. Li, F. J. Fattah, Y. Dong, E. A. Bey, M. Patel,
J. Gao, and D. A. Boothman. 2014. Review of poly
(ADP-ribose) polymerase (PARP) mechanisms of action
and rationale for targeting in cancer and other diseases.
Crit. Rev. Eukaryot. Gene Expr. 24:15–28. doi:10.1615/
critreveukaryotgeneexpr.2013006875.
Mosmann, T. 1983. Rapid colorimetric assay for cellular
growth and survival: Application to proliferation and cyto-
toxicity assays. J. Immunol. Meth. 65:55–63. doi:10.1016/
0022-1759(83)90303-4.
Namekawa, T., K. Ikeda, K. Horie-Inoue, and S. Inoue. 2019.
Application of prostate cancer models for preclinical study:
Advantages and limitations of cell lines, patient-derived
xenografts, and three-dimensional culture of
patient-derived cells. Cells 8:74. doi:10.3390/cells8010074.
Nitulescu, G. M., M. Van De Venter, G. Nitulescu,
A. Ungurianu, P. Juzenas, Q. Peng, O. T. Olaru,
D. Grădinaru, A. Tsatsakis, D. Tsoukalas, et al. 2018. The
Akt pathway in oncology therapy and beyond (Review). Int.
J. Oncol. 53:2319–31. doi:10.3892/ijo.2018.4597.
OECD. 2016. Test No. 489: In vivo mammalian alkaline comet
assay. In OECD guidelines for the testing of chemicals, sec-
tion 4, Paris: OECD Publishing. 1–27.
Pezaro, C., H. H. Woo, and I. D. Davis. 2014. Prostate cancer:
measuring PSA. Intern Med J 44:433–40. doi:10.1111/
imj.12407.
Proietti De Santis, L., A. S. Balajee, C. L. Garcia, G. Pepe,
A. M. Worboys, and F. Palitti. 2003. Inhibition of p53,
p21 and Bax by pifithrin-alpha does not affect UV induced
apoptotic response in CS-B cells. DNA Repair (Amst.)
2:891–900. doi:10.1016/s1568-7864(03)00088-0.
Repetto, G., A. Del Peso, and J. L. Zurita. 2008. Neutral
red uptake assay for the estimation of cell viability/
cytotoxicity. Nat. Protoc. 3:1125–31. doi:10.1038/
nprot.2008.75.
Resende, F. A., C. H. Nogueira, L. G. Espanha, P. K. Boldrin,
A. P. Oliveira-Höhne, M. S. de Camargo, C. Q. da Rocha,
W. Vilegas, and E. A. Varanda. 2017. In vitro toxicological
assessment of Arrabidaea brachypoda (DC.) Bureau:
Mutagenicity and estrogenicity studies. Regul. Toxicol.
Pharmacol. 90:29–35. doi:10.1016/j.yrtph.2017.08.010.
JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH, PART A 11
Rocha, T. A., D. F. de Moura, M. M. da Silva, T. G. Dos Santos
Souza, M. A. D. C. de Lira, D. D. M. Barros, A. G. da Silva,
R. M. Ximenes, E. P. D. S. Falcão, C. A. Chagas, et al. 2019a.
Evaluation of cytotoxic potential, oral toxicity, genotoxicity,
and mutagenicity of organic extracts of Pityrocarpa
moniliformis. J. Toxicol. Environ. Health Part A 82:216–31.
doi:10.1080/15287394.2019.1576563.
Rocha, V. P. C., C. Q. da Rocha, E. Ferreira Queiroz,
L. Marcourt, W. Vilegas, G. B. Grimaldi, P. Furrer,
É. Allémann, J.-L. Wolfender, and M. B. P. Soares. 2019b.
Antileishmanial activity of dimeric flavonoids isolated from
Arrabidaea brachypoda. Molecules 24:1. doi:10.3390/
molecules24010001.
Rodrigues, E., and E. A. Carlini. 2005. Ritual use of plants with
possible action on the central nervous system by the Kraho
Indians, Brazil. Phytother. Res. 19:129–35. doi:10.1002/
ptr.1636.
Rodrigues, V. P., C. Q. da Rocha, L. L. Périco,
R. D. C. D. Santos, R. Ohara, C. M. Nishijima, E. Ferreira
Queiroz, J.-L. Wolfender, L. R. M. da Rocha, A. R. S. Santos,
et al. 2017. Involvement of opioid system, TRPM8, and
ASIC receptors in antinociceptive effect of Arrabidaea bra-
chypoda (DC) Bureau. Int. J. Mol. Sci. 18:2304. doi:10.3390/
ijms18112304.
Russo, M., C. Spagnuolo, I. Tedesco, and G. L. Russo. 2010.
Phytochemicals in cancer prevention and therapy: truth or
dare? Toxins (Basel) 2:517–51. doi:10.3390/toxins2040517.
Ryhänen, T., J. M. T. Hyttinen, J. Kopitz, K. Rilla, E. Kuusisto,
E. Mannermaa, J. Viiri, C. I. Holmberg, I. Immonen,
S. Meri, et al. 2009. Crosstalk between Hsp70 molecular
chaperone, lysosomes and proteasomes in
autophagy-mediated proteolysis in human retinal pigment
epithelial cells. J. Cell. Mol. Med. 13:3616–31. doi:10.1111/
j.1582-4934.2008.00577.x.
Sasaki, Y. F., T. Nakamura, and S. Kawaguchi. 2007. What is
better experimental design for in vitro comet assay to detect
chemical genotoxicity? AATEX 14:499–504.
Serpeloni, J. M., A. Specian, D. L. Ribeiro, L. M. Benício,
H. L. Nunes, L. P. Franchi, C. Q. Rocha, W. Vilegas,
E. A. Varanda, and I. Cólus. 2020. Fridericia platyphylla
(Cham.) L.G. Lohmann root extract exerts cytotoxic and
antiproliferative effects on gastric tumor cells and down-
regulates BCL-XL, BIRC5, and MET genes. Human Exp.
Toxicol. 39:338–54. doi:10.1177/0960327119888261.
Serpeloni, J. M., A. F. L. Specian, D. L. Ribeiro, K. Tuttis,
W. Vilegas, W. Martínez-López, A. L. Dokkedal,
L. L. Saldanha, I. M. D. S. Cólus, and E. A. Varanda. 2015.
Antimutagenicity and induction of antioxidant defense by
flavonoid rich extract of Myrcia bella Cambess. in normal
and tumor gastric cells. J. Ethnopharmacol. 176:345–55.
doi:10.1016/j.jep.2015.11.003.
Siegel, R. L., K. D. Miller, and A. Jemal. 2016. Cancer statistics,
2016. CA Cancer J. Clin. 66:7–30. doi:10.3322/caac.21332.
Sikes, R. A. 2007. Chemistry and pharmacology of anticancer
drugs. Br. J. Cancer 97:1713. doi:10.1038/sj.bjc.6604075.
Singh, N. P., M. T. McCoy, R. R. Tice, and E. L. Schneider.
1988. A simple technique for quantitation of low levels of
DNA damage in individual cells. Exp. Cell Res. 175:184–91.
doi:10.1016/0014-4827(88)90265-0.
Specian, A. F. L., J. M. Serpeloni, K. Tuttis, D. L. Ribeiro,
H. L. Cilião, E. A. Varanda, M. Sannomiya, W. Martinez-
Lopez, W. Vilegas, and I. M. S. Cólus. 2016. LDH, prolif-
eration curves and cell cycle analysis are the most suitable
assays to identify and characterize new phytotherapeutic
compounds. Cytotechnology 68:2729–44. doi:10.1007/
s10616-016-9998-6.
Tice, R. R., E. Agurell, D. Anderson, B. Burlinson,
A. Hartmann, H. Kobayashi, Y. Miyamae, E. Rojas, J.-
C. Ryu, and Y. F. Sasaki. 2000. Single cell gel/comet assay:
Guidelines for in vitro and in vivo genetic toxicology testing.
Environ. Mol. Mutagen. 35:206–21. doi:10.1002/(SICI)
1098-2280(2000)35:3<206::AID-EM8>3.0.CO;2-J.
Tuttis, K., D. L. M. G. da Costa, H. L. Nunes, A. F. L. Specian,
J. M. Serpeloni, L. C. Dos Santos, E. A. Varanda, W. Vilegas,
W. Martínez-Lopez, and I. M. D. S. Cólus. 2018. Pouteria
ramiora (Mart.) Radlk. extract: Flavonoids quantification
and chemopreventive effect on HepG2 cells. J. Toxicol.
Environ. Health A 81:792–804. doi:10.1080/
15287394.2018.1491911.
Woodrum, D. A., A. Kawashima, K. R. Gorny, and
L. A. Mynderse. 2017. Prostate cancer: State of the art
imaging and focal treatment. Clin. Radiol. 72:665–79.
doi:10.1016/j.crad.2017.02.010.
Yesil-Celiktas, O., C. Sevimli, E. Bedir, and F. Vardar-Sukan.
2010. Inhibitory effects of rosemary extracts, carnosic acid
and rosmarinic acid on the growth of various human cancer
cell lines. Plant Foods Human Nutr. 65:158–63. doi:10.1007/
s11130-010-0166-4.
12 H. L. NUNES ET AL.
... It was recently reported that crude hydroethanolic extracts and the three brachydins separately exhibited antitumor activity (Nunes et al. 2020, Serpeloni et al. 2020. However, the mechanisms by which these uncommon flavonoids act as antitumor agents remain unclear. ...
... Regarding cytotoxicity, the DCMF significantly reduced the viability of tumor cells, corroborating with previous studies in which the antitumor activity was demonstrated for extracts (Serpeloni et al. 2020) and for each of the three brachydins separately (Nunes et al. 2020, de Oliveira et al. 2021). Recently, de Lima et al. (2022) described this ability also in glycosylated brachydins. ...
... Our results agree with other authors who evaluated the genotoxic or mutagenic potential of brachydins. Resende et al. (2017) reported that the DCMF did not cause mutations in bacteria, while Nunes et al. (2020) tested each brachydin individually and did not detect genotoxicity. DNA damage-inducing chemotherapeutics maintain a prominent position among the strategies used to treat cancer. ...
Article
Notwithstanding the advances in molecular target-based drugs, chemotherapy remains the most common cancer treatment, despite its high toxicity. Consequently, effective anticancer therapies with fewer adverse effects are needed. Therefore, this study aimed to determine the anticancer activity of the dichloromethane fraction (DCMF) isolated from Arrabidae brachypoda roots, whose components are three unusual dimeric flavonoids. The toxicity of DCMF was investigated in breast (MCF-7), prostate (DU145), and cervical (HeLa) tumor cells, as well as non-tumor cells (PNT2), using sulforhodamine B (cell viability), Comet (genotoxicity), clonogenicity (reproductive capacity) and wound healing (cell migration) assays, and atomic force microscopy (AFM) for ultrastructural cell membrane alterations. Molecular docking revealed affinity between albumin and each rare flavonoid, supporting the impact of fetal bovine serum in DCMF antitumor activity. The IC50 values for MCF7, HeLa, and DU145 were 2.77, 2.46, and 2.51 µg/mL, respectively, and 4.08 µg/mL for PNT2. DCFM was not genotoxic to tumor or normal cells when exposed to twice the IC50 for up to 24 h, but it inhibited tumor cell migration and reproduction compared to normal cells. Additionally, AFM revealed alterations in the ultrastructure of tumor nuclear membrane surfaces, with a positive correlation between DCMF concentration and tumor cell roughness. Finally, we found a negative correlation between roughness and the ability of DCMF-treated tumor cells to migrate and form colonies with more than 50 cells. These findings suggest that DCFM acts by causing ultrastructural changes in tumor cell membranes while having fewer toxicological effects on normal cells.
... Among them are two glycosylated phenylethanoid derivatives, seven glycosylated dimeric flavonoids, and three previously unreported dimeric flavonoids that are rare within the Bignoniaceae family. These newly discovered flavonoids have been named brachydins 1, 2, and 3 [7,8] and have been reported in the literature with promising activity against promastigote and amastigote forms of Leishmania amazonensis [9], against prostate tumor cells PC-3 [10], metastatic prostate cancer (DU145) in 2D and 3D cells [11][12][13][14][15][16], with anti-inflammatory activity [17], microbial activity by inhibiting the NorA gene in Staphylococcus aureus strain [18], and antiproliferative activity in gastric tumors [19]. ...
Article
Full-text available
Fridericia platyphylla (Cham.) L.G. Lohmann is a species native to the Brazilian cerrado, with promising bioactivity. The organic fraction of the roots is rich in unusual dimeric flavonoids, reported as potential candidates for cancer treatment. The exploration of these flavonoids is very important, considering their diverse biological activities and the need for innovative therapeutic options. This work aimed to develop and characterize a microemulsion loaded with a non-polar fraction (DCM). The constituents were chosen, and the pseudo-ternary diagram was constructed to determine the region of microemulsion formation. The microemulsions blank (ME), with 3% (ME3) and 5% (ME5) of fraction DCM, were characterized in terms of droplet size, zeta potential, and polydispersity index. Both MEs showed particle sizes <100 nm; only ME3 exhibited better values for polydispersity index and zeta potential and was therefore selected for further study. The organoleptic and physicochemical characteristics were evaluated, revealing limpidity and transparency typical of these microstructures, physiologically acceptable pH, refractive index of 1.42±0.01, and density of 1.017 g/cm3±0.01. The stability tests showed good stability profiles even after exposure to extreme thermal conditions, with minimal changes in pH and the content of the incorporated fraction. The in vitro release study demonstrated that ME3 enabled the controlled release of the fraction, with a cumulative amount released over 60% within 6 h. Furthermore, fraction DCM and ME3 exhibited no toxicity in Tenebrio molitor larvae. The developed microemulsion exhibited excellent properties, so this study represents the first successful attempt to develop a formulation that incorporates the dimeric flavonoid fraction.
... Plants have been characterized as an important source of pharmaceutical products with therapeutic or prophylactic properties which are associated with anti-inflammatory, antioxidant, antibacterial, antigenotoxic and antiproliferative activities of their bioactive compounds [1][2][3][4]. Due to their beneficial properties, their use for various therapeutic purposes has increased tremendously over the last decades worldwide. Presently, many plants are used as a source of ingredients to prevent and/or treat a plethora of diseases [5]. ...
Article
Full-text available
This study investigated the beneficial properties of prickly pear peel (PPP) extracts from Opuntia ficus-indica (L.) Mill. Extracts were obtained via the Soxhlet extraction method using methanol (P1), ethanol (P2) and ethanol-water (P3) as extraction solvents. Their total phenolic and flavonoid content (TPC and TFC, respectively) and their antioxidant activity (AA) were determined. The PPP extracts were characterized in detail using mass spectrometry techniques. Their cyto-genotoxic effect and antigenotoxic potential against mitomycin C were evaluated via the cytokinesis block micronucleus (CBMN) assay on human lymphocytes. Enhanced TPC, TFC and AA values were recorded for all the extracts. Moreover, P1 and P2 were cytotoxic only at the highest concentrations, whereas P3 was found to be cytotoxic in all cases. No significant micronucleus induction was observed in the tested extracts. The PPP extracts contain bioactive compounds such as flavonoids, carboxylic acids, alkaloids, fatty acids and minerals (mainly K, Si, Mg, Ca, P and Zn). The results showed that all three extracts exerted high antigenotoxic activity. Our findings confirm the beneficial and genoprotective properties of PPP extracts and further studies on the bioactive compounds of Opuntia ficus-indica (L.) Mill. are recommended, as it constitutes a promising plant in pharmaceutical applications.
... Searching for regional and national biodiversity, chemical richness and novel therapeutic options mean utilizing existing popular culture by adding value to traditional knowledge and investing in scientific and technological growth to improve quality of life and health of population (Pereira et al. 2015;Pereira-Freire et al. 2018;Silva et al. 2020;Valli and Bolzani 2019). To underline such therapeutic potential, preclinical investigations are required and several methods have been incorporated to predict general mechanisms and broadspectrum toxicity employing animal and plant cells, microalgae, invertebrates, and mammals (Almeida et al. 2020;Carvalho et al. 2016;Ferreira et al. 2009Ferreira et al. , 2019aNunes et al. 2020;Sbardelotto et al. 2021;Sousa et al. 2012). Herein, we used in vitro and in vivo methods were used to investigate possible toxicogenetic and antiproliferative properties of EtAcSur aerial parts on murine and vegetal cells. ...
Article
Stevia urticifolia Thunb. is an underexploited herb possessing bioactive flavonoids, saponins, and terpenoids. The aim of this study was to examine the antiproliferative and toxicogenetic properties of the ethyl acetate extract from Stevia urticifolia aerial parts (EtAcSur) upon Artemia salina, erythrocytes, Allium cepa and sarcoma 180 cells and fibroblasts, as well as in vivo studies on mice to determine systemic, macroscopic, and behavioral alterations and bone marrow chromosomal damage. The assessment using A. salina larvae and mouse blood cells revealed LC50 and EC50 values of 68.9 and 113.6 µg/ml, respectively. Root growth and mitosis were inhibited by EtAcSur, and chromosomal aberrations were detected only at 100 μg/ml. EtAcSur exhibited potent concentration-dependent viability reduction of S180 and L-929 cells and antioxidant capacity employing ABTS• and DPPH•. No previous in vivo studies were performed before with the EtAcSur. Signals of acute toxicity were not observed at 300 mg/kg. Physiological and toxicological investigations at 25 and 50 mg/mg/day i.p. for 8 days did not markedly change body or organ relative weights, nor patterns of spontaneous locomotor and exploratory activities. In contrast, clastogenic effects on bone marrow were found at 50 mg/mg/day. EtAcSur was found to (1) produce toxicity in microcrustaceans, (2) capacity as free radical scavenger, (3) antimitotic, cytotoxic and clastogenic activties upon vegetal and mammalian cells, and (4) lethality on both tumor and normal murine cells indistinctly. In vivo damage systemic effects were not remarkable and clinical signals of toxicity were not observed, suggesting the significant pharmacological potential of S. urticifolia for the development of antineoplastic agents.Abbreviations: ABTS: 2,2'-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid); DMSO: dimethylsulfoxide; DPPH: 1,1-diphenyl-2-picrylhydrazyl; EC50: effective concentration 50%; EtAcSur: ethyl acetate extract from Stevia urticifolia aerial parts; Hb, hemoglobin; IC50: inhibitory concentration 50%; LC50,: lethal concentration 50%; MI: mitotic index; RBC, red blood cells; Trolox: 6-hydroxy-2,5,7,8-tetramethylchroman-2-carboxylic acid.
Article
In the Mid-19th century, Rudolf Virchow considered necrosis to be a prominent form of cell death; since then, pathologists have recognized necrosis as both a cause and a consequence of disease. About a century later, the mechanism of apoptosis, another form of cell death, was discovered, and we now know that this process is regulated by several molecular mechanisms that "programme" the cell to die. However, discoveries on cell death mechanisms are not limited to these, and recent studies have allowed the identification of novel cell death pathways that can be molecularly distinguished from necrotic and apoptotic cell death mechanisms. Moreover, the main goal of current cancer therapy is to discover and develop drugs that target apoptosis. However, resistance to chemotherapeutic agents targeting apoptosis is mainly responsible for the failure of clinical therapy and adverse side effects of the chemotherapeutic agents currently in use pose a major threat to the well-being and lives of patients. Therefore, the development of natural-based anticancer drugs with low cellular and organismal side effects is of great interest. In this comprehensive review, we thoroughly examine and discuss natural anticancer compounds that specifically target non-canonical cell death mechanisms.
Article
Ethnopharmacological relevance: Aloysia gratissima leaves are popularly used to treat respiratory, digestive, and nervous system disorders. Several studies have been carried out to determine the biological activity of A. gratissima, such as its antibacterial and anti-edematogenic activities, but despite the beneficial uses of A. gratissima, few studies have examined the toxicological profile of this plant. Aim of the study: This study aimed to determine the chemical composition, cytotoxic, genotoxic, mutagenic potential, and antioxidant activity of an aqueous extract of A. gratissima leaves (AG-AEL). Material and methods: The phytochemical constitution of AG-AEL was assessed by colorimetric analyses and High-performance liquid chromatography (HPLC). The inorganic elements were detected by Particle-Induced X-ray Emission (PIXE). The antioxidant, cytotoxicity, genotoxic, and mutagenic activities were evaluated in vitro by Di(phenyl)-(2,4,6-trinitrophenyl)iminoazanium (DPPH), Sulforhodamine B (SRB) assay, comet assay, and Salmonella/microsome assays. Results: AG-AEL indicated the presence of terpenoids, flavonoids, and phenolic acids. HPLC detected rutin at 2.41 ± 0.33 mg/100 mg. PIXE analysis indicated the presence of Mg, Si, P, S, K, Ca, Mn, and Zn. The 50% inhibitory concentration was 84.17 ± 3.17 μg/mL in the DPPH assay. Genotoxic effects were observed using the Comet assay in neuroblastoma (SH-SY5Y) cells and mutations were observed in TA102 and TA97a strains. The extract showed cytotoxic activities against ovarian (OVCAR-3), glioblastoma (U87MG), and colon (HT-29) cancer cell lines. Conclusions: In conclusion, AG-AEL increased DNA damage, induced frameshift, and oxidative mutations, and showed cytotoxic activities against different cancer cells. The in vitro toxicological effects observed suggest that this plant preparation should be used with caution, despite its pharmacological potential.
Article
Brachydin B (BrB) is a unique dimeric flavonoid extracted from Fridericia platyphylla (Cham.) LG Lohmann with different biological activities. However, the antitumoral potential of this flavonoid is unclear. In our study, we evaluated the effects of the BrB flavonoid on cell viability (MTT, resazurin, and lactate dehydrogenase assays), proliferation (protein dosage and clonogenic assay), and migration/invasion (3D ECM gel, wound-healing, and transwell assays) of metastatic prostate (DU145) cells cultured both as traditional 2D monolayers and 3D tumor spheroids in vitro. The results showed that the BrB flavonoid promotes cytotoxic effects from ≥1.50 μM after 24 h of treatment in DU145 cells in monolayers. In 3D prostate tumor spheroids, BrB also induced cytotoxic effects at higher concentrations after longer treatment (48, 72, and 168 h). Furthermore, BrB treatment is associated with reduced DU145 clonogenicity in 2D cultures, as well as decreased area/volume of 3D tumor spheroids. Finally, BrB (6 μM) reduced cell migration/invasion in 2D monolayers and promoted antimigratory effects in DU145 tumor spheroids (≥30 μM). In conclusion, the antitumoral and antimigratory effects observed in DU145 cells cultured in 2D and 3D models are promising results for future studies with BrB using in vivo models and confirm this molecule as a candidate for metastatic prostate cancer therapy.
Preprint
Medicinal plants for bone grafts are promising because they are free from infecting microorganisms and biocompatible. We report a novel study evaluating osteoregeneration of bone grafts from extracts of Alternanthera brasiliana and Fridericia platyphylla after bone injury induced by radius fracture in rats. Grafts were obtained using 2% chitosan gel and 0.5% hydroalcoholic extract. The rats were randomly divided into four experimental groups (N= 12): Negative control (NC) - Chitosan gel; Positive control (PC) - bovine mineral bone graft (Lumina Bone®, fine powder 0.5); F. platyphylla Graft (FRID) - 0.5% bone graft; A. brasiliana Graft (ABRA) - 0.5% bone graft. The animals were evaluated for three periods, 30, 60, and 90 days after fracture induction. Bone alkaline phosphatase (BSAP) and radiographic and histological evaluations were followed. After 90 days, there was an increase in BSAP for the ABRA group on the newly formed bone matrix but not for FRID, indicating the presence of active osteoblasts. Additionally, mature bone tissue and bone remodeling were observed, and a conspicuous presence of type I collagen for both FRID and ABRA. We demonstrated that FRID and ABRA grafts produced early bone neoformation. New perspectives for these species as a graft are suggested.
Article
Brachydin C (BrC) has demonstrated in vitro cytotoxic and antiproliferative effects in prostate cancer cells. In the present study, we compare the anticancer effects of BrC in DU145 cells grown in common bidimensional cultures (2D) and multicellular tumor spheroids (MCTS), often denominated 3D in vitro models, that can better mimic the microenvironment of tissues. BrC IC50 values obtained in the resazurin assay after 24 h of treatment were 47.31 μM (2D) and 229.8 μM (3D) and these cytotoxic effects were time dependent only in 3D. BrC (5 to 60 μM) interfered with the growth of MCTS and reduced cell viability after 11 days of treatment, a result that is not attributable to oxidative stress evaluated using the CM‐H2DCFDA probe. BrC (6.0 μM) impaired horizontal (wound healing) and vertical cell migration and invasion (transwell assay) in 2D and BrC (5.0 to 60 μM) in 3D (ECM Gel®). BrC modulated the expression of genes BIRC5, TNF‐α, CASP3, NKX3.1, MMP9, MMP11, CDH1, and ITGAM and downregulated proteins CASP7, BAX, and TNF‐α in western blotting analysis. In conclusion, BrC stimulated cell death and decreased epithelial‐mesenchymal transition. Furthermore, DU145 MCTS displayed higher resistance to BrC‐ induced cell death than 2D cultures, a difference that should be considered in future approaches in prostatic cancer studies. BrC stimulated cell death modulating BIRC5, CASP3, and CASP7. BrC decreased epithelial‐mesenchymal transition and cell migration, modulating the MMP9, MMP11, CDH1, and ITGAM. Furthermore, DU145 MCTS displayed higher resistance to BrC‐ induced cell death than 2D cultures, a difference to be considered in future approaches in prostatic cancer studies.
Article
Full-text available
Fridericia platyphylla (Cham.) L.G. Lohmann (FP) has cytotoxic, anti-inflammatory, and analgesic properties. We aimed to characterize the cytotoxic and antiproliferative effects of FP extract on normal (GAS) and tumor-derived (ACP02 and HepG2) cell lines. The effective concentrations (EC 50 s) by tetrazolium bromide assay (MTT) were 56.16, 43.68, and 42.57 µg mL ⁻¹ and 69.38, 41.73, and 52.39 µg mL ⁻¹ by neutral red assay for GAS, ACP02, and HepG2 cells, respectively. The extract decreased nuclear division indices, which was not reflected in cell proliferation curves. Flow cytometric analyses showed that even 30 µg mL ⁻¹ extract (shown to be noncytotoxic by MTT assay) increased the sub-G1 population, indicating cell death due to apoptosis and necrosis. A cytokinesis-block micronucleus cytome assay showed that 30 µg mL ⁻¹ of the extract increased the frequency of nuclear buds in tumor cells. Real-time quantitative polymerase chain reaction showed CCND1 upregulation in doxorubicin-treated GAS cells and BCL-XL, BIRC5, and MET downregulation in 5 or 30 µg mL ⁻¹ in FP extract-treated ACP02 cells. In conclusion, FP extract modulated apoptosis- and cell cycle-related genes and presented selective cytotoxicity toward tumor cells that deserves further investigation by testing other cell types. Our results demonstrated that even medicinal plants exert adverse effects depending on the extract concentrations used and tissues investigated.
Article
Full-text available
We synthesized eight new bipyridine and bipyrimidine gold (III) dithiocarbamate-containing complexes (C1–C8) and tested them in a panel of human cancer cell lines. We used osteosarcoma (MG-63), lung (A549), prostate (PC3 and DU145), breast (MCF-7), ovarian (A2780 and A2780cis, cisplatin- and doxorubicin-resistant), and cervical (ME-180 and R-ME-180, cisplatin resistant) cancer cell lines. We found that C2, C3, C6, and C7 were more cytotoxic than cisplatin in all cell lines tested and overcame cisplatin and doxorubicin resistance in A2780cis and R-ME-180 cells. In the PC3 prostate cancer cell line, the gold (III) complex C6 ([Au2(BPM)(DMDTC)2]Cl4) induced apoptosis and double-stranded DNA breaks, modified cell cycle phases, increased Reactive Oxigen Species (ROS) generation, and reduced thioredoxin reductase and proteasome activities. It inhibited PC3 cell migration and was more cytotoxic against PC3 cells than normal human adipose-derived stromal cells. In mice bearing PC3 tumor xenografts, C6 reduced tumor growth by more than 70% without causing weight loss. Altogether, our results demonstrate the anticancer activity of these new gold (III) complexes and support the potential of C6 as a new agent for prostate cancer treatment.
Article
Full-text available
Various preclinical models have been developed to clarify the pathophysiology of prostate cancer (PCa). Traditional PCa cell lines from clinical metastatic lesions, as exemplified by DU-145, PC-3, and LNCaP cells, are useful tools to define mechanisms underlying tumorigenesis and drug resistance. Cell line-based experiments, however, have limitations for preclinical studies because those cells are basically adapted to 2-dimensional monolayer culture conditions, in which the majority of primary PCa cells cannot survive. Recent tissue engineering enables generation of PCa patient-derived xenografts (PDXs) from both primary and metastatic lesions. Compared with fresh PCa tissue transplantation in athymic mice, co-injection of PCa tissues with extracellular matrix in highly immunodeficient mice has remarkably improved the success rate of PDX generation. PDX models have advantages to appropriately recapitulate the molecular diversity, cellular heterogeneity, and histology of original patient tumors. In contrast to PDX models, patient-derived organoid and spheroid PCa models in 3-dimensional culture are more feasible tools for in vitro studies for retaining the characteristics of patient tumors. In this article, we review PCa preclinical model cell lines and their sublines, PDXs, and patient-derived organoid and spheroid models. These PCa models will be applied to the development of new strategies for cancer precision medicine.
Article
Full-text available
Leishmaniasis are diseases caused by parasites belonging to Leishmania genus. The treatment with pentavalent antimonials present high toxicity. Secondary line drugs, such as amphotericin B and miltefosine also have a narrow therapeutic index. Therefore, there is an urgent need to develop new drugs to treat leishmaniasis. Here, we present the in vitro anti-leishmanial activity of unusual dimeric flavonoids purified from Arrabidaea brachypoda. Three compounds were tested against Leishmana sp. Compound 2 was the most active against promastigotes. Quantifying the in vitro infected macrophages revealed that compound 2 was also the most active against intracellular amastigotes of L. amazonensis, without displaying host cell toxicity. Drug combinations presented an additive effect, suggesting the absence of interaction between amphotericin B and compound 2. Amastigotes treated with compound 2 demonstrated alterations in the Golgi and accumulation of vesicles inside the flagellar pocket. Compound 2-treated amastigotes presented a high accumulation of cytoplasmic vesicles and a myelin-like structure. When administered in L. amazonensis-infected mice, neither the oral nor the topical treatments were effective against the parasite. Based on the high in vitro activity, dimeric flavonoids can be used as a lead structure for the development of new molecules that could be useful for structure-active studies against Leishmania.
Article
Full-text available
Protein kinase B (Akt), similar to many other protein kinases, is at the crossroads of cell death and survival, playing a pivotal role in multiple interconnected cell signaling mechanisms implicated in cell metabolism, growth and division, apoptosis suppression and angiogenesis. Akt protein kinase displays important metabolic effects, among which are glucose uptake in muscle and fat cells or the suppression of neuronal cell death. Disruptions in the Akt‑regulated pathways are associated with cancer, diabetes, cardiovascular and neurological diseases. The regulation of the Akt signaling pathway renders Akt a valuable therapeutic target. The discovery process of Akt inhibitors using various strategies has led to the identification of inhibitors with great selectivity, low side‑effects and toxicity. The usefulness of Akt emerges beyond cancer therapy and extends to other major diseases, such as diabetes, heart diseases, or neurodegeneration. This review presents key features of Akt structure and functions, and presents the progress of Akt inhibitors in regards to drug development, and their preclinical and clinical activity in regards to therapeutic efficacy and safety for patients.
Article
Carnosic acid (CA) is a phenolic diterpene with many important biological activities including antimicrobial, antioxidant, anti-inflammatory properties, and anti-proliferative properties. The aim of the present study was to investigate cytotoxic activity, cell cycle, apoptotic, and molecular effects attributed to CA in non-tumoral IMR-90 (human fetal lung fibroblasts), as well as tumoral NCI-H460 (human non–small-cell lung cancer) cell lines. Cell proliferation was evaluated by Real-Time Cell Analysis system, while apoptosis and cell cycle were assessed using flow cytometry. RT-qPCR was used to estimate the relative expression of genes involved in cell cycle regulation, DNA damage and repair, and apoptosis induction. CA inhibited proliferation of IMR-90 and NCI-H460 cells via cell cycle arrest at G0/G1 and G2/M phases, according to the treatment concentration. The mRNA levels of genes encoding cyclins A2, B1, and B2 were downregulated in response to CA treatment of IMR-90 cells. Apoptosis was induced and proapoptotic gene PUMA was upregulated in both cell lines. mRNA levels of genes ATR, CCND1, CHK1, CHK2, MYC, GADD45A, H2AFX, MTOR, TP53, and BCL2, CASP3 were not markedly changed following CA treatments. Although CA exerted antiproliferative activity against NCI-H460 tumor cells, this phytochemical induced toxic effects in non-tumoral cells, and thus needs to be considered carefully prior to pharmacological use therapeutically.
Article
Natural products are still a promising source of bioactive molecules. Food and Drug Administration data showed that approximately 49% of the approved molecules originate naturally or chemically-resemble these substances, of which more than 70% are being used in anticancer therapy. It is noteworthy that at present there are no scientific studies to prove the effectiveness and safety of a number of plants used in folk medicine such as in the case of Calyptranthes grandifolia O. Berg (Myrtaceae) originally from South America. The aim of the present study was to determine the biological potential and toxicological effects of the aqueous leaf extract of C. grandifolia. The main detected phytoconstituents were condensed tannins and flavonoids and a high quantity of polyphenols. Regarding the antimicrobial potential, the extract exerted inhibitory activity against Pseudomonas aeruginosa. The results also revealed the extract induced DNA damage in a concentration-dependent manner in RAW 264.7 cells. In addition, C. grandifolia produced cytotoxicity in leukemia cell lines (HL60 and Kasumi-1) without affecting isolated human lymphocytes but significantly inhibited JAK3 and p38α enzyme activity. Taken together, these findings add important information on the biological and toxicological effects of C. grandifolia, indicating that aqueous extract may be a source of natural antimicrobial and antileukemic constituents.
Article
Salix alba (SA), commonly known as white willow, is a plant used in folk medicine for the treatment of chronic and acute inflammation, infection, pain, and fever. The phytochemical characterization of the bark extract of this plant indicated that its main component is salicin, a precursor of the anti-inflammatory agent acetylsalicylic acid. Considering the lack of studies evaluating the genetic toxicity and cytotoxic action of SA bark extract on human cells, as well as the chemical characterization of its major phenolic compounds, the present study was designed to (1) investigate the cytotoxic and genotoxic potential of SA bark extract on human peripheral leukocyte cells and human hepatoma cell line HepG2, and (2) characterize its major phenolic constituents. The phenolic compounds found were salicylic acid, salicin, salidroside, saligenin, tremulodin, salicoylsalicin, salicortin, and tremulacin. The results using trypan blue staining test showed viability decreases (viability less than 70%) for concentrations of SA extract equal and higher to 200 µg/ml. Low genotoxic activity (comet assay) was exhibited for 50 and 100 µg/ml SA extract in human leukocytes. SA did not exert a marked clastogenic/aneugenic effect on leukocytes and HepG2 human cells. Data suggest that the genotoxic effects of SA bark extract occur when it is not metabolized by liver enzymes.
Article
Arrabidaea brachypoda is a native shrub of the Brazilian Cerrado widely used in the folk medicine for treatment of renal diseases and articular pains. This study aimed to, first, evaluate the antimicrobial activity of both extracts and isolated molecules Brachydins BR-A and BR-B obtained from the flowers of A. brachypoda against Staphylococcus aureus, Escherchia coli and Candida albicans species. A second objective was to investigate if these natural products were able to potentiate the Norfloxacin activity against the strain Staphylococcus aureus SA1199-B that overexpress the norA gene encoding the NorA efflux pump. Extracts and isolated compounds were analysed by HPLC-PDA and LC-ESI-MS respectively. Minimal inhibitory concentrations of Norfloxacin or Ethidium Bromide (EtBr) were determined in the presence or absence of ethanolic extract, dichloromethane fraction, as well as BR-A or BR-B by microdilution method. Only BR-B showed activity against Candida albicans. Addition of ethanolic extract, dichloromethane fraction or BR-B to the growth media at sub-inhibitory concentrations enhanced the activity of both Norfloxacin and EtBr against S. aureus SA1199-B, indicating that these natural products and its isolated compound BR-B were able to modulate the fluoroquinolone-resistance possibly by inhibition of NorA. Moreover, BR-B inhibited the EtBr efflux in the SA1199-B strain confirming that it is a NorA inhibitor. Isolated BR-B was able to inhibit an important mechanism of multidrug-resistance very prevalent in S. aureus strains, thus its use in combination with Norfloxacin could be considered as an alternative for the treatment of infections caused by S. aureus strains overexpressing norA.
Article
The objective of this study was to determine the cytotoxicity of organic extracts of P. moniliformis in vitro and identify the acute toxicity and genotoxicity in vivo. The leaves were extracted using three organic solvents (cyclohexane [EP1], ethyl acetate [EP2], and methanol [EP3]). Phytochemical qualitative analysis was performed by thin layer chromatography (TLC). Cytotoxicity tests were performed on human embryonic kidney (HEK) cells and J774 murine macrophages. Acute toxicity in mice was measured after intraperitoneal (ip) administration of 2000 mg/kg, while evaluation of genotoxicity and mutagenicity were assessed using the comet assay and the micronucleus (MN) test, respectively. The TLC analysis of the extracts revealed the presence of flavonoids, triterpenes, steroids, and saponins. In the cytotoxicity assay, extracts EP1 and EP3 altered proliferation of HEK cells, and all organic extracts increased the viability of J774 cells. In the toxicity tests, no deaths or behavioral alterations were observed in mice exposed to the acute dose of the extracts. Although some extracts led to changes in hematological and histological parameters, these results did not indicate physiological changes. In relation to the MN test and comet assay, no significant changes were detected in the DNA of the animals tested with the extracts EP1, EP2, and EP3. Thus, extracts of P. moniliformis were not considered to be toxic and did not induce formation of MN or damage to cellular DNA in the genotoxicity tests.