ArticlePDF Available

Abstract and Figures

Angiogenesis is the process of vascular network development and plays a crucial role in cancer growth, progression, and metastasis. Phthalates are a class of environmental pollutants that have detrimental effects on human health and are reported to increase cancer risk. However, the interplay between phthalate exposure and angiogenesis has not been investigated thoroughly. In this study, we investigated the effect of prolonged di (2-ethylhexyl) phthalate (DEHP) treatment on the angiogenic potential of triple-negative breast cancer. MDA-MB-231 cells were exposed to physiological concentrations of DEHP for more than three months. Prolonged DEHP exposure induced angiogenesis in breast cancer cells. Endoglin (ENG)/CD105 is a membrane glycoprotein and an auxiliary receptor of the TGFβ receptor complex. In endothelial cells, ENG is highly expressed and it is a prerequisite for developmental angiogenesis. A literature review highlights endoglin as a well known mesenchymal stem cell marker responsible for vascular development and angiogenesis. NGS analysis showed that endoglin overexpression in DEHP-exposed MDA-MB-231 cells correlated with tumor development and growth. An in vivo zebrafish xenograft assay showed that VEGFA induced sprouting of the subintestinal vein (SIV) in embryos injected with DEHP-exposed cells. Endoglin knockdown reduced SIV sprouting and VEGFA expression in zebrafish embryos. An in vitro HUVEC tube formation assay showed that endoglin depletion reversed DEHP-induced VEGF-mediated HUVEC tube formation in coculture. DEHP-induced endoglin activated TGFβ/SMAD3/VEGF and MAPK/p38 signaling in MDA-MB-231 cells. A cytokine angiogenesis antibody array showed induced expression of the inflammatory cytokines IL1α, IL1β, IL6, and IL8, along with GMCSF and VEGF. Endoglin knockdown reversed DEHP-induced activation of the TGFβ/SMAD3/VEGF signaling axis, MAPK/p38 signaling, and cytokine regulation, limiting angiogenesis potential both in vivo and in vitro. Targeting endoglin might serve as a potential alternative treatment to control angiogenesis, leading to metastasis and limiting cancer progression. Biomedicines
This content is subject to copyright.


Citation: Jadhao, M.; Chen, C.-L.;
Liu, W.; Deshmukh, D.; Liao, W.-T.;
Chen, J.Y.-F.; Urade, R.; Tsai, E.-M.;
Hsu, S.-K.; Wang, L.-F.; et al.
Endoglin Modulates TGFβR2
Induced VEGF and Proinflammatory
Cytokine Axis Mediated
Angiogenesis in Prolonged
DEHP-Exposed Breast Cancer Cells.
Biomedicines 2022,10, 417.
https://doi.org/10.3390/
biomedicines10020417
Academic Editor: Randolph
C. Elble
Received: 22 December 2021
Accepted: 3 February 2022
Published: 10 February 2022
Publisher’s Note: MDPI stays neutral
with regard to jurisdictional claims in
published maps and institutional affil-
iations.
Copyright: © 2022 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
biomedicines
Article
Endoglin Modulates TGFβR2 Induced VEGF and
Proinflammatory Cytokine Axis Mediated Angiogenesis in
Prolonged DEHP-Exposed Breast Cancer Cells
Mahendra Jadhao 1, Chun-Lin Chen 2,3 , Wangta Liu 4, Dhanashri Deshmukh 1, Wei-Ting Liao 4,
Jeff Yi-Fu Chen 4, Ritesh Urade 2, Eing-Mei Tsai 5,6, Sheng-Kai Hsu 4, Li-Fang Wang 1,*
and Chien-Chih Chiu 2,4,6,7,8,*
1Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
mah.jadhao@yahoo.com (M.J.); dhanashrivdeshmukh1990@gmail.com (D.D.)
2Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan;
chunlinchen@mail.nsysu.edu.tw (C.-L.C.); uraderit@gmail.com (R.U.)
3Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University,
Kaohsiung 807, Taiwan
4Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
liuwangta@kmu.edu.tw (W.L.); wtliao@kmu.edu.tw (W.-T.L.); yifuc@kmu.edu.tw (J.Y.-F.C.);
b043100050@gmail.com (S.-K.H.)
5Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
tsaieing@kmu.edu.tw
6The Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University,
Kaohsiung 807, Taiwan
7Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
8Center for Cancer Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University,
Kaohsiung 807, Taiwan
*Correspondence: lfwang@kmu.edu.tw (L.-F.W.); cchiu@kmu.edu.tw (C.-C.C.);
Tel.: +886-67-312-1101 (ext. 2217) (L.-F.W.); +886-67-312-1101 (ext. 2368) (C.-C.C.);
Fax: +886-67-312-5339 (L.-F.W.)
Abstract:
Angiogenesis is the process of vascular network development and plays a crucial role in
cancer growth, progression, and metastasis. Phthalates are a class of environmental pollutants that
have detrimental effects on human health and are reported to increase cancer risk. However, the
interplay between phthalate exposure and angiogenesis has not been investigated thoroughly. In
this study, we investigated the effect of prolonged di (2-ethylhexyl) phthalate (DEHP) treatment
on the angiogenic potential of triple-negative breast cancer. MDA-MB-231 cells were exposed to
physiological concentrations of DEHP for more than three months. Prolonged DEHP exposure
induced angiogenesis in breast cancer cells. Endoglin (ENG)/CD105 is a membrane glycoprotein and
an auxiliary receptor of the TGF
β
receptor complex. In endothelial cells, ENG is highly expressed and
it is a prerequisite for developmental angiogenesis. A literature review highlights endoglin as a well-
known mesenchymal stem cell marker responsible for vascular development and angiogenesis. NGS
analysis showed that endoglin overexpression in DEHP-exposed MDA-MB-231 cells correlated with
tumor development and growth. An
in vivo
zebrafish xenograft assay showed that VEGFA induced
sprouting of the subintestinal vein (SIV) in embryos injected with DEHP-exposed cells. Endoglin
knockdown reduced SIV sprouting and VEGFA expression in zebrafish embryos. An
in vitro
HUVEC
tube formation assay showed that endoglin depletion reversed DEHP-induced VEGF-mediated
HUVEC tube formation in coculture. DEHP-induced endoglin activated TGF
β
/SMAD3/VEGF and
MAPK/p38 signaling in MDA-MB-231 cells. A cytokine angiogenesis antibody array showed induced
expression of the inflammatory cytokines IL1
α
, IL1
β
, IL6, and IL8, along with GMCSF and VEGF.
Endoglin knockdown reversed DEHP-induced activation of the TGF
β
/SMAD3/VEGF signaling
axis, MAPK/p38 signaling, and cytokine regulation, limiting angiogenesis potential both
in vivo
and
in vitro
. Targeting endoglin might serve as a potential alternative treatment to control angiogenesis,
leading to metastasis and limiting cancer progression.
Biomedicines 2022,10, 417. https://doi.org/10.3390/biomedicines10020417 https://www.mdpi.com/journal/biomedicines
Biomedicines 2022,10, 417 2 of 21
Keywords: DEHP; angiogenesis; endoglin; VEGF; TGFβsignaling; inflammatory cytokines
1. Introduction
In the growth and progression of breast cancer, new blood vessel generation from pre-
existing vessels is pivotal and is known as neovascularization. An increase in angiogenic
cues and mutation at a genetic level are some of the factors that accompany the growing
tumor. In quiescent vessels, endothelial cells (ECs) line up by forming a lumen surrounded
by a thick layer of pericytes or vascular smooth muscle cells (VSMCs), supporting the
structural integrity of the vessel [
1
]. Angiogenic cues or ischemia increase endothelial
permeability, giving rise to matrix metalloproteins for extracellular matrix degradation
and relieving pericyte-EC contact [
2
]. This provides space and a chance for the adjacent
cells to influx fluids and/or macromolecules due to the absence of cell-to-cell contact.
Through coordinated activation, endothelial cells tend to proliferate and migrate toward
promigratory phenomena (such as VEGF) to reach their final destination, where they
undergo morphogenesis to form a lumen and branches [
3
]. Several factors, such as VEGF,
thrombin, and sphingosine 1 phosphate, control EC permeability and lead to reversible
loss of junctional integrity [
4
]. Angiogenesis provides oxygen, nutrients, and metabolites to
growing tumors simultaneously, removes waste products from them, and nourishes them
to form a solid mass. Angiogenesis is mostly controlled by proangiogenic factors such as
VEGF and its tyrosine kinase receptors VEGFR1 and VEGFR2. Activation of the VEGF
receptor by its ligand leads to activation of a number of downstream signaling pathways,
such as PI3K, MAPK, and PLCγ, which mostly occur during angiogenesis [5].
Cancer highjacks and diverts the flow of blood vessels toward it to fulfil its require-
ments, leading to enhanced proliferation and metastatic spread. The angiogenesis niche
is triggered by hypoxia, leading to the production of angiogenic factors and immunosup-
pressive cytokines. For instance, in neuroblastoma (NB), extracellular microenvironment
hypoxia is reported to promote extracellular adenosine generation, which induces VEGF
production by binding with A3 adenosine receptor and activating HIF-1
α
/2
α
/VEGF
axis [
6
]. In addition, a recent study conducted by Shen demonstrated that HIF-1
α
expres-
sion is parallel with VEGF expression, and both expressions are positively correlated with
poor prognosis of ovarian cancer [
7
]. In contrast, pituitary adenylate cyclase-activating
polypeptide (PACAP) and its receptor PAC1R suppress angiogenic pathway and mesenchy-
mal markers, such as vimentin and MMP-2 by inhibition of MAPK/PI3K/Akt signaling
in the hypoxia niche of glioblastoma [
8
,
9
]. Maugeri et al. indicated that caffeine exerts
anti-tumor effects on glioblastoma multiforme (GBM), characterized by extensive hypoxia
through significant downregulation of HIF-1
α
as well as VEGF [
10
]. Metformin, widely
prescribed for type II diabetes, is also suggested to circumvent hypoxia-mediated overex-
pression of pro-angiogenic factors (VEGF) and aberrant angiogenesis through promoting
perfusion [
11
]. In our previous study, a reduced ROS level was observed in MDA-MB-231
under prolonged exposure to DEHP [
12
]. Chen et al. revealed that hypoxia serves as a
double-edged sword in ROS production. On one hand, HIF decreases ROS production
via suppressing the tricarboxylic acid (TCA) cycle; on the other hand, it promotes ROS
formation by NADPH oxidase (NOX) because NOX is a downstream gene of HIF [
13
].
Collectively, there is an intimate interplay between hypoxic microenvironment, ROS, and
angiogenesis. Some of the angiogenic factors involved in tumor-induced angiogenesis
include vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF),
fibroblast growth factor (FGF), and cytokines such as interleukins (ILs), tumor necrosis
factors (TNFs), interferons (INFs), and tumor growth factors (TGFs) [
14
]. The process of
angiogenesis is governed by activators and inhibitors [
15
]. Determination and localization
of angiogenic activators and inhibitors can lead us to design a drug to reduce the metastatic
spread rate of the respective cancer.
Biomedicines 2022,10, 417 3 of 21
Endocrine disrupters (EDs)/endocrine disrupting chemicals (EDCs) are naturally
occurring or synthetic compounds that mimic endocrine hormonal activity to affect the
normal physiological functioning of the endocrine system [
16
]. EDs are often found in
many products, such as plastic, cosmetics and hygiene products, personal care products,
packaging, medical devices, heavy metals, and furniture [
17
]. One ED/EDC is phthalates
or phthalic acid, which are used to enhance the durability and flexibility of plastic products
and are reported to exert detrimental effects on human health [
18
]. Some of the studies
showed its ubiquitous presence in urine in industrialized countries. Its effects range
from an imbalance in the hormonal system to human sexual and reproductive system
development [
19
]. Frequently found phthalates include diethyl phthalate (DEP), dimethyl
phthalate (DMP), di (2-ethylhexyl) phthalate (DEHP), diisononyl phthalate (DINP), and
dibutyl phthalate (DBP) [
20
]. Among all phthalates, DEHP is the most widely used; it is
noncovalently bound in plastics and results in leaching under different conditions, resulting
in ubiquitous environmental distribution [
18
,
21
]. Phthalates may result in human exposure
in one of several ways, such as dermal contact, inhalation, and ingestion, which could
pose a risk to potential health conditions, such as cancer progression and disturbance of
reproductive and respiratory systems [
22
,
23
]. DEHP exposure in uterine leiomyoma cells
increased cell viability and anti-apoptotic proteins [
24
]. In lung cancer cells, it increased
cell viability, cell proliferation, and inflammatory proteins [
25
]. Inhibition of angiogenesis
reduced placental growth and development, followed by DEHP exposure, in pregnant
mice [
26
]. Angiogenic factors such as sFlt-1 and PIGF are affected by treatment with
DEHP, suggesting a strong correlation between angiogenesis and phthalates [
27
]. An
increase in cancer cell viability and suppression of angiogenesis adversely both affect
cancer development. An increase in cancer cell viability spares a chance to proliferate
and metastasize and increases the niche to develop a solid tumor. At the molecular level,
several signaling pathways correlated with cancer growth and differentiation following
phthalate exposure have been recorded. Zu et al. reported that phthalate exposure activated
ERK5/p38 signaling, which promoted the spread of prostate cancer [
28
]. Activation of
MAPK (ERK1/2) and JNK signaling following DBP treatment induced testicular and liver
damage in a murine model [
29
,
30
]. Similarly, in our previous study, we demonstrated the
involvement and activation of PI3K/Akt signaling in continuous and long-term exposure
to DEHP-induced multidrug resistance in breast cancer cells [12].
Endoglin (CD105/ENG) is a transmembrane glycoprotein and a coreceptor of the
tumor growth factor-
β
(TGF
β
) family. Endoglin is highly expressed in proliferating vascu-
lar endothelial cells [
31
,
32
], and it interacts with their serine/threonine kinase receptors
(TGF
β
RI or TGF
β
RII), forming a heterotrimer along with ligand [
33
35
]. Other ligands in-
clude TGF
β
, activins, GDFs, and BMPs [
36
]. Ligand binding leads to receptor dimerization
and autophosphorylation at serine residues, recruiting R-SMAD and Co-SMAD proteins to
the nucleus and transcribing target genes, leading to several biological processes, namely,
proliferation, migration, differentiation, cell death, and angiogenesis [
37
]. Significant evi-
dence has shown increased endoglin in the TGF-
β
signaling pathway in tumor-associated
endothelial cells, making it ‘a marker’ for tumor-induced angiogenesis [
38
]. Solid tumors
such as prostate, cervical, and breast cancer showed an increased level of endoglin in
the endothelium [
39
41
]. The expression of endoglin increases during wound healing,
inflammation, vasculogenesis, and angiogenesis [
42
,
43
]. Liu et al. showed that endoglin
is indispensable for VEGF-induced angiogenesis. Endoglin-deficient mouse embryos die
due to malformation of blood vessels [
44
]. Proliferation and angiogenesis are inhibited in
ovarian cancer endothelial cells following endoglin depletion/knockdown [
45
]. In murine
mammary carcinoma, endoglin silencing reduces the growth and number of vessels formed
during tumor progression [
46
]. Endoglin interacts with VEGFR2 in a VEGF-dependent
manner to sustain and stabilize VEGFRII on the cell surface to promote tip cell formation
for tumor progression and growth [
47
]. In the current study, we demonstrated the detailed
mechanism of endoglin-mediated regulation of TGF
β
, MAPK/p38 signaling, and cytokines
Biomedicines 2022,10, 417 4 of 21
controlling angiogenesis in prolonged DEHP-exposed MDA-MB-231 cells
in vitro
and
in vivo.
2. Materials and Method
2.1. Cell Culture
MDA-MB-231 cells, human breast cancer cells, and human umbilical vein endothelial
cells (HUVECs) were used as experimental models for the study. MDA-MB-231 cells were
acquired from the American Type Culture Collection (ATCC) and maintained in DMEM
(Gibco, Grand Island, NE, USA) supplemented with penicillin, streptomycin antibiotics,
10% FBS, 0.03% glutamine, and 1 mM sodium pyruvate in a 5% CO
2
incubator at 37
C in
humidified conditions. A sample of 293T cells were acquired from ATCC and maintained
in low antibiotic (0.1% penicillin/streptomycin) DMEM containing FBS (10%), glutamine
(0.03%), and sodium pyruvate (1 mM). Human umbilical vein endothelial cells (HUVECs)
were purchased from Lonza (CC2519, Basel, Switzerland) and maintained in EGM
TM
-
2 endothelial cell growth medium (CC-3121, Lonza, Basel, Switzerland) supplemented
with an EGM
TM
-2 supplement pack (CC-4122, Lonza, Basel, Switzerland) containing
bovine brain extract (BBE) and free of exogenous VEGF in a 5% CO2incubator at 37 C in
humidified conditions.
2.2. Reagents and Antibodies
DEHP (Sigma-Aldrich, 36735, Saint Louis, MO, USA) was reconstituted in DMSO
(Sigma-Aldrich, Saint Louis, MO, USA). Doxorubicin (Dox) (Sigma-Aldrich, D1515, Saint Louis,
MO, USA) was dissolved in DMSO (Sigma-Aldrich, Saint Louis, MO, USA). Primary an-
tibodies for proteins: Endoglin/CD105 (71 kDa, Proteintech, 10862-1-AP, Rosemont, IL,
USA), MAPK (ERK1/2) (44 kDa, Cell Signaling, 137F5, Danvers, MA, USA), phospho-
MAPK (ERK1/2) (Thr202/Tyr204) (44 kDa, Cell Signaling, 9101 s, Danvers, MA, USA), p38
(38 kDa, Proteintech, 14064-1-AP, Rosemont, IL, USA), phospho-p38 (Tyr182) (38 kDa, Santa
Cruz, SC-166182, Santa Cruz, CA, USA), GAPDH (35.5 kDa, EMD Millipore, MAB374,
Burlington, MA, USA), Smad2/3 (52, 60 kDa, Cell Signaling, 8685, Danvers, MA, USA),
phospho-Smad2 (Ser465/467)/Smad3 (Ser423/425) (52, 60 kDa, Cell Signaling, 8828, Dan-
vers, MA, USA),
β
actin (43 kDa, Santa Cruz, SC-47778, Santa Cruz, CA, USA), and VEGFA
(43 kDa, Proteintech, 66828-1-IG, Rosemont, IL, USA). Secondary antibodies: Goat anti-
rabbit IgG (Alexa Fluor 594, Invitrogen, A11012, Waltham, MA, USA) was used as the
secondary antibody.
2.3. DEHP Exposure and Stable Clone Establishment
MDA-MB-231 cells were exposed to DEHP at a low concentration (100 nM) for three
months in cell culture medium. DEHP stock was directly diluted in culture medium
to achieve a final concentration of 100 nM. Cells were cultured in 100 mm cell culture
dish supplemented with 10–12 mL of DEHP containing DEHP. Upon reaching 80–90%
confluence,
1
4
cells were passaged and continuous culture with DEHP treatment was
maintained for 3 months. After 3 months, DEHP treatment was terminated permanently,
and cells were treated with 10 nM Dox for 72 h. Colonies formed following dox treatment
were selected and maintained for further study. Untreated MDA-MB-231 cells are denoted
as the control (Ctrl), and DEHP-exposed cells are denoted as DEHP from here onward. Both
untreated and DEHP-treated cells were maintained and equally passaged (10–12 passages)
to reduce senescence effects.
2.4. Zebrafish Xenograft
Transgenic zebrafish strain Tg (fli1: EGFP) was raised and maintained at 28.5
C in the
Zebrafish Core Facility at KMU. Embryos were obtained by pairwise mating and incubating
them in 0.03% phenylthiourea (PTU) at 28.5
C in an incubator for 48 h. Forty-eight hpf
embryos were injected with Vybrant
®
DiI-stained control and DEHP-exposed MDA-MB-
231 cells using a microinjector setup. Following injection, embryos were maintained in
Biomedicines 2022,10, 417 5 of 21
an incubator at 28.5
C for 24 h. Embryos were further observed for SIV sprouting, and
images were captured under a fluorescence microscope (MZ10F, Leica, Singapore) using
Metaview software (version 7.8.0.0).
2.5. RNA Sequencing
To evaluate the underlying mechanism of DEHP-induced angiogenesis in MDA-MB-
231 cells, next-generation sequencing (NGS) was performed on the control and DEHP-
exposed MDA-MB-231 cells as described in our previous publication [
12
]. Briefly, 3
µ
g of
isolated RNA from Biotools Co. Ltd. (Taipei, Taiwan) was used for sequencing. RNA was
sequenced, and data were analyzed by Illumina software. DEGs and GO were analyzed by
TopHat (v2.0.12) and GoSeq & topGO (2.12); KEGG analysis was performed by KOBAS
(v2.0). For data confirmation and validation, the log ratio of expression obtained by NGS
was further evaluated by QIAGEN Ingenuity Pathway Analysis (IPA
®
, QIAGEN Redwood,
Redwood City, CA, USA, Available online: www.qiagen.com/ingenuity (accessed on
21 December 2021).
2.6. Lentiviral Transfection
Envelop plasmid (pMD2. G), packaging plasmid (pCMV-dR8.91), and short hairpin
RNA (shRNA) containing hairpin-pLKO.1 vector was used for lentiviral particle prepa-
ration; pMD2. G, pCMV-dR8.91, scrambled/mock shRNA (clone ID: ASN0000000004)
and shENG (clone ID: TRCN0000083140) were purchased from the RNAi core facility
(Academia Sinica, Taipei, Taiwan). Scramble shRNA or sheng, along with pMD2. G and
pCMV-dR8.91, were transfected into 293T cells using Lipofectamine 2000 (Thermo Fisher,
11668019, Waltham, MA, USA) in OptiMEM for 18 h. Packaged lentiviruses were harvested
in FBS/BSA-enriched DMEM at 36 h and 48 h post-transfection. Collected lentiviruses
were concentrated using a 100 K molecular weight cutoff filter unit (MAP100C38, Pall
Corporation, New York, NY, USA). Lentiviral transfection with shScr (mock) or shENG con-
taining lentivirus was performed using Lipofectamine 2000 in the control and DEHP-treated
MDA-MB-231 cells for 24 h. Transfected cells were exposed to puromycin (
1–2 µg/mL
) for
selection and establishment of stable knockdown cells.
2.7. Quantitative Polymerase Chain Reaction (qPCR)
Total RNA was extracted from 25–30 noninjected and tumor cell-injected embryos
using TRIzol reagent. Similarly, cellular RNA from the control and DEHP-exposed (mock-
treated and ENG knockdown) cells was isolated using TRIzol reagent. Extracted RNA was
reverse transcribed to cDNA, and qPCR was performed using SYBR Green master mix
(Applied BiosystemsTM, Waltham, MA, USA). cDNA from zebrafish embryos or cells and
zebrafish-specific primers against VEGFa (XM_009292018.3) (5
0
-CAGTTATTTCTCGCGGCTCT-
3
0
; 5
0
- TCCCCCTTCTTTGGGTATGT-3
0
) and GAPDH (NM_001115114.1) (5
0
-GTGGAGTCTA
CTGGTGTCTTC-3
0
and 5
0
-GTGCAGGAGGCATTGCTTACA-3
0
) and human-specific primers
against endoglin (NM_001278138.2) (5
0
-CGCCAACCACAACATGCAG-3
0
and 5
0
- GCTCCA
CGAAGGATGCCAC-3
0
); TGF
β
RII (NM_003242.6) (5
0
-GCTTTGCTGAGGTCTATAAGGC-
3
0
and 5
0
-GGTACTCCTGTAGGTTGCCCT-3
0
); SMAD3 (NM_001145103.2) (5
0
-GTCTGCAAG
ATCCCACCAG-3
0
and 5
0
-AGCCCTGGTTGACCGACT-3
0
);
β
-actin (NM_001101.5) (5
0
-
TGAGACCTTCAACACCCCAGCCAT-30and 50-CGTAGATGGGCACAGTGTGGGTG-30)
was purchased from Genomics (New Taipei, Taiwan) and analyzed on a QuantStudio
TM
5 Real-Time PCR System (A28574, Applied Biosystems
TM
, Waltham, MA, USA). The data
analysis was performed using QuantStudio
TM
5 Design & Analysis Software (version 1.5.1,
Applied Biosystems
TM
, Waltham, MA, USA), and relative mRNA expression was calculated
by the equation 2
∆∆Ct
. Data quantification was performed by SigmaPlot (version 12.3,
Systat Software, Inc., Erkrath, Germany).
Biomedicines 2022,10, 417 6 of 21
2.8. Western Blotting
Protein samples (30–40
µ
g) were separated by SDS–PAGE, followed by transfer to
PVDF membranes. PVDF membrane blocking was performed in 5% non-fat milk and
subsequent incubation with specific primary antibodies against ENG, MAPK (ERK1/2),
phospho-MAPK (ERK1/2), phospho-p38, and GAPDH at 4
C. The following day, mem-
branes were washed with blocking buffer to remove unbound antibodies, and host-specific
secondary antibody treatment was performed. Finally, membranes were washed, and
chemiluminescence signal detection was performed using an ECL
TM
detection kit and
luminescent image analyzer (Amersham Imager 680, GE Healthcare and Bioscience ab,
Princeton, NJ, USA).
2.9. In Vitro-Angiogenesis/Tube Formation Assay
In vitro
angiogenesis/tube formation was performed by coculturing HUVECs with
the control and DEHP-treated MDA-MB-231 cells. Briefly, 1
×
10
4
HUVECs were seeded
on Matrigel (Culturex
®
5X BME)-coated 24-well plates. A total of 1
×
10
5
of the control and
DEHP-exposed cells were seeded in 0.4
µ
m Transwell inserts (3413, Corning Incorporated,
Corning, NY, USA) and placed in HUVEC seeded plates. Cultures were incubated at 37
C
in an incubator, and images were captured 8 h after seeding. Tube formation data analysis
was performed by web-based WIMASIS Image Analysis software (Onimagin Technologies
SCA, Cordoba, Spain). Data quantification was performed by SigmaPlot.
2.10. Quantitative Enzyme-Linked Immunosorbent Assay (ELISA)
Approximately 2
×
10
5
of the control and DEHP-exposed (mock-treated and ENG
knockdown) MDA-MB-231 cells were seeded in 6-well plates and incubated overnight.
After incubation, serum-free culture medium was replaced, and cells were incubated for
48–72 h. Culture medium was collected and used to perform VEGF quantification using a
Human VEFG
165
Standard TBM ELISA Development kit (900-T10, PEPROTech, Hamburg,
Germany), as described previously [48].
2.11. Immunofluorescence
Approximately 2
×
10
4
of the control and DEHP-exposed (mock-treated and ENG
knockdown) MDA-MB-231 cells were seeded in cell culture chamber slides (30114, SPL
Lifesciences, Pocheon-si, Korea) and incubated overnight at 37
C in an incubator. Cells
were fixed with 4% paraformaldehyde (PFA), permeabilized with 0.1% Triton X-100, and
blocked with blocking buffer (3% BSA in PBS). Cells were incubated with primary anti-
body (SMAD3), washed with blocking buffer, and incubated with host-specific secondary
antibody (goat anti-rabbit IgG, Alexa Fluor 594), along with nuclear staining with DAPI.
Fluorescence imaging was performed on a fluorescence microspore (Olympus, 1X71, Shin-
juku, Japan). Data analysis and quantification were performed by SigmaPlot (version 12.3,
Systat Software, Inc., Erkrath, Germany).
2.12. Antibody Angiogenesis Array
An antibody angiogenesis microarray was performed using a Human Angiogene-
sis Antibody Array-membrane kit (ab193655, Abcam, MA, USA), according to the man-
ufacturer’s instructions. Briefly, control and DEHP-exposed (mock-treated and ENG
knockdown) MDA-MB-231 cells were lysed, and the protein concentration was estimated;
200–250 µg
of total protein was used. The protein-conjugated membranes provided in the
kit were blocked with blocking buffer for 30 min. After incubation, half of the blocking
buffer was removed and replaced with an equal volume of protein sample and then incu-
bated for 1.5–5 h at room temperature. Membranes were washed and conjugated with a
biotinylated antibody cocktail for 1.5–2 h at room temperature. Membranes were washed
and incubated with HRP-conjugated streptavidin for 2 h at room temperature. Finally,
membranes were incubated with detection buffer, and chemiluminescence signals were
detected by a luminescence imager analyzer (Amersham Imager 680). Quantification of
Biomedicines 2022,10, 417 7 of 21
signal intensity was performed by ImageJ (version 1.53, NIH, USA). Data quantification
was performed using SigmaPlot.
2.13. Statistical Analysis
Statistical analysis of data was performed by SigmaPlot. Standard one-way ANOVA
and Student’s t tests were used to compare different groups. Data presented is as a
±
standard deviation (SD). A pvalue was considered statistically significant if it was <0.05.
3. Results
3.1. Prolonged DEHP Exposure Enhances the Angiogenesis Potential of MDA-MB-231 Cells
In Vivo
To evaluate the effect of prolonged DEHP exposure at physiological concentrations,
a zebrafish xenograft assay was performed. Control and DEHP-treated MDA-MB-231
cells injected in 48 hpf Tg (fli1: EGFP) embryo sacs showed enhanced SIV sprouting in
DEHP-treated MDA-MB-231-injected embryos compared to control MDA-MB-231-injected
embryos (Figure 1A). On average, 55% and 12% of embryos (n= 50) showed SIV sprouting
in DEHP-treated and control MDA-MB-231-injected embryos, respectively (Figure 1B). A
43% difference in SIV sprouting is indicative of pro-angiogenic effects of DEHP exposure in
MDA-MB-231 cells.
Biomedicines 2022, 10, x FOR PEER REVIEW 7 of 23
detected by a luminescence imager analyzer (Amersham Imager 680). Quantification of
signal intensity was performed by ImageJ (version 1.53, NIH, USA). Data quantification
was performed using SigmaPlot.
2.13. Statistical Analysis
Statistical analysis of data was performed by SigmaPlot. Standard one-way ANOVA
and Student’s t tests were used to compare different groups. Data presented is as a ±stand-
ard deviation (SD). A p value was considered statistically significant if it was <0.05.
3. Results
3.1. Prolonged DEHP Exposure Enhances the Angiogenesis Potential of MDA-MB-231 Cells In
Vivo
To evaluate the effect of prolonged DEHP exposure at physiological concentrations,
a zebrafish xenograft assay was performed. Control and DEHP-treated MDA-MB-231
cells injected in 48 hpf Tg (fli1: EGFP) embryo sacs showed enhanced SIV sprouting in
DEHP-treated MDA-MB-231-injected embryos compared to control MDA-MB-231-in-
jected embryos (Figure 1A). On average, 55% and 12% of embryos (n = 50) showed SIV sprout-
ing in DEHP-treated and control MDA-MB-231-injected embryos, respectively (Figure 1B). A
43% difference in SIV sprouting is indicative of pro-angiogenic effects of DEHP exposure
in MDA-MB-231 cells.
Figure 1. Representative and quantitative results of the zebrafish xenograft angiogenesis assay. (A)
DEHP-exposed MDA-MB-231 cells enhance SIV sprouting in Tg (fli1: EGFP) zebrafish embryos at
24 hpi. Red fluorescence: DiI stained breast cancer cells; Green fluorescence: vascular network of
zebrafish embryo. Scale bar = 1000 µm. White arrow: Indicate site of SIV sprouting. (B) Quantitative
results of angiogenesis in zebrafish showing SIV sprouting (n = 50); ** p < 0.001.
Figure 1.
Representative and quantitative results of the zebrafish xenograft angiogenesis assay.
(
A
) DEHP-exposed MDA-MB-231 cells enhance SIV sprouting in Tg (fli1: EGFP) zebrafish embryos at
24 hpi. Red fluorescence: DiI stained breast cancer cells; Green fluorescence: vascular network of
zebrafish embryo. Scale bar = 1000
µ
m. White arrow: Indicate site of SIV sprouting. (
B
) Quantitative
results of angiogenesis in zebrafish showing SIV sprouting (n= 50); ** p< 0.001.
3.2. Endoglin Predicted as a Regulator of DEHP-Induced Angiogenesis in Breast Cancer Cells
To identify the underlying mechanism and regulators of DEHP-induced angiogenesis
potential in MDA-MB-231 cells, RNA sequencing was performed. Gene ontology (GO)
enrichment analysis of differentially expressed genes (DEGs) was performed for control and
DEHP treated MDA-MB-231 cells. GO enrichment analysis showed the 30 most significantly
enriched terms related to biological process, cellular component, and molecular function.
Biomedicines 2022,10, 417 8 of 21
Regulation of cell growth, organ development, regulation of cell development, system
development, locomotion, response to wounding, and cell growth were identified among
the top 30 enriched GO terms (Figure 2A). GO terms such as cellular growth, organ
development, system development, and locomotion are closely related to angiogenesis
or vascular development, which is the initial phase of system or organ development.
During cancer progression, angiogenesis is a prerequisite for tumor growth and metastasis.
We further used IPA, a bioinformatics tool to identify and predict the profile of gene
expression and pathways underlying the impact of prolonged DEHP exposure to breast
cancer cells, and IPA analysis showed the significant enrichment of endothelial tissue
development and cell development under disease and function (Figure 2B). Downstream
effect analysis highlighted the involvement of endoglin with a log expression ratio of
4.457 (Figure 2C). Further disease and function analysis found enrichment of growth of
malignant tumors and growth of solid tumors (Figure 2D). Interestingly, endoglin was
found to be positively correlated with both the growth of malignant tumors and the growth
of solid tumors. To our surprise, the matrix metalloproteins MMP1 and MMP3, along
with the inflammatory cytokines IL1
β
and IL1
α
, were predicted to be activated with a
high log expression ratio (Figure 2E,F). The expression of endoglin was further confirmed
through Western blotting and qPCR. Prolonged DEHP exposure increased the expression
of endoglin almost 2-fold at protein and 3-fold at mRNA levels (Figure 2G,H). Collectively,
endoglin was predicted to be involved in endothelial tissue and cell development, along
with malignant and solid tumor growth, indicating a central role of endoglin. Considering
the pathophysiological conditions, angiogenesis/vascular development plays a crucial role
in normal tissue development or tumor growth [
49
,
50
]. An increase in endoglin expression
consistent with prediction of NGS data points towards its involvement in regulating DEHP-
induced angiogenesis, tumor growth, and ultimately metastasis.
3.3. Endoglin Depletion Reverses the DEHP-Induced Angiogenic Potential of MDA-MB-231 Cells
To confirm and validate the involvement of endoglin in DEHP-induced angiogenesis in
MDA-MB-231 cells, endoglin was knocked down using lentivirus-mediated shRNA trans-
fection. Western blotting showed upregulation of endoglin in scrambled/mock shRNA-
treated DEHP-exposed MDA-MB-231 cells. However, endoglin expression was completely
depleted in shENG-treated control and DEHP-exposed MDA-MB-231 cells (Figure 3A).
Next, control and DEHP-exposed MDA-MB-231 cells (mock- and shENG-treated) were
injected into the embryo sacs of 48 hpf Tg (fli1: EGFP) embryos. SIV sprouting evaluation at
24 h after injection showed that endoglin knockdown significantly reduced SIV sprouting
(Figure 3B). The number of embryos showing SIV sprouting following xenografting was
reduced from 50% to 12.5% in endoglin knockdown DEHP-exposed MDA-MB-231 cell-
injected mock-treated embryos compared with DEHP-exposed MDA-MB-231 cell-injected
embryos (Figure 3C). However, no change was observed in mock- and shENG-treated
control MDA-MB-231 cells. To understand the mechanism of cancer cell-induced SIV
sprouting of Tg (fli1: EGFP) embryos, mRNA levels of zebrafish VEGFA were investigated.
qPCR results showed that VEGFA mRNA levels were significantly increased in breast
cancer cell-injected embryos compared to embryos without injection. The highest VEGFA
mRNA levels were recorded in mock-treated DEHP-exposed MDA-MB-231 cell-injected
embryos, followed by embryos injected with mock-treated control MDA-MB-231 cells.
Interestingly, DEHP knockdown significantly reduced VEGFA mRNA levels in embryos
injected with shENG-treated DEHP-exposed MDA-MB-231 cells (Figure 3D). Surprisingly,
ENG knockdown in control cell-injected embryos showed no significant change in VEGFA
mRNA levels. Overall, endoglin controls and regulates DEHP-induced SIV sprouting and
VEGFA expression in Tg (fli1: EGFP) embryos injected with breast cancer cells.
Biomedicines 2022,10, 417 9 of 21
Biomedicines 2022, 10, x FOR PEER REVIEW 9 of 23
Figure 2. Representative results of RNA sequencing and IPA of control and DEHP-treated MDA-
MB-231 cells. (A) Bar chart of significantly enriched GO terms and the number of DEGs enriched in
Figure 2.
Representative results of RNA sequencing and IPA of control and DEHP-treated MDA-
MB-231 cells. (A) Bar chart of significantly enriched GO terms and the number of DEGs enriched in
biological processes (green), cellular components (orange), and molecular function (violet). (
B
) IPA-
derived heatmap analysis of development under cellular diseases and functions of the DEGs involved.
(
C
) Downstream analysis of genes involved in the development of endothelial tissue and cell devel-
opment. (
D
) IPA-derived heatmap analysis of cancer growth under cellular diseases and functions of
the DEGs involved. (
E
,
F
) Downstream analysis of genes involved in the growth of malignant tumors
and the growth of solid tumors in control and DEHP-exposed MDA-MB-231 cells, highlighting the
involvement of endoglin/CD105 and its prediction as a regulator of DEHP-induced angiogenesis.
(
G
,
H
) Prolonged DEHP exposure increased endoglin expression at the protein and mRNA level in
MDA-MB-231 cells; ** p< 0.001.
Biomedicines 2022,10, 417 10 of 21
Biomedicines 2022, 10, x FOR PEER REVIEW 11 of 23
Figure 3. Representative and quantitative results showing that DEHP-induced endoglin-controlled
VEGFA-mediated angiogenesis in vivo. (A) Prolonged DEHP treatment upregulated endoglin ex-
pression in MDA-MB-231 cells, and shENG treatment depleted endoglin expression in control and
DEHP-exposed MDA-MB-231 cells. (B) Endoglin knockdown reduced DEHP-induced SIV sprout-
ing in Tg (fli1: EGFP) zebrafish embryos at 24 hpi. Scale bar =1000 µ m. (C) Quantitative results of
angiogenesis showing a reduced number of zebrafish embryos showing SIV sprouting following
endoglin knockdown (n = 50). (D) Depletion of VEGFA mRNA levels in zebrafish embryos follow-
ing endoglin knockdown cell xenografts of DEHP-exposed MDA-MB-231 cells; ** p < 0.001.
3.4. Endoglin Regulates HUVEC Tube Formation through VEGF Production in Prolonged
DEHP-Treated MDA-MB-231 Cells
To validate the results of the in vivo zebrafish angiogenesis assay, we performed an
in vitro angiogenesis assay by coculturing HUVECs with control and DEHP-exposed
MDA-MB-231 cells (mock- and shENG-treated). Induced tube formation was observed
under all coculture conditions compared to HUVECs alone, and mock-treated DEHP-ex-
posed MDA-MB-231 cell coculture showed the highest tube formation potential (Figure 4A).
Endoglin knockdown in DEHP-exposed cells significantly reduced HUVEC tube
Figure 3.
Representative and quantitative results showing that DEHP-induced endoglin-controlled
VEGFA-mediated angiogenesis
in vivo
. (
A
) Prolonged DEHP treatment upregulated endoglin ex-
pression in MDA-MB-231 cells, and shENG treatment depleted endoglin expression in control and
DEHP-exposed MDA-MB-231 cells. (
B
) Endoglin knockdown reduced DEHP-induced SIV sprouting
in Tg (fli1: EGFP) zebrafish embryos at 24 hpi. Scale bar =1000
µ
m. (
C
) Quantitative results of
angiogenesis showing a reduced number of zebrafish embryos showing SIV sprouting following
endoglin knockdown (n= 50). (
D
) Depletion of VEGFA mRNA levels in zebrafish embryos following
endoglin knockdown cell xenografts of DEHP-exposed MDA-MB-231 cells; ** p< 0.001.
3.4. Endoglin Regulates HUVEC Tube Formation through VEGF Production in Prolonged
DEHP-Treated MDA-MB-231 Cells
To validate the results of the
in vivo
zebrafish angiogenesis assay, we performed an
in vitro
angiogenesis assay by coculturing HUVECs with control and DEHP-exposed MDA-
MB-231 cells (mock- and shENG-treated). Induced tube formation was observed under all
coculture conditions compared to HUVECs alone, and mock-treated DEHP-exposed MDA-
MB-231 cell coculture showed the highest tube formation potential (Figure 4A). Endoglin
knockdown in DEHP-exposed cells significantly reduced HUVEC tube formation, consis-
Biomedicines 2022,10, 417 11 of 21
tent with the
in vivo
results. The number of tubes, average tube length, and number of
nodes showed similar observations. The average number of tubes, average tube length, and
number of nodes formed were highest in mock-treated DEHP-exposed MDA-MB-231 cells,
and HUVECs without coculture were lowest among all groups. Endoglin knockdown
in DEHP-exposed MDA-MB-231 cells significantly reduced the average number of tubes,
average tube length, and number of nodes formed in coculture. Interestingly, endoglin
knockdown in control MDA-MB-231 cells showed a slight increase in the average number
of tubes and average tube length; however, it did not affect the number of nodes formed
in coculture (Figure 4B–D). To understand the mechanism of HUVEC tube formation
in coculture, quantitative ELISA was performed. Cell culture medium of control and
DEHP-exposed MDA-MB-231 cells (mock- and shENG-treated) was used to evaluate VEGF
concentrations. The results showed that mock-treated DEHP-exposed MDA-MB-231 cell
culture medium contained the highest VEGF concentration of 432 pg/
µ
L, followed by
320 pg/
µ
L in mock-treated control MDA-MB-231 cell culture medium. Endoglin knock-
down in DEHP-exposed MDA-MB-231 cells resulted in significant depletion of VEGF levels,
as low as 97 pg/
µ
L. However, endoglin knockdown did not affect VEGF levels in control
MDA-MB-231 cells (Figure 4E). Collectively, prolonged DEHP exposure induced HUVEC
tube formation in coculture through endoglin-mediated VEGF production.
3.5. Endoglin Maintains the TGFβ/SMAD3/VEGF Signaling Axis in Prolonged DEHP-Treated
MDA-MB-231 Cells
To understand the underlying mechanism of DEHP-induced angiogenesis potential
through VEGF expression, gene set enrichment analysis (GSEA) was performed. TGF
β
signaling was enriched and found to be positively correlated with DEHP-exposed MDA-
MB-231 cells, with an enrichment score of 0.28 compared to
0.67 of the control MDA-MB-
231 cells (Figure 5A). Individual DEGs of the TGF
β
gene set showed core enrichment in
DEHP-treated MDA-MB-231 cells (Figure 5B). Endoglin is a TGF
β
coreceptor, and endoglin
upregulation following DEHP treatment is postulated to stabilize and induce TGF
β
signal-
ing, supporting the results. mRNA levels of TGF
β
signaling markers TGF
β
RII and SMAD3,
along with endoglin and VEGF, were evaluated and showed that DEHP treatment increased
mRNA levels of endoglin, TGF
β
RII, SMAD3, and VEGF; however, endoglin knockdown
resulted in a significant decline in mRNA levels of endoglin, TGF
β
RII, SMAD3, and
VEGF (Figure 5C–F). qPCR results show that activation of canonical TGF
β
/SMAD3/VEGF
signaling regulated through endoglin in DEHP-exposed MDA-MB-231 cells, consistent
with GSEA results. To verify the transcriptional regulatory activity of SMAD3, IF was
performed, and consistent with prior observations, SMAD3 expression was highest in
DEHP-exposed MDA-MB-231 cells. SMAD3 was observed to be colocalized in the nucleus
of DEHP-exposed cells, indicating SMAD3 activation and nuclear translocation. Interest-
ingly, endoglin knockdown not only reduced the expression of SMAD3 but also depleted
nuclear translocation, limiting its activation and SMAD3-mediated transcriptional reg-
ulation of VEGF (Figure 5G–I). The effect of DEHP exposure and endoglin knockdown
was evaluated on the expression of total and phospho-SMAD3 and showed that DEHP
exposure upregulates total and phospho-SMAD3. Consistent with qPCR and IF findings,
endoglin knockdown downregulates expression of both total and phospho-SMAD3 in
DEHP-exposed MDA-MB-231 cells (Figure 5J). Overall, prolonged DEHP exposure induces
endoglin expression, which stabilizes TGFβRII and activates TGFβsignaling, resulting in
increased VEGF production and secretion and enhancement of the angiogenesis potential
of MDA-MB-231 cells.
Biomedicines 2022,10, 417 12 of 21
Biomedicines 2022, 10, x FOR PEER REVIEW 13 of 23
Figure 4. Representative and quantitative results of endoglin-mediated HUVEC tube formation. (A)
Induced HUVEC tube formation in coculture with control and DEHP-exposed MDA-MB-231 cells
at 8 h after seeding; endoglin knockdown reversed DEHP-induced HUVEC tube formation. Scale
bar = 250 µ m. (B) Quantitative evaluation of the number of tubes formed in coculture at 8 h after
cell seeding. (C) Quantitative evaluation of average tube length in coculture at 8 h after cell seeding.
(D) Quantitative evaluation of the number of nodes formed in coculture at 8 h after cell seeding. (E)
Results of quantitative ELISA for VEGF levels in the cell culture medium of control and DEHP-
exposed MDA-MB-231 cells (mock- and shENG-treated); ** p < 0.001.
Figure 4.
Representative and quantitative results of endoglin-mediated HUVEC tube formation.
(
A
) Induced HUVEC tube formation in coculture with control and DEHP-exposed MDA-MB-231
cells at 8 h after seeding; endoglin knockdown reversed DEHP-induced HUVEC tube formation.
Scale bar = 250
µ
m. (
B
) Quantitative evaluation of the number of tubes formed in coculture at 8 h
after cell seeding. (
C
) Quantitative evaluation of average tube length in coculture at 8 h after cell
seeding. (
D
) Quantitative evaluation of the number of nodes formed in coculture at 8 h after cell
seeding. (
E
) Results of quantitative ELISA for VEGF levels in the cell culture medium of control and
DEHP-exposed MDA-MB-231 cells (mock- and shENG-treated); ** p< 0.001.
Biomedicines 2022,10, 417 13 of 21
Biomedicines 2022, 10, x FOR PEER REVIEW 15 of 23
Figure 5. Representative and quantitative results of endoglin-mediated regulation of the
TGFβ/SMAD3/VEGF signaling axis. (A) GSEA of TGFβ signaling showing a positive correlation
with DEHP-treated MDA-MB-231 cells (enrichment score). (B) BluePink O gram showing core
Figure 5.
Representative and quantitative results of endoglin-mediated regulation of the
TGF
β
/SMAD3/VEGF signaling axis. (
A
) GSEA of TGF
β
signaling showing a positive correla-
tion with DEHP-treated MDA-MB-231 cells (enrichment score). (
B
) Blue–Pink O’ gram showing core
enrichment of individual genes in the TGF
β
signaling gene set (red: upregulation; blue: downregula-
tion). (
C
F
) qPCR analysis showing the mRNA expression of endoglin, TGF
β
RII, SMAD3, and VEGF
in control and DEHP-exposed MDA-MB-231 cells (mock- and shENG-treated). (
G
) IF results showing
expression changes and nuclear localization of p-SMAD3. Scale bar = 100
µ
m. (
H
,
I
) Quantitative
analysis of p-SMAD3 IF showing total and nuclear p-SMAD3 expression levels. (
J
) SMAD3 and
phospho-SMAD3 expression evaluated by Western blotting; ** p< 0.001.
Biomedicines 2022,10, 417 14 of 21
3.6. Endoglin-Mediated MAPK/p38 Signaling and Secretory Cytokine Production May Contribute
to DEHP-Induced Angiogenesis in MDA-MB-231 Cells
Western blotting was performed to evaluate the effect of prolonged DEHP treatment on
MDA-MB-231 cells and showed activation of MAPK/p38 signaling. Upregulation of MAPK,
phospho-MAPK, and phospho-p38 was observed in DEHP-treated cells compared to control
MDA-MB cells. Endoglin knockdown reduced the expression of MAPK, phospho-MAPK,
and phospho-p38 in DEHP-exposed MDA-MB-231 cells; however, endoglin knockdown in
control MDA-MB-231 cells upregulated all three proteins (Figure 6A). Overall, these results
indicate that endoglin can positively regulate MAPK/p38 signaling in DEHP-exposed cells
and that MAPK/p38 may not be involved in angiogenesis progression in control MDA-
MB-231 cells. Next, to understand the molecular mechanism of MAPK/p38 signaling,
we performed an angiogenesis antibody array to screen multiple candidate proteins that
might be involved in ENG-mediated angiogenesis in MDA-MB-231 cells. The results
showed six proteins involved in endoglin-mediated angiogenesis among the panel of
43 angiogenesis-associated markers evaluated. The inflammatory cytokines IL1
α
, IL1
β
,
IL6, IL8, GMCSF, and VEGF were highly expressed in DEHP-treated cells compared to
control MDA-MB-231 cells. Endoglin knockdown reversed the expression of all six markers
in both control and DEHP-exposed MDA-MB-231 cells (Figure 6B–H). Overall, it can be
predicted that endoglin-mediated MAPK/p38 signaling may regulate the expression of the
abovementioned inflammatory cytokines and that GMCSF contributes to DEHP-induced
angiogenesis potential in MDA-MB-231 cells. Moreover, in control MDA-MB-231 cells,
endoglin knockdown resulted in MAPK/p38 activation, but it reduced the expression of
cytokines that might not be interrelated with each other. Endoglin depletion still affected
the expression of secretory cytokines in control MDA-MB-231 cells, which may be regulated
by different signaling mechanisms, warranting further investigation.
Biomedicines 2022,10, 417 15 of 21
Biomedicines 2022, 10, x FOR PEER REVIEW 17 of 23
Figure 6. Representative and quantitative results of endoglin-mediated MAPK/p38 signaling and
cytokine regulation. (A) Western blotting results showing upregulation of MAPK, phospho-MAPK,
p38, and phospho-p38 in DEHP-treated MDA-MB-231 cells, and endoglin reversed protein expres-
sion. (B) Representative results of the antibody angiogenesis array showing endoglin-mediated ex-
pression of the inflammatory cytokines GMCSF and VEGF. (CH) Quantification of the expression
of IL1α, IL1β, IL6, IL8, GMCSF, and VEGF in control and DEHP-exposed MDA-MB-231 cells (mock-
and shENG-treated). +ve ctrl: positive control; ** p < 0.001.
Figure 6.
Representative and quantitative results of endoglin-mediated MAPK/p38 signaling and
cytokine regulation. (
A
) Western blotting results showing upregulation of MAPK, phospho-MAPK,
p38, and phospho-p38 in DEHP-treated MDA-MB-231 cells, and endoglin reversed protein expression.
(
B
) Representative results of the antibody angiogenesis array showing endoglin-mediated expression
of the inflammatory cytokines GMCSF and VEGF. (
C
H
) Quantification of the expression of IL1
α
,
IL1
β
, IL6, IL8, GMCSF, and VEGF in control and DEHP-exposed MDA-MB-231 cells (mock- and
shENG-treated). +ve ctrl: positive control; ** p< 0.001.
Biomedicines 2022,10, 417 16 of 21
4. Discussion
Di-2-ethylhexyl phthalate (DEHP) is widely used as a plasticizer in plastic products
and is ubiquitous in daily life [
51
]. In particular, Chinese populations are exposed to higher
phthalate concentrations than Western populations, leading to exposure from contami-
nated food and air [
52
]. A large amount of evidence suggests that DEHP exposure is
intimately correlated with several disorders, such as Alzheimer’s disease, male infertility,
and increased risk of breast cancer [
53
55
]. The effects of DEHP exposure
in vivo
has
been evaluated in several studies. Barkat et al. demonstrated that high oral DEHP doses
in pregnant mice resulted in infertile male offspring [
56
]. However, other groups found
prenatal DEHP exposure affected female reproduction in F1–F3 generations, leading to
reproductive aging [
57
]. These studies indicate that DEHP exposure exerts significant
effects on reproductive system
in vivo
; however, long-term effects with low concentrations
of DEHP have not been demonstrated. A strategic study of cancer progression in long-term
DEHP-exposed mice might provide an insight into physiological condition and the effects
of DEHP exposure on cancer progression. Moreover, this type of study might lack the
actual prolonged DEHP exposure to tumor cells, considering the proliferation rate of tumor
implants
in vivo
. To overcome this issue, we exposed breast cancer cells with physiological
DEHP concentration (100 nM) long term in an attempt to mimic real life conditions. In
addition, phthalate has been confirmed to promote cancer progression through induced
proliferation, drug resistance, and angiogenesis. Crobeddu et al. demonstrated that DEHP
and its primary metabolite mono (2-ethylhexyl) phthalate (MEHP) enhance the prolifer-
ation of human breast ductal carcinoma cells via upregulation of progesterone receptor
(PR) [55].
In our previous study, we showed that long-term DEHP exposure at a concentra-
tion can induce multidrug resistance through increased expression of ABC transporters
and reduced intracellular ROS generation [
12
]. Over the past few decades, most studies
have reported the role of DEHP in placental development [
26
,
58
]. However, the associ-
ation between DEHP and angiogenesis in cancer remains largely unknown. Tsai et al.
reported that benzyl butyl phthalate (BBP) can induce angiogenesis in hepatocellular car-
cinoma (HCC) through the aryl hydrocarbon receptor AhR/ERK/VEGF axis [
59
]. This
finding indicates that phthalate has the potential to promote angiogenesis. Consistent
with previous reports, our data showed that SIV sprouting was remarkably increased
in zebrafish embryos implanted with prolonged DEHP-exposed MDA-MB-231 cells. We
demonstrated the proangiogenic activity of DEHP treatment to mimic physiological DEHP
exposure for an extended period of time because most DEHP-related studies emphasize
high-concentration DEHP treatment for a short period and research on low-dose and
prolonged DEHP exposure–mediated tumor progression is relatively insufficient.
Endoglin, also known as CD105, is a transmembrane glycoprotein that serves as a
coreceptor for TGF-
β
receptors I and II and is important for angiogenesis [
60
]. Kasprzak
et al. revealed that endoglin plays a significant role in angiogenesis in HCC. It cannot only
promote the proliferation of liver sinusoidal endothelial cells (ECs) but also enhance the
resistance of ECs to apoptosis [
61
]. Endoglin is also reported to promote colorectal cancer
liver metastasis by cancer-associated fibroblast (CAF)-expressing endoglin and TGF-
β
signaling [
62
]. Consistent with these findings, our study showed that endoglin knockout
reduced SIV sprouting and VEGFa expression in zebrafish embryos. Moreover, endoglin
depletion seriously disrupted DEHP-induced VEGF-mediated HUVEC tube formation.
Collectively, DEHP-induced angiogenesis is mainly mediated by endoglin.
A literature review revealed that endoglin is the auxiliary receptor of the TGF-
β
recep-
tor that facilitates the initiation of the TGF-
β
/SMAD signaling pathway. Phosphorylated
SMAD serves as a transcription factor (TF) to promote the expression of the downstream
effector genes responsible for cell proliferation, migration, and angiogenesis [
63
]. SMAD3
functions as a TF once phosphorylated following TGF
β
signaling activation, which in-
duces the expression of its downstream transcripts, especially VEGFA [
64
,
65
]. Our results
indicated that prolonged DEHP exposure activated the TGF
β
/SMAD3/VEGF signaling
Biomedicines 2022,10, 417 17 of 21
axis. Moreover, DEHP treatment induced nuclear localization of SMAD3, indicating the
transcriptional regulator activity of SMAD3. Endoglin depletion reversed the activation of
TGFβsignaling and reduced the nuclear translocation of SMAD3.
Mitogen-activated protein kinase (MAPK) is an important signaling pathway involved
in several cellular events, such as cell differentiation, proliferation, inflammation and
apoptosis. There are three major components of the MAPK cascade, including c-Jun N-
terminal kinases (JNKs), p38, and extracellular signal-regulated kinases
(ERKs) [66,67].
Preliminary evidence indicates that VEGFA overexpression is triggered by both the PI3K
and MAPK/p38 signaling pathways, which further drives angiogenesis [
68
]. Our results
corroborated the activation of MAPK/p38 in DEHP-exposed MDA-MB-231 cells. Con-
versely, endoglin knockdown inhibited phosphorylation and activation. Taken together,
DEHP exposure can induce angiogenesis through ENG-controlled TGF-
β
/SMAD3/VEGF
and MAPK/p38 signaling.
Accumulating studies have suggested that MAPK/p38 is also involved in the produc-
tion of cytokines, including IL-1 and IL-6, and granulocyte-macrophage colony-stimulating
factor (GM-CSF) [
69
71
]. In proteome profiling, the relationship between inflammation
and angiogenesis was apparently observed; in addition, IL-1
β
and VEGF had considerable
overlaps on biological functions and signal transduction of pro-angiogenic effects, espe-
cially with both of them activating MAPK in HUVECs [
72
]. Another finding suggested that
IL-1
β
also plays a crucial role in invasiveness and metastasis. Angiogenesis is capable of
providing sufficient nutrients because metastasis requires abundant energy to complete [
73
].
Interleukin-6 (IL-6), a pleiotropic cytokine, has been suggested to induce angiogenesis in
mounting studies. IL-6 can contribute to not only the upregulation of VEGF but also in-
creased endothelial tip cell sprouting through the pSTAT3 and jagged-1/Notch-3 signaling
pathways, respectively [
74
]. Gopinathan et al. highlighted that IL-6 shares similarities with
VEGF on pro-angiogenic function; however, the difference lies in the fact that a vessel with
aberrant pericytes coverage was observed in IL-6-induced angiogenesis, but was absent
in the VEGF-stimulated one [
75
]. Our results indicated that prolonged DEHP exposure
induced the expression of the inflammatory cytokines IL1
α
, IL1
β
, IL6, IL8, and GM-CSF
through endoglin regulation in MDA-MB-231 cells.
5. Conclusions
In conclusion, our study confirmed that prolonged DEHP exposure can promote an-
giogenesis in TNBC via the ENG/TGF
β
/SMAD3/VEGF signaling axis
in vitro
and
in vivo
.
Alternatively, long-term DEHP exposure can also induce upregulation of the inflammatory
cytokines IL1
α
, IL1
β
, IL6, and GM-CSF through the MAPK/p38 signaling pathway to
facilitate breast cancer progression. Hence, targeting ENG and its downstream signaling
pathway might be a promising strategy to normalize vasculature or curb angiogenesis and
subsequently inhibit the progression of breast cancer (Figure 7).
Biomedicines 2022,10, 417 18 of 21
Figure 7.
Schematic representation of prolonged DEHP exposure-induced angiogenesis potential
in breast cancer cells. Prolonged DEHP exposure at physiological concentrations upregulate the
expression of endoglin (ENG). Endoglin overexpression activates TGF
β
and MAPK/p38 signaling-
mediated production of VEGF; inflammatory cytokines IL1
α
, IL1
β
, IL6, IL8; and GMCSF, contributing
to enhanced DEHP-induced angiogenesis potential in breast cancer cells. shRNA mediated ENG
knockdown reversed ENG induced angiogenesis through downregulation of TGF
β
and MAPK/p38
signaling along with reduced expression of inflammatory cytokines IL1
α
, IL1
β
, IL6, IL8; and GMCSF.
Author Contributions:
Conceptualization, M.J., C.-C.C., E.-M.T. and L.-F.W.; data curation, M.J.,
D.D. and R.U.; formal analysis, M.J., D.D., W.-T.L. and J.Y.-F.C.; funding acquisition, C.-L.C., L.-F.W.
and C.-C.C.; investigation, M.J., J.Y.-F.C. and C.-C.C.; methodology, M.J., W.L., D.D., W.-T.L., R.U.
and S.-K.H.; project administration, L.-F.W. and C.-C.C.; resources, C.-L.C., W.L., W.-T.L., J.Y.-F.C.,
E.-M.T., L.-F.W. and C.-C.C.; software, W.L.; supervision, L.-F.W. and C.-C.C.; validation, M.J. and
D.D.; writing—original draft, M.J., R.U. and S.-K.H.; writing—review and editing, C.-C.C. All authors
have read and agreed to the published version of the manuscript.
Funding:
We thank the following institutions for providing financial support: Ministry of Science
and Technology, Taiwan (grants MOST 107-2320-B-110-002-MY3, 109-2320-B-037-017-MY3, 109-2314-
B-037-069-MY3 and 110-2320-B- 110-004); NSYSU-KMU joint grants (grant NSYSUKMU 110-I006,
111-P25 and 111-P12), the Kaohsiung Medical University Research Center, Taiwan (KMU-TC109A04),
and the Kaohsiung Medical University, Taiwan (KMU-DK(A)111001).
Institutional Review Board Statement:
All animal experiments were conducted according to the
guidelines of the Institutional Animal Care and Use Committee (IACUC), Kaohsiung Medical Uni-
versity, Kaohsiung, Taiwan. KMU, IACUC Approval No: 108182 Date of approval:
27 February 2020.
Informed Consent Statement: Not applicable.
Data Availability Statement:
The authors confirm that the data supporting the findings of this study
are available within the article and will be provided on request.
Acknowledgments:
We are grateful to the Center for Research Resources and Development (Kaohsi-
ung Medical University, Kaohsiung, Taiwan) for instrument support (real-time PCR and fluorescence
microscopy) and for providing access to Ingenuity Pathway Analysis Software (IPA
®
, QIAGEN
Redwood, Redwood City, CA, USA, www.qiagen.com/ingenuity accessed on 21 December 2021).
Biomedicines 2022,10, 417 19 of 21
Conflicts of Interest: The authors report no conflict of interest.
References
1. Carmeliet, P.; Jain, R.K. Molecular mechanisms and clinical applications of angiogenesis. Nature 2011,473, 298–307. [CrossRef]
2.
Ghajar, C.M.; George, S.C.; Putnam, A.J. Matrix metalloproteinase control of capillary morphogenesis. Crit. Rev. Eukaryot.
Gene Expr. 2008,18, 251–278. [CrossRef] [PubMed]
3.
Bryan, B.A.; D’amore, P.A. What tangled webs they weave: Rho-GTPase control of angiogenesis. Cell. Mol. Life Sci.
2007
,64,
2053–2065. [CrossRef] [PubMed]
4.
Sukriti, S.; Tauseef, M.; Yazbeck, P.; Mehta, D. Mechanisms Regulating Endothelial Permeability. Pulm. Circ.
2014
,4, 535–551.
[CrossRef] [PubMed]
5. Ferrara, N.; Gerber, H.-P.; LeCouter, J. The biology of VEGF and its receptors. Nat. Med. 2003,9, 669–676. [CrossRef]
6.
Maugeri, G.; D’Amico, A.G.; Federico, C.; Saccone, S.; Giunta, S.; Cavallaro, S.; D’Agata, V. Involvement of A3 Adenosine
Receptor in Neuroblastoma Progression via Modulation of the Hypoxic/Angiogenic Pathway. J. Mol. Neurosci.
2019
,69, 166–176.
[CrossRef]
7.
Shen, W.; Li, H.-L.; Liu, L.-Z.; Cheng, J.-X. Expression levels of PTEN, HIF-1
α
, and VEGF as prognostic factors in ovarian cancer.
Eur. Rev. Med. Pharmacol. Sci. 2017,21, 2596–2603.
8.
Maugeri, G.; D’Amico, A.; Saccone, S.; Federico, C.; Rasà, D.; Caltabiano, R.; Broggi, G.; Giunta, S.; Musumeci, G.; D’Agata, V.
Effect of PACAP on Hypoxia-Induced Angiogenesis and Epithelial–Mesenchymal Transition in Glioblastoma. Biomedicines
2021
,
9, 965. [CrossRef]
9.
D’Amico, A.G.; D’Amico, A.G.; Maugeri, G.; Vanella, L.; Pittalà, V.; Reglodi, D.; D’Agata, V. Multimodal Role of PACAP in
Glioblastoma. Brain Sci. 2021,11, 994. [CrossRef]
10.
Maugeri, G.; D’amico, A.G.; Rasà, D.M.; Saccone, S.; Federico, C.; Magro, G.; Cavallaro, S.; D’agata, V. Caffeine Effect on
HIFs/VEGF Pathway in Human Glioblastoma Cells Exposed to Hypoxia. Anti-Cancer Agents Med. Chem.
2018
,18, 1432–1439.
[CrossRef]
11.
Wang, J.-C.; Li, G.-Y.; Li, P.-P.; Sun, X.; Li, W.-M.; Li, Y.; Lu, S.-Y.; Liu, P.-J. Suppression of hypoxia-induced excessive angiogenesis
by metformin via elevating tumor blood perfusion. Oncotarget 2017,8, 73892–73904. [CrossRef]
12.
Jadhao, M.; Tsai, E.-M.; Yang, H.-C.; Chen, Y.-F.; Liang, S.-S.; Wang, T.-N.; Teng, Y.-N.; Huang, H.-W.; Wang, L.-F.; Chiu, C.-C. The
Long-Term DEHP Exposure Confers Multidrug Resistance of Triple-Negative Breast Cancer Cells through ABC Transporters and
Intracellular ROS. Antioxidants 2021,10, 949. [CrossRef] [PubMed]
13.
Chen, R.; Lai, U.H.; Zhu, L.; Singh, A.; Ahmed, M.; Forsyth, N.R. Reactive Oxygen Species Formation in the Brain at Different
Oxygen Levels: The Role of Hypoxia Inducible Factors. Front. Cell Dev. Biol. 2018,6, 132. [CrossRef] [PubMed]
14.
Chulpanova, D.S.; Kitaeva, K.V.; Green, A.R.; Rizvanov, A.A.; Solovyeva, V.V. Molecular Aspects and Future Perspectives of
Cytokine-Based Anti-cancer Immunotherapy. Front. Cell Dev. Biol. 2020,8, 402. [CrossRef]
15.
Nishida, N.; Yano, H.; Nishida, T.; Kamura, T.; Kojiro, M. Angiogenesis in cancer. Vasc. Health Risk Manag.
2006
,2, 213–219.
[CrossRef]
16.
Gore, A.C.; Chappell, V.A.; Fenton, S.E.; Flaws, J.A.; Nadal, A.; Prins, G.S.; Toppari, J.; Zoeller, R.T. EDC-2: The Endocrine
Society’s Second Scientific Statement on Endocrine-Disrupting Chemicals. Endocr. Rev. 2015,36, E1–E150. [PubMed]
17.
Teitelbaum, S.L.; Belpoggi, F.; Reinlib, L. Advancing research on endocrine disrupting chemicals in breast cancer: Expert panel
recommendations. Reprod. Toxicol. 2015,54, 141–147. [CrossRef]
18. Wang, Y.; Qian, H. Phthalates and Their Impacts on Human Health. Healthcare 2021,9, 603. [CrossRef]
19.
Wormuth, M.; Scheringer, M.; Vollenweider, M.; Hungerbühler, K. What are the sources of exposure to eight frequently used
phthalic acid esters in Europeans? Risk Anal. 2006,26, 803–824. [CrossRef]
20.
Panagiotou, E.M.; Ojasalo, V.; Damdimopoulou, P. Phthalates, ovarian function and fertility in adulthood. Best Pract. Res. Clin.
Endocrinol. Metab. 2021,35, 101552. [CrossRef]
21.
Tanay Das, M.; Kumar, M.; Thakur, I.S. Differential toxicological endpoints of di(2-ethylhexyl) phthalate (DEHP) exposure in
MCF-7 and MDA-MB-231 cell lines: Possible estrogen receptor alpha (ERalpha) independent modulations. Indian J. Exp. Biol.
2014,52, 1052–1061. [PubMed]
22.
Rowdhwal, S.S.S.; Chen, J. Toxic Effects of Di-2-ethylhexyl Phthalate: An Overview. BioMed Res. Int.
2018
,2018, 1–10. [CrossRef]
[PubMed]
23.
Zarean, M.; Keikha, M.; Poursafa, P.; Khalighinejad, P.; Amin, M.; Kelishadi, R. A systematic review on the adverse health effects
of di-2-ethylhexyl phthalate. Environ. Sci. Pollut. Res. 2016,23, 24642–24693. [CrossRef] [PubMed]
24.
Kim, J.H. Analysis of the
in vitro
effects of di-(2-ethylhexyl) phthalate exposure on human uterine leiomyoma cells.
Exp. Ther. Med.
2018,15, 4972–4978. [CrossRef]
25.
Kim, J.H. Di(2-ethylhexyl) phthalate promotes lung cancer cell line A549 progression via Wnt/
β
-catenin signaling. J. Toxicol. Sci.
2019,44, 237–244. [CrossRef]
26.
Zhang, L.; Zhang, T.-L.; Zong, T.; Chen, Y.-L.; Ren, M.; Yu, X.-C.; Kuang, H.-B. Effect of di-(2-ethylhexyl) phthalate exposure on
placental development in pregnant mice. J. South. Med. Univ. 2016,36, 467–471.
27.
Ferguson, K.K.; McElrath, T.F.; Cantonwine, D.E.; Mukherjee, B.; Meeker, J.D. Phthalate metabolites and bisphenol-A in association
with circulating angiogenic biomarkers across pregnancy. Placenta 2015,36, 699–703. [CrossRef]
Biomedicines 2022,10, 417 20 of 21
28.
Zhu, M.; Huang, C.; Ma, X.; Wu, R.; Zhu, W.; Li, X.; Liang, Z.; Deng, F.; Wu, J.; Geng, S.; et al. Phthalates promote prostate cancer
cell proliferation through activation of ERK5 and p38. Environ. Toxicol. Pharmacol. 2018,63, 29–33. [CrossRef]
29.
Bhattacharya, N.; Dufour, J.M.; Vo, M.-N.; Okita, J.; Okita, R.; Kim, K.H. Differential Effects of Phthalates on the Testis and the
Liver1. Biol. Reprod. 2005,72, 745–754. [CrossRef]
30.
Wang, H.; Zhou, W.; Zhang, J.; Li, H. Role of JNK and ERK1/2 MAPK signaling pathway in testicular injury of rats induced by
di-N-butyl-phthalate (DBP). Biol. Res. 2019,52, 41. [CrossRef]
31.
Gougos, A.; Letarte, M. Primary structure of endoglin, an RGD-containing glycoprotein of human endothelial cells. J. Biol. Chem.
1990,265, 8361–8364. [CrossRef]
32.
Miller, D.W.; Graulich, W.; Karges, B.; Stahl, S.; Ernst, M.; Ramaswamy, A.; Sedlacek, H.H.; Müller, R.; Adamkiewicz, J. Elevated
expression of endoglin, a component of the TGF-beta-receptor complex, correlates with proliferation of tumor endothelial cells.
Int. J. Cancer 1999,81, 568–572. [CrossRef]
33.
Cheifetz, S.; Bellón, T.; Calés, C.; Vera, S.; Bernabeu, C.; Massagué, J.; Letarte, M. Endoglin is a component of the transforming
growth factor-beta receptor system in human endothelial cells. J. Biol. Chem. 1992,267, 19027–19030. [CrossRef]
34.
Koleva, R.I.; Conley, B.A.; Romero, D.; Riley, K.S.; Marto, J.A.; Lux, A.; Vary, C.P. Endoglin structure and function: Determinants
of endoglin phosphorylation by transforming growth factor-beta receptors. J. Biol. Chem. 2006,281, 25110–25123. [CrossRef]
35.
Liu, Z.; Lebrin, F.; Maring, J.A.; Driesche, S.V.D.; Van Der Brink, S.; van Dinther, M.; Thorikay, M.; Martin, S.; Kobayashi, K.;
Hawinkels, L.J.A.C.; et al. ENDOGLIN Is Dispensable for Vasculogenesis, but Required for Vascular Endothelial Growth
Factor-Induced Angiogenesis. PLoS ONE 2014,9, e86273. [CrossRef]
36.
Barbara, N.P.; Wrana, J.L.; Letarte, M. Endoglin is an accessory protein that interacts with the signaling receptor complex of
multiple members of the transforming growth factor-beta superfamily. J. Biol. Chem. 1999,274, 584–594. [CrossRef] [PubMed]
37.
Hata, A.; Chen, Y.G. TGF-beta Signaling from Receptors to Smads. Cold Spring Harb. Perspect. Biol.
2016
,8, a022061. [CrossRef]
[PubMed]
38. Nassiri, F.; Cusimano, M.D.; Scheithauer, B.W.; Rotondo, F.; Fazio, A.; Yousef, G.M.; Syro, L.V.; Kovacs, K.; Lloyd, R.V. Endoglin
(CD105): A review of its role in angiogenesis and tumor diagnosis, progression and therapy. Anticancer Res.
2011
,31, 2283–2290.
[PubMed]
39.
Beresford, M.; Harris, A.L.; Ah-See, M.; Daley, F.; Padhani, A.R.; Makris, A. The relationship of the neo-angiogenic marker,
endoglin, with response to neoadjuvant chemotherapy in breast cancer. Br. J. Cancer 2006,95, 1683–1688. [CrossRef] [PubMed]
40.
Brewer, C.A.; Setterdahl, J.J.; Li, M.J.; Johnston, J.M.; Mann, J.L.; McAsey, M.E. Endoglin expression as a measure of microvessel
density in cervical cancer. Obstet. Gynecol. 2000,96, 224–228.
41. Wikström, P.; Lissbrant, I.F.; Stattin, P.; Egevad, L.; Bergh, A. Endoglin (CD105) is expressed on immature blood vessels and is a
marker for survival in prostate cancer. Prostate 2002,51, 268–275. [CrossRef] [PubMed]
42.
Jonker, L.; Arthur, H.M. Endoglin expression in early development is associated with vasculogenesis and angiogenesis. Mech. Dev.
2002,110, 193–196. [CrossRef]
43.
Torsney, E.; Charlton, R.; Parums, D.; Collis, M.; Arthur, H.M. Inducible expression of human endoglin during inflammation and
wound healing in vivo. Agents Actions 2002,51, 464–470. [CrossRef]
44.
Li, D.Y.; Sorensen, L.K.; Brooke, B.S.; Urness, L.D.; Davis, E.C.; Taylor, D.G.; Boak, B.B.; Wendel, D.P. Defective Angiogenesis in
Mice Lacking Endoglin. Science 1999,284, 1534–1537. [CrossRef] [PubMed]
45.
Xu, Y.; Wang, D.; Zhao, L.-M.; Zhao, X.-L.; Shen, J.-J.; Xie, Y.; Cao, L.-L.; Chen, Z.-B.; Luo, Y.-M.; Bao, B.-H.; et al. Endoglin is
necessary for angiogenesis in human ovarian carcinoma-derived primary endothelial cells. Cancer Biol. Ther.
2013
,14, 937–948.
[CrossRef]
46.
Dolinsek, T.; Markelc, B.; Sersa, G.; Coer, A.; Stimac, M.; Lavrencak, J.; Brozic, A.; Kranjc, S.; Cemazar, M. Multiple Delivery of
siRNA against Endoglin into Murine Mammary Adenocarcinoma Prevents Angiogenesis and Delays Tumor Growth. PLoS ONE
2013,8, e58723. [CrossRef]
47.
Tian, H.; Huang, J.J.; Golzio, C.; Gao, X.; Hector-Greene, M.; Katsanis, N.; Blobe, G.C. Endoglin interacts with VEGFR2 to promote
angiogenesis. FASEB J. 2018,32, 2934–2949. [CrossRef]
48.
Nellinger, S.; Schmidt, I.; Heine, S.; Volz, A.; Kluger, P.J. Adipose stem cell-derived extracellular matrix represents a promising
biomaterial by inducing spontaneous formation of prevascular-like structures by mvECs. Biotechnol. Bioeng.
2020
,117, 3160–3172.
[CrossRef]
49. Folkman, J. Role of angiogenesis in tumor growth and metastasis. Semin. Oncol. 2002,29, 15–18. [CrossRef]
50.
Lugano, R.; Ramachandran, M.; Dimberg, A. Tumor angiogenesis: Causes, consequences, challenges and opportunities. Cell. Mol.
Life Sci. 2020,77, 1745–1770. [CrossRef]
51.
Sun, Y.; Shen, J.; Zeng, L.; Yang, D.; Shao, S.; Wang, J.; Wei, J.; Xiong, J.; Chen, J. Role of autophagy in di-2-ethylhexyl phthalate
(DEHP)-induced apoptosis in mouse Leydig cells. Environ. Pollut. 2018,243, 563–572. [CrossRef]
52.
Wang, W.; Leung, A.O.W.; Chu, L.H.; Wong, M.H. Phthalates contamination in China: Status, trends and human exposure-with
an emphasis on oral intake. Environ. Pollut. 2018,238, 771–782. [CrossRef] [PubMed]
53.
Yen, P.L.; How, C.M.; Liao, V.H.-C. Early-life and chronic exposure to di(2-ethylhexyl) phthalate enhances amyloid-beta toxicity
associated with an autophagy-related gene in Caenorhabditis elegans Alzheimer’s disease models. Chemosphere
2021
,273, 128594.
[CrossRef] [PubMed]
Biomedicines 2022,10, 417 21 of 21
54.
Khasin, L.G.; Della Rosa, J.; Petersen, N.; Moeller, J.; Kriegsfeld, L.J.; Lishko, P.V. The Impact of Di-2-Ethylhexyl Phthalate on
Sperm Fertility. Front. Cell Dev. Biol. 2020,8, 426. [CrossRef] [PubMed]
55.
Crobeddu, B.; Ferraris, E.; Kolasa, E.; Plante, I. Di(2-ethylhexyl) phthalate (DEHP) increases proliferation of epithelial breast
cancer cells through progesterone receptor dysregulation. Environ. Res. 2019,173, 165–173. [CrossRef] [PubMed]
56.
Barakat, R.; Lin, P.-C.P.; Rattan, S.; Brehm, E.; Canisso, I.F.; Abosalum, M.E.; Flaws, J.A.; Hess, R.; Ko, C. Prenatal Exposure to
DEHP Induces Premature Reproductive Senescence in Male Mice. Toxicol. Sci. 2017,156, 96–108. [CrossRef] [PubMed]
57.
Brehm, E.; Rattan, S.; Gao, L.; Flaws, J.A. Prenatal Exposure to Di(2-Ethylhexyl) Phthalate Causes Long-Term Transgenerational
Effects on Female Reproduction in Mice. Endocrinology 2017,159, 795–809. [CrossRef]
58.
Martínez-Razo, L.D.; Martínez-Ibarra, A.; Vázquez-Martínez, E.R.; Cerbón, M. The impact of Di-(2-ethylhexyl) Phthalate and
Mono(2-ethylhexyl) Phthalate in placental development, function, and pathophysiology. Environ. Int.
2020
,146, 106228.
[CrossRef]
59.
Tsai, C.F.; Hsieh, T.H.; Lee, J.N.; Hsu, C.Y.; Wang, Y.C.; Lai, F.J.; Kuo, K.K.; Wu, H.L.; Tsai, E.M.; Kuo, P.L. Benzyl butyl phthalate
induces migration, invasion, and angiogenesis of Huh7 hepatocellular carcinoma cells through nongenomic AhR/G-protein
signaling. BMC Cancer 2014,14, 556. [CrossRef]
60.
Rossi, E.; Bernabeu, C.; Smadja, D.M. Endoglin as an Adhesion Molecule in Mature and Progenitor Endothelial Cells: A Function
Beyond TGF-beta. Front. Med. 2019,6, 10. [CrossRef]
61.
Kasprzak, A.; Adamek, A. Role of Endoglin (CD105) in the Progression of Hepatocellular Carcinoma and Anti-Angiogenic
Therapy. Int. J. Mol. Sci. 2018,19, 3887. [CrossRef] [PubMed]
62.
Paauwe, M.; Schoonderwoerd, M.J.; Helderman, R.F.; Harryvan, T.J.; Groenewoud, A.; Van Pelt, G.W.; Bor, R.; Hemmer, D.M.;
Versteeg, H.H.; Snaar-Jagalska, B.E.; et al. Endoglin Expression on Cancer-Associated Fibroblasts Regulates Invasion and
Stimulates Colorectal Cancer Metastasis. Clin. Cancer Res. 2018,24, 6331–6344. [CrossRef] [PubMed]
63.
Perez-Gomez, E.; Del Castillo, G.; Santibáñez, J.F.; Lêpez-Novoa, J.M.; Bernabéu, C.; Quintanilla, M. The role of the TGF-beta
coreceptor endoglin in cancer. Sci. World J. 2010,10, 2367–2384. [CrossRef] [PubMed]
64. Frick, C.L.; Yarka, C.; Nunns, H.; Goentoro, L. Sensing relative signal in the Tgf-beta/Smad pathway. Proc. Natl. Acad. Sci. USA
2017,114, E2975–E2982. [CrossRef] [PubMed]
65.
Shi, X.; Guo, L.W.; Seedial, S.M.; Si, Y.; Wang, B.; Takayama, T.; Suwanabol, P.A.; Ghosh, S.; DiRenzo, D.; Liu, B.; et al. TGF-
beta/Smad3 inhibit vascular smooth muscle cell apoptosis through an autocrine signaling mechanism involving VEGF-A.
Cell Death Dis. 2014,5, e1317. [CrossRef]
66.
Chang, W.-T.; Bow, Y.-D.; Fu, P.-J.; Li, C.-Y.; Wu, C.-Y.; Chang, Y.-H.; Teng, Y.-N.; Li, R.-N.; Lu, M.-C.; Liu, Y.-C.; et al. A Marine
Terpenoid, Heteronemin, Induces Both the Apoptosis and Ferroptosis of Hepatocellular Carcinoma Cells and Involves the ROS
and MAPK Pathways. Oxid. Med. Cell. Longev. 2021,2021, 1–12. [CrossRef]
67.
Guo, Y.J.; Pan, W.W.; Liu, S.B.; Shen, Z.F.; Xu, Y.; Hu, L.L. ERK/MAPK signalling pathway and tumorigenesis. Exp. Ther. Med.
2020,19, 1997–2007. [CrossRef]
68.
Luangdilok, S.; Box, C.; Harrington, K.; Rh
ˆ
ys-Evans, P.; Eccles, S. MAPK and PI3K signalling differentially regulate angiogenic
and lymphangiogenic cytokine secretion in squamous cell carcinoma of the head and neck. Eur. J. Cancer
2011
,47, 520–529.
[CrossRef]
69.
Koga, Y.; Hisada, T.; Ishizuka, T.; Utsugi, M.; Ono, A.; Yatomi, M.; Kamide, Y.; Aoki-Saito, H.; Tsurumaki, H.; Dobashi, K.; et al.
CREB regulates TNF-
α
-induced GM-CSF secretion via p38 MAPK in human lung fibroblasts. Allergol. Int.
2016
,65, 406–413.
[CrossRef]
70.
Klemm, C.; Bruchhagen, C.; Van Krüchten, A.; Niemann, S.; Löffler, B.; Peters, G.; Ludwig, S.; Ehrhardt, C. Mitogen-activated
protein kinases (MAPKs) regulate IL-6 over-production during concomitant influenza virus and Staphylococcus aureus infection.
Sci. Rep. 2017,7, srep42473. [CrossRef]
71.
Sinfield, J.K.; Das, A.; O’Regan, D.J.; Ball, S.G.; Porter, K.E.; Turner, N.A. p38 MAPK alpha mediates cytokine-induced IL-6 and
MMP-3 expression in human cardiac fibroblasts. Biochem. Biophys. Res. Commun. 2013,430, 419–424. [CrossRef] [PubMed]
72.
Mohr, T.; Haudek-Prinz, V.; Slany, A.; Grillari, J.; Micksche, M.; Gerner, C. Proteome profiling in IL-1beta and VEGF-activated
human umbilical vein endothelial cells delineates the interlink between inflammation and angiogenesis. PLoS ONE
2017
,
12, e0179065. [CrossRef] [PubMed]
73.
Fahey, E.; Doyle, S.L. IL-1 Family Cytokine Regulation of Vascular Permeability and Angiogenesis. Front. Immunol.
2019
,10, 1426.
[CrossRef]
74.
Middleton, K.; Jones, J.; Lwin, Z.; Coward, J.I. Interleukin-6: An angiogenic target in solid tumours. Crit. Rev. Oncol. Hematol.
2014,89, 129–139. [CrossRef] [PubMed]
75.
Gopinathan, G.; Milagre, C.; Pearce, O.; Reynolds, L.E.; Hodivala-Dilke, K.; Leinster, D.A.; Zhong, H.; Hollingsworth, R.E.;
Thompson, R.G.; Whiteford, J.R.; et al. Interleukin-6 Stimulates Defective Angiogenesis. Cancer Res.
2015
,75, 3098–3107.
[CrossRef] [PubMed]
... minor modifications [25]. Briefly, approximately 3 × 10 4 cells were seeded in 12-well plates coated with a layer of growth factor-reduced Matrigel (BME001-05, R&D systems, Minneapolis, MN, USA). ...
... VEGFR-induced MAPK-p38 signaling induced HUVEC migration, and mechanistic depletion of p38 repressed VEGF-induced migration [65]. In our previous study, we demonstrated that prolonged DEHP exposure regulates TGFβ/SMAD3/VEGF and MAPK/p38 signaling-mediated angiogenesis in HUVECs [25]. Similarly, Ye et al. reported that MAPK/p38 signaling inhibition attenuates HUVEC and fibroblast angiogenic potential in coculture with tumor-conditioned medium [66]. ...
Article
Full-text available
The medicinal properties of natural/edible plant products and their use are popular in traditional practice owing to their nutritional contents with little to no side effects. Lepista nuda (L. nuda), an edible mushroom (Clitocybe nuda, commonly known as blewit), has attracted researchers to evaluate its contents and the mechanism of its activities. In the current study, we focused on evaluating the antiangiogenic effects of L. nuda water extract on zebrafish development and in vitro human umbilical vein endothelial cell (HUVEC) tube formation. Bioactive components such as ergothioneine, eritadenine, and adenosine were identified and quantified by HPLC analysis. The L. nuda extract showed antiangiogenic properties and inhibited intersegmental vessel (ISV), caudal vein plexus (CVP), hyaloid vessel (HV), and subintestinal vessel (SIV) development in Tg (fli1: EGFP) zebrafish embryos. The expression of angiogenesis-related genes (vegfaa, kdrl, vegfba, flt1, kdr) was affected following L. nuda extract treatment. L. nuda extract attenuated in vitro HUVEC tube formation, migration, and invasion. Furthermore, inhibition of MAPK/p38 signaling and depletion of proangiogenic genes, including growth factors (fgf, ang2, and vegfa); primary and accessory receptors (tie2, vegfr2, and eng); MMPs (mmp1 and mmp2); and cytokines (il-1α, il-1β, il-6, and tnf-α) was observed in HUVECs following L. nuda treatment. An in vivo zebrafish xenograft assay showed that L. nuda extract inhibited HuCCT1 cell-induced SIV sprouting in HuCCT1-injected embryos. Collectively, the results suggest that L. nuda could be a potential inhibitor of angiogenesis limiting cancer progression.
... In humans, the impact of phthalates on the cardiovascular system has been explored, and most of the studies have examined their effects on the fetoplacental vasculature (Lorigo and Cairrao, 2022). Phthalates may be involved in the initiation of the atherosclerotic process as they may contribute to endothelial dysfunction by inducing oxidative stress and apoptosis (Ban et al., 2014), and by disrupting the regulation of certain inflammatory cytokines (Jadhao et al., 2022). ...
Article
Full-text available
Phthalates may be associated with an increased risk of cardiometabolic diseases by interfering with glucose and lipid metabolism and by promoting adipogenesis. This study aimed to perform a systematic review and meta-analysis of the association between phthalate exposure and subclinical carotid atherosclerosis, using surrogate markers such as carotid intima-media thickness (IMT) and carotid plaques. The literature search was performed using four databases (Web of Science, Medline, PubMed, and Scopus), and this systematic review includes all available observational studies until July 6th, 2023. The Joanna Briggs Institute critical appraisal tool was used to assess the risk of bias. Meta-analyses were performed, and random effects models were used. Six high-quality cross-sectional studies and 2,570 participants aged 12 to 70 were included. Six phthalate metabolites showed significant associations with subclinical carotid atherosclerosis. Exposure to MBzP, ΣDEHP, and MnBP was associated with increased carotid IMT. Exposure to MEP was associated with a higher prevalence of carotid plaques, and MiBP was associated with a lower prevalence. Mixed results were observed for MMP in older adults. The meta-analyses showed a high degree of heterogeneity, and the results are based on single studies. This study accurately describes the evidence of this association to date, suggesting that phthalates are associated with increased carotid IMT and a higher prevalence of carotid plaques. Further research is needed to elucidate this association, as phthalates are still used in the manufacture of everyday products, humans continue to be exposed to them, and atherosclerosis is a public health concern.
... Smad proteins play crucial roles in a multitude of intricate biological processes, including cellular differentiation. These proteins exhibit a wide range of biological activities when interacting with various signaling proteins [9]. For instance, they possess the capability to modulate the response of specific genes to TGF-β by forming long-lasting protein-protein interactions with additional transcription factors [10]. ...
Article
Full-text available
Smad intranuclear binding protein 1 (SNIP1), a highly conserved nuclear protein, functions as a transcriptional regulator and exerts a significant influence on disease progression. In addition, the N-terminal domain of SNIP1 facilitates its interaction with Smad4, a signaling protein associated with the TGF-β family, and RelA/p65, a transcription factor connected to NF-κB. This interaction further enhances the transcriptional activation of c-Myc-dependent genes. Presently, the primary emphasis in research is directed towards targeting the catalytic domain of SNIP1, as it holds promise as a potential therapeutic target for various diseases. While the significance of SNIP1 in pathological mechanisms remains uncertain, this review aims to comprehensively examine the existing literature on the association between SNIP1 and proteins implicated in the regulation of diverse clinical conditions, including cancer, inflammation, and related diseases.
... CD105, a cell membrane glycoprotein, is the most important marker for MSCs [52]; however, CD105 acts as an accessory receptor for TGF-β [53][54][55], found overexpressed in actively proliferating endothelial cells, and it is considered as a powerful marker of new vessel formation. Together with CD34, CD105 can also be considered a marker for early stages of vascularization. ...
Article
Full-text available
Among all strategies directed at developing new tools to support re-vascularization of damaged tissues, the use of pro-angiogenic soluble factors, derived from mesenchymal stem cells (MSCs), appears a promising approach for regenerative medicine. Here, we compared the feasibility of two devices, generated by coupling soluble factors of human dental pulp mesenchymal stem cells (DPSCs), with a nanostructured scaffold, to support angiogenesis once transplanted in mice. DPSCs were obtained from impacted wisdom tooth removal, usually considered surgical waste material. After 28 days, we verified the presence of active blood vessels inside the scaffold through optical and scansion electron microscopy. The mRNA expression of surface antigens related to macrophage polarization (CD68, CD80, CD86, CD163, CD206), as well as pro-angiogenic markers (CD31, CD34, CD105, Angpt1, Angpt2, CDH5) was evaluated by real-time PCR. Our results demonstrate the capability of DPSC–scaffold and DPSC soluble factors–scaffold to support angiogenesis, similarly to adipose stem cells, whereas the absence of blood vessels was found in the scaffold grafted alone. Our results provide evidence that DPSC-conditioned medium can be proposed as a cell-free preparation able to support angiogenesis, thus, providing a relevant tool to overcome the issues and restrictions associated with the use of cells.
... In contrast, endoglin deficiency decreased production of VEGF in HPMCs. Obviously, just as previous studies have shown an interconnection between endoglin and the VEGF signaling pathway, we speculate that the same is true for peritoneal angiogenesis during PD (Li et al., 2015;Jadhao et al., 2022). Although the relationship in detail needs to further elucidate, it is sure that endoglin may also be a therapeutic target for the treatment of peritoneal angiogenesis and UF. ...
Article
Full-text available
Background: Peritoneal fibrosis (PF) is an intractable complication in patients on long-term peritoneal dialysis (PD). Transforming growth factor-β (TGF-β) is a key pro-fibrogenic factor involved in PD-associated PF, and endoglin, as a coreceptor for TGF-β, plays a role in balancing the TGF-β signaling pathway. Here, we investigated whether endoglin could be a potential therapeutic target for PF. Methods: In vivo, we established PF model in SD rats by daily intraperitoneal injection of peritoneal dialysis fluids (PDF) containing 4.25% glucose for 6 weeks and downregulated endoglin expression by tail vein injection of AAV9-ENG on day 14 to assess the effect of endoglin on peritoneal morphology and markers related to fibrosis, angiogenesis, and epithelial-mesenchymal transition (EMT). In vitro, we treated human peritoneal mesothelial cells (HPMCs) transfected with ENG siRNA in high glucose medium to explore the potential mechanism of endoglin in PF. Results: Compared to control group, continuous exposure to biologically incompatible PDF induced exacerbated PF, accompanied by a significant increase in endoglin expression. Conversely, knockdown of endoglin ameliorated peritoneal injury characterized by increased peritoneal thickening and collagen deposition, angiogenesis, as well as EMT. Consistently, HPMCs cultured in high glucose medium underwent the EMT process and exhibited over-expression of fibronectin, collagen type I, vascular endothelial growth factor (VEGF), whereas these aforementioned alterations were alleviated after ENG siRNA transfection. In addition, we also found that ENG siRNA inhibited TGF-β-induced phosphorylation of Smad2/3 and Smad1/5/9 in HPMCs treated with high glucose (HG). Conclusion: Our findings confirmed for the first time that endoglin exacerbated PF by regulating the activation of TGF-β/ALK/Smads signaling, which will provide a novel potential therapeutic target in PF.
Article
Breast cancer is the most common invasive malignancy among women worldwide and constitutes a complex and heterogeneous disease. Interest has recently grown in the role of the aryl hydrocarbon receptor (AhR) in breast cancer and the contribution of environment-polluting AhR agonists. Here, we present a literature review addressing AhR ligands, including pesticides hexachlorobenzene and chlorpyrifos, polycyclic aromatic hydrocarbons, polychlorinated dibenzo-p-dioxins and dibenzofurans , polychlorinated biphenyls, parabens, and phthalates. The objectives of this review are a) to summarize recent original experimental, preclinical, and clinical studies on the biological mechanisms of AhR agonists which interfere with the regulation of breast endocrine functions, and b) to examine the biological effects of AhR ligands and their impact on breast cancer development and progression. We discuss biological mechanisms of action in cell viability, cell cycle, proliferation, epigenetic changes, epithelial to mesenchymal transition, and cell migration and invasion. In addition, we examine the effects of AhR ligands on angiogenic processes, metastasis, chemoresistance, and stem cell renewal. We conclude that exposure to AhR agonists stimulates pathways that promote breast cancer development and may contribute to tumor progression. Given the massive use of industrial and agricultural chemicals, ongoing evaluation of their effects in laboratory assays and preclinical studies in breast cancer at environmentally relevant doses is deemed essential. Likewise, awareness should be raised in the population regarding the most harmful toxicants to eradicate or minimize their use.
Article
Diisobutyl adipate (DIBA), as a novel non-phthalate plasticizer, is widely used in various products. However, little effort has been made to investigate whether DIBA might have adverse effects on human health. In this study, we integrated an in silico and in vitro strategy to assess the impact of DIBA on cellular homeostasis. Since numerous plasticizers could activate peroxisome proliferator-activated receptor γ (PPARγ) pathway to interrupt metabolism systems, we first utilized molecular docking to analyze interaction between DIBA and PPARγ. Results indicated that DIBA had strong affinity with the ligand-binding domain of PPARγ (PPARγ-LBD) at Histidine 499. Afterwards, we used cellular models to investigate in vitro effects of DIBA. Results demonstrated that DIBA exposure increased intracellular lipid content in murine and human hepatocytes, and altered transcriptional expression of genes related to PPARγ signaling and lipid metabolism pathways. At last, target genes regulated by DIBA were predicted and enriched for Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. Protein-protein interaction (PPI) network and transcriptional factors (TFs)-genes network were established accordingly. Target genes were enriched in Phospholipase D signaling pathway, phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) and Epidermal growth factor receptor (EGFR) signaling pathway which were related to lipid metabolism. These findings suggested that DIBA exposure might disturb intracellular lipid metabolism homeostasis via targeting PPARγ. This study also demonstrated that this integrated in silico and in vitro methodology could be utilized as a high throughput, cost-saving and effective tool to assess the potential risk of various environmental chemicals on human health.
Article
Phthalates are one of the most widely used plasticizers in polymer products, and they are increasingly being exposed to people all over the world, generating health concerns. Phthalates are often used as excipients in controlled-release capsules and enteric coatings, and patients taking these drugs may be at risk. In both animals and human, phthalates are mainly responsible for testicular dysfunction, ovarian toxicity, reduction in steroidogenesis. In this regard, for a better understanding of the health concerns corresponding to phthalates and their metabolites, still more research is required. Significantly, multifarious forms of phthalates and their biomedical effects are need to be beneficial to investigate in the various tissues or organs. Based on these investigations, researchers can decipher their toxicity concerns and related mechanisms in the body after phthalate's exposure. This review summarizes the chemical interactions, mechanisms, and their biomedical applications of phthalates in animals and human.
Article
Full-text available
Pituitary adenylate cyclase-activating polypeptide (PACAP) exerts different effects in various human cancer. In glioblastoma (GBM), PACAP has been shown to interfere with the hypoxic micro-environment through the modulation of hypoxia-inducible factors via PI3K/AKT and MAPK/ERK pathways inhibition. Considering that hypoxic tumor micro-environment is strictly linked to angiogenesis and Epithelial–Mesenchymal transition (EMT), in the present study, we have investigated the ability of PACAP to regulate these events. Results have demonstrated that PACAP and its related receptor, PAC1R, are expressed in hypoxic area of human GBM colocalizing either in epithelial or mesenchymal cells. By using an in vitro model of GBM cells, we have observed that PACAP interferes with hypoxic/angiogenic pathway by reducing vascular-endothelial growth factor (VEGF) release and inhibiting formation of vessel-like structures in H5V endothelial cells cultured with GBM-conditioned medium. Moreover, PACAP treatment decreased the expression of mesenchymal markers such as vimentin, matrix metalloproteinase 2 (MMP-2) and matrix metalloproteinase 9 (MMP-9) as well as CD44 in GBM cells by affecting their invasiveness. In conclusion, our study provides new insights regarding the multimodal role of PACAP in GBM malignancy.
Article
Full-text available
Glioblastoma multiforme (GBM) is the deadliest form of brain tumors. To date, the GBM therapeutical approach consists of surgery, radiation-therapy and chemotherapy combined with molecules improving cancer responsiveness to treatments. In this review, we will present a brief overview of the GBM classification and pathogenesis, as well as the therapeutic approach currently used. Then, we will focus on the modulatory role exerted by pituitary adenylate cyclase-activating peptide, known as PACAP, on GBM malignancy. Specifically, we will describe PACAP ability to interfere with GBM cell proliferation, as well as the tumoral microenvironment. Considering its anti-oncogenic role in GBM, synthesis of PACAP agonist molecules may open new perspectives for combined therapy to existing gold standard treatment.
Article
Full-text available
The characteristics of phthalates had been thought to be similar to endocrine disruptors, which increases cancer risk. The role of phthalates in acquired drug resistance remains unclear. In this study, we investigated the effect of di-(2-ethylhexyl) phthalate (DEHP) on acquired drug resistance in breast cancer. MCF7 and MDA-MB-231 breast cancer cells were exposed to long-term physiological concentration of DEHP for more than three months. Long-exposure DEHP permanently attenuated the anti-proliferative effect of doxorubicin with estrogen receptor-independent activity even after withdrawal of DEHP. Long term DEHP exposure significantly reduced ROS (O2−) level in MDA-MB-231 cells while increased in MCF7 cells. ATP-binding cassette (ABC) transporters possess a widely recognized mechanism of drug resistance and are considered a target for drug therapy. Upregulation of ABC family proteins, ABCB-1 and ABCC-1 observed in DEHP-exposed clones compared to doxorubicin-resistant (DoxR) and parental MDA-MB-231 cells. A viability assay showed enhanced multidrug resistance in DEHP-exposed clones against Dox, topotecan, and irinotecan. Inhibition of ABC transporters with tariquidar, enhanced drug cytotoxicity through increased drug accumulation reversing acquired multidrug resistance in MDA-MB-231 breast cancer cells. Tariquidar enhanced Dox cytotoxicity by increasing intracellular ROS production leading to caspase-3 mediated apoptosis. Activation of PI3K/Akt signaling enhanced proliferation and growth of DEHP-exposed MDA-MB-231 cells. Overall, long-term DEHP exposure resulted in acquired multidrug resistance by upregulating ABCB-1 and ABCC1; apart from proliferation PI3K/Akt may be responsible for acquired drug resistance through ABC transporter upregulation. Targeting ABCB1 and ABCC1 with tariquidar may be a promising strategy for reversing the acquired multidrug resistance of triple-negative breast cancer cells.
Article
Full-text available
Phthalates are a family of high-production volume industrial chemicals used in the manufacture of plastics. Some phthalates are regulated as endocrine disrupting chemicals (EDCs) and reproductive toxicants based on adverse effects in the male. Potential effects in females are less understood although exposure levels can be higher in women compared to men. Here, we review the literature on the effects of phthalate exposures in adulthood on ovarian function and fertility in women. Experimental studies using cell cultures and rodents combined with human evidence from epidemiological studies suggest that phthalates pose a hazard to ovaries. Phthalates can disrupt follicle growth pattern, increase oxidative stress and cause follicle death. These effects could lead to infertility, faster depletion of ovarian reserve, and earlier reproductive senescence. However, more studies using more realistic exposure levels will be needed to properly assess the risks in women.
Article
Full-text available
Phthalates are a series of widely used chemicals that demonstrate to be endocrine disruptors and are detrimental to human health. Phthalates can be found in most products that have contact with plastics during producing, packaging, or delivering. Despite the short half-lives in tissues, chronic exposure to phthalates will adversely influence the endocrine system and functioning of multiple organs, which has negative long-term impacts on the success of pregnancy, child growth and development, and reproductive systems in both young children and adolescents. Several countries have established restrictions and regulations on some types of phthalates; however, we think that more countries should establish constraints or substitute measures for phthalates to reduce health risks. This article aims to summarize the adverse impacts of phthalates on human health, analyze the toxicity mechanism, assess the risks, and finally provide feasible strategies to reduce exposure of the public to phthalates.
Article
Full-text available
Hepatocellular carcinoma (HCC) is a leading cause of death, resulting in over 700 thousand deaths annually worldwide. Chemotherapy is the primary therapeutic strategy for patients with late-stage HCC. Heteronemin is a marine natural product isolated from Hippospongia sp. that has been found to protect against carcinogenesis in cholangiocarcinoma, prostate cancer, and acute myeloid leukemia. In this study, heteronemin was found to inhibit the proliferation of the HCC cell lines HA22T and HA59T and induce apoptosis via the caspase pathway. Heteronemin treatment also induced the formation of reactive oxygen species (ROS), which are associated with heteronemin-induced cell death, and to trigger ROS removal by mitochondrial SOD2 rather than cytosolic SOD1. The mitogen-activated protein kinase (MAPK) signaling pathway was associated with ROS-induced cell death, and heteronemin downregulated the expression of ERK, a MAPK that is associated with cell proliferation. Inhibitors of JNK and p38, which are MAPKs associated with apoptosis, restored heteronemin-induced cell death. In addition, heteronemin treatment reduced the expression of GPX4, a protein that inhibits ferroptosis, which is a novel form of nonapoptotic programmed cell death. Ferroptosis inhibitor treatment also restored heteronemin-induced cell death. Thus, with appropriate structural modification, heteronemin can act as a potent therapeutic against HCC.
Article
Full-text available
Di(2-ethylhexyl) phthalate (DEHP) is a chemical widely distributed in the environment as is extensively used in the plastic industry. DEHP is considered an endocrine disruptor chemical (EDC) and humans are inevitably and unintentionally exposed to this EDC through several sources including food, beverages, cosmetics, medical devices, among others. DEHP exposure has been associated and may be involved in the development of various pathologies; importantly, pregnant women are a particular risk group considering that endocrine alterations during gestation may impact fetal programming leading to the development of several chronic diseases in adulthood. Recent studies have indicated that exposure to DEHP and its metabolite Mono(2-ethylhexyl) phthalate (MEHP) may impair placental development and function, which in turn would have a negative impact on fetal growth. Studies performed in several trophoblastic and placental models have shown the negative impact of DEHP and MEHP in key processes related to placental development such as implantation, differentiation, invasion and angiogenesis. In addition, many alterations in placental functions like hormone signaling, metabolism, transfer of nutrients, immunomodulation and oxidative stress response have been reported. Moreover, clinical-epidemiological evidence supports the association between DEHP exposure and adverse pregnancy outcomes and pathologies. In this review, we aim to summarize for the first time current knowledge about the impact of DEHP and MEHP exposure on placental development and pathophysiology, as well as the mechanisms involved. We also remark the importance of exploring DEHP and MEHP effects in different trophoblast cell populations and discuss new perspectives regarding this topic.
Article
Full-text available
Tissue constructs of physiologically relevant scale require a vascular system to maintain cell viability. However, in vitro vascularization of engineered tissues is still a major challenge. Successful approaches are based on a feeder layer (FL) to support vascularization. Here, we investigated whether the supporting effect on the self‐assembled formation of prevascular‐like structures by microvascular endothelial cells (mvECs) originates from the FL itself or from its extracellular matrix (ECM). Therefore, we compared the influence of ECM, either derived from adipose‐derived stem cells (ASCs) or adipogenically differentiated ASCs, with the classical cell‐based FL. All cell‐derived ECM (cdECM) substrates enabled mvEC growth with high viability. Prevascular‐like structures were visualized by immunofluorescence staining of endothelial surface protein CD31 and could be observed on all cdECM and FL substrates but not on control substrate collagen I. On adipogenically differentiated ECM, longer and higher branched structures could be found compared with stem cell cdECM. An increased concentration of proangiogenic factors was found in cdECM substrates and FL approaches compared with controls. Finally, the expression of proteins associated with tube formation (E‐selectin and thrombomodulin) was confirmed. These results highlight cdECM as promising biomaterial for adipose tissue engineering by inducing the spontaneous formation of prevascular‐like structures by mvECs.
Article
Full-text available
A growing number of studies point to reduced fertility upon chronic exposure to endocrine-disrupting chemicals (EDCs) such as phthalates and plasticizers. These toxins are ubiquitous and are often found in food and beverage containers, medical devices, as well as in common household and personal care items. Animal studies with EDCs, such as phthalates and bisphenol A have shown a dose-dependent decrease in fertility and embryo toxicity upon chronic exposure. However, limited research has been conducted on the acute effects of these EDCs on male fertility. Here we used a murine model to test the acute effects of four ubiquitous environmental toxins: bisphenol A (BPA), di-2-ethylhexyl phthalate (DEHP), diethyl phthalate (DEP), and dimethyl phthalate (DMP) on sperm fertilizing ability and pre-implantation embryo development. The most potent of these toxins, di-2-ethylhexyl phthalate (DEHP), was further evaluated for its effect on sperm ion channel activity, capacitation status, acrosome reaction and generation of reactive oxygen species (ROS). DEHP demonstrated a profound hazardous effect on sperm fertility by producing an altered capacitation profile, impairing the acrosome reaction, and, interestingly, also increasing ROS production. These results indicate that in addition to its known chronic impact on reproductive potential, DEHP also imposes acute and profound damage to spermatozoa, and thus, represents a significant risk to male fertility.
Article
The widespread use of di(2-ethylhexyl) phthalate (DEHP) has resulted in its ubiquitous presence in the environment, which has led to serious health concerns. One of these concerns is its possible link to Alzheimer’s disease (AD), which is the most common neurodegenerative disease in aged individuals. This study investigated whether early-life and chronic exposure to DEHP affects AD via the toxicity of amyloid-β (Aβ), which has been implicated in the pathogenesis of AD, using Caenorhabditis elegans AD models (strains CL4176 and CL2006). We show that early-life DEHP exposure increased Aβ toxicity in C. elegans strains CL4176 and CL2006. Early-life and chronic exposure to DEHP also significantly increased intracellular ROS levels and Aβ deposition in aged CL2006 nematodes. Moreover, it was found that DEHP-induced Aβ toxicity does not require transcription factors DAF-16 or SKN-1, while early-life and chronic exposure to DEHP significantly increased the accumulation of lysosome-related organelles and the mRNA levels of the autophagy-related gene bec-1 in aged CL2006 nematodes. Our findings suggest that early-life and chronic exposure to DEHP enhances Aβ toxicity, which may be associated with the autophagy–lysosomal degradation pathway in C. elegans.