You are on page 1of 11

BIOENGINEERED

2022, VOL. 13, NO. 1, 206–216


https://doi.org/10.1080/21655979.2021.2012906

RESEARCH PAPER

C1q/tumor necrosis factor related protein 6 (CTRP6) regulates the phenotypes of


high glucose-induced gestational trophoblast cells via peroxisome proliferator-
activated receptor gamma (PPARγ) signaling
Jin Zhang and Wen-Pei Bai
Department of Obstetrics and Gynaecology, Beijing Shijitan Hospital, Capital Medical University, Beijing, P.R. China

ABSTRACT ARTICLE HISTORY


Multiple studies have confirmed that adipokines are compactly relevant to insulin resistance and Received 26 October 2021
participate in the pathogenesis of gestational diabetes mellitus (GDM). This paper aimed to study Revised 26 November 2021
the effects of C1q/tumor necrosis factor related protein (CTRP)6 on the phenotypes of trophoblast Accepted 27 November
2021
cells, covering cell proliferation, invasion and migration, and initially explore the mechanism. High
glucose was used to induce trophoblast cells to establish an in vitro model. The expression levels KEYWORDS
of CTRP6 were firstly determined, and then the effects of CTRP6 knockdown on cell viability, CTRP6; gestational
apoptosis, migration and invasion were assessed using CCK8, TUNEL, wound healing, Transwell trophoblast cells; PPARγ;
assays. Moreover, the role of peroxisome proliferator-activated receptor gamma (PPARγ), probable gestational diabetes
target of CTRP6, was evaluated through co-transfection with PPARγ overexpression vector. The mellitus; high glucose
results of the present study revealed that CTRP6 and PPARγ were both upregulated in high
glucose-induced cells. And CTRP6 knockdown could significantly elevate the abilities of cell
viability, migration and invasion, and avoid cell apoptosis. In addition, PPARγ overexpression
was found to restrain the protective effects of CTRP6 knockdown on the above aspects, indicating
CTRP6 played a role in trophoblast cells via inhibiting PPARγ expression. In conclusion, CTRP6
regulated the viability, migration and invasion of high glucose-induced gestational trophoblast
cells through PPARγ signaling.

Introduction
Gestational diabetes mellitus (GDM) refers to nor­ diabetes [5]. It is generally believed that the patho­
mal glucose metabolism before pregnancy and genesis of GDM is complex, which may be the
abnormal glucose tolerance first discovered during result of multiple factors such as inflammatory
pregnancy, which is one of the most familiar com­ factors, insulin resistance (IR), pancreatic β-cell
plications of pregnancy [1]. It is manifested as function defects, genetics and environment [6].
metabolic and endocrine disorders, including sig­ Recently, considerable studies have indicated that
nificant changes in the body fluid environment, some adipokines, such as leptin, adiponectin and
such as alterations in the levels of adipokines and visfatin, are compactly relevant to IR and partici­
inflammatory cytokines [2]. As the alterations in pate in the pathogenesis of GDM [2,7,8].
the childbearing age, dietary patterns and lifestyles C1q/tumor necrosis factor related protein is
of contemporary women, the morbidity of GDM is a newly discovered highly conserved adiponectin
increasing gradually [3]. After childbirth, the glu­ family consisting of 15 members (CTRP1-
cose tolerance of most pregnant women with CTRP15) [9]. Therein, CTRP6 is widely expressed
GDM returned to normal, whereas a few would in a variety of tissues and organs, including adi­
develop type 2 diabetes [4]. Moreover, several stu­ pose tissue, heart, brain and placenta [10]. It plays
dies have identified that, compared with healthy different roles in different physiological or patho­
pregnant women, the offspring of pregnant logical processes, and has been proven to be
women with GDM are at a relatively higher risk involved in inflammation, diabetes, and cardiovas­
of long-term complications such as obesity and cular disease [11]. In addition, a previous study

CONTACT Wen-pei Bai baiwp@bjsjth.cn Department of Obstetrics and Gynecology, Beijing Shijitan Hospital, Capital Medical University, 10 Tieyi
Road, Haidian District, Beijing 100038, P.R. China
© 2021 The Author(s). Published by Informa UK Limited, trading as Taylor & Francis Group.
This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted
use, distribution, and reproduction in any medium, provided the original work is properly cited.
BIOENGINEERED 207

revealed that CTRP6 could inhibit the prolifera­ information on the tissue and cell distribution of
tion, invasion and migration of squamous cell all 24,000 human proteins [17] (Figure 1a).
carcinoma cells [12]. And another study deemed Therefore, this study put forward the hypothesis
that CTRP6 could inhibit the proliferation of vas­ that CTRP6 may play a role in GDM. High glucose
cular smooth muscle cells induced by platelet- was used to induce trophoblast cells to establish an
derived growth factor-bb [13]. Furthermore, in vitro model. This paper explored the effects of
knockout of CTRP6 was found to inhibit obesity CTRP6 on the cell phenotypes, covering cell pro­
in mice caused by high-fat diet [14], particularly, liferation, invasion and migration and initially
obesity has been generally considered to be inse­ explored the mechanism.
parable from diabetic pregnancy-eclampsia [15].
The adverse environment in the body of mother
with GDM affects the fetus through the placenta, Methods
and the material exchange between mother and
Cell culture and grouping
fetus principally occurs in the placental tropho­
blast [16]. It happens that CTRP6 is expressed in Human chorionic trophoblast cells (HTR-8/SVneo
the gestational trophoblast cells according to the cells) were obtained from American Type Culture
human protein atlas (HPA) website (https://www. Collection (ATCC) and can be used for the study
proteinatlas.org/), which is dedicated to providing of placental function and trophoblast biology [18].

Figure 1. (a) Expression levels of CTRP6 in different types of cells from the analysis result of HPA website. (b) Cell viability in each
group was assessed using a CCK-8 assay. Expression levels of CTRP6 in each group of cells were determined using (c) Western
blotting and (d) RT-qPCR. Expression levels of PPARγ were determined using (e) Western blotting and (f) RT-qPCR. *p < 0.05,
**P < 0.01, ***P < 0.001.
208 J. ZHANG AND W.-P. BAI

The cells grew adherently, and the culture condi­ a microplate reader (Molecular devices, Shanghai,
tion was RPMI 1640 medium (5.5 mM glucose; China) [21].
Gibco, Thermo Fisher Scientific, China): FBS:
penicillin/streptomycin with 100:10:1, and the
Reverse transcription-quantitative PCR (RT-qPCR)
incubator was adjusted to 37°C, 5% CO2 condi­
tion. The cells were randomly divided into Total RNA was extracted from cells using TRIzol®
a control group and high glucose groups, each (Invitrogen; Thermo Fisher Scientific, China) on ice
containing the final concentration of D-glucose: for 15 min. The purity of the RNA was evaluated
the control group (5.5 mM); the high glucose using a NanoDropTM One Spectrophotometer
group (10, 20, 30 mM) [19]. (Thermo Fisher Scientific, China). Following reverse
transcription of RNA into cDNA product with
PrimeScript RT reagent kit (Takara Biotechnology,
Cell transfection and grouping
Dalian, China), SYBR Green Realtime PCR Master
Cells were seeded in a six-well plate at a density of Mix kit (TOYOBO, Shanghai, China) was used
3 × 105, and transfection was performed [20] when cell according to the manufacturer’s protocol for qPCR.
density reached 50%~60% next day. CTRP6 knock­ The thermocycling condition was as follows: Initial
down was performed with the pLKO.1-short hairpin denaturation at 95°C for 10 min, 40 cycles of dena­
(sh)RNA lentiviral vector (shRNA-CTRP6-1/2), turation at 95°C for 2 sec and annealing at 60°C for
whereas non-targeted shRNA was as the negative 20 sec, and extension at 70°C for 10 sec. The normal­
control (shRNA-NC; Vectorbuilder Biotechnology, ization method was adopted with GADPH as the
Guangzhou, China). Target sequences of shRNAs internal reference, and 2−ΔΔCq method [22] was
were as follows: shRNA-CTRP6-1, 5ʹ-GATGTGT used to perform relative quantification. The primers
GAGATCCCTATGGT-3ʹ; shRNA-CTRP6-2, 5ʹ- (5ʹ-3ʹ) used are as follows: CTRP6, forward:
GCACTTCTCAAACTTGGAAAT-3ʹ; shRNA-NC, CCATCCTGAAAGGTGACAAAGG, reverse: AG
5ʹ-CAACAAGATGAAGAGCACCAA-3ʹ Peroxiso TAATGCGTCTGGCACGAG; PPARγ, forward:
me proliferator-activated receptor gamma (PPARγ) AATGGAAGACCACTCCCACT, reverse: GGTA
overexpression (Ov) was performed with CTCTTGAAGTTTCAGGTC; GAPDH, forward:
a pcDNA3.1 vector (Ov-PPARγ), whereas the empty GGAGCGAGATCCCTCCAAAAT, reverse: GGCT
vector was set as the negative control (Ov-NC; GTTGTCATACTTCTCATGG.
Fenghui Biotechnology, Hunan, China). Cells were
transfected with 10 nM shRNAs and pcDNAs using
Western blot analysis
Lipofectamine® 2000 reagent (Invitrogen; Thermo
Fisher Scientific, China) at 37°C, according to the Protein was extracted from cells on ice with pre­
manufacturer’s protocol. Following 4–6 h of transfec­ cooled RIPA lysis buffer (contains protease inhi­
tion, the wells were washed twice with PBS, and then bitors; Beyotime Biotechnology) for 10 min, and
filled with RPMI-1640 medium containing 10% FBS then quantified using a BCA assay kit (Abcam,
to continue the culture for 48 h. Afterward, the cells Shanghai, China). Protein samples (30 µg/lane)
were divided into a control group, high glucose group were separated by 10% SDS-PAGE at 120 V and
(30 mM), and transfection groups. transferred to PVDF membranes (Millipore,
Thermo Fisher Scientific) at 280 mV for 90 min.
The membranes were first blocked with 5%
CCK-8 assay
skimmed milk for 2 h at room temperature.
The cells were seeded in a 96-well plate at a density Afterward, they were incubated with diluted pri­
of 3 × 103 cells/well. Following the culture main­ mary antibodies against CTRP6 (ab36898; 1:500),
tained for 72 h, 10 µl of CCK-8 reagent (Beyotime PPARγ (ab178860; 1:1,000), Bcl2 (ab32124;
Biotechnology, Shanghai, China) was added, and 1:1,000), Bax (ab32503; 1:1,000), Cleaved caspase3
cells were continued to be incubated at 37°C for (ab32042; 1:500) and GAPDH (ab181602;
1 h in the dark. The absorbance of each well at 1:10,000) at 4°C overnight followed by HRP-
a wavelength of 450 nm was measured using conjugated goat anti-rabbit IgG secondary
BIOENGINEERED 209

antibody (ab6721; 1:10,000; all Abcam) at room room temperature. The results were observed under
temperature for 1 h. The chemiluminescence reac­ an inverted microscope (magnification, x100; Nikon
tion was performed using an ECL kit (Yeasen Corporation) [26].
BioTechnology, Shanghai, China), and densitome­
try was analyzed using the ImageJ software (v1.8;
Statistical analysis
National Institutes of Health) [23].
Each cellular experiment was repeated indepen­
dently at least three times. Data were presented
TUNEL assay
as the mean ± SD and processed using GraphPad
Cells were seeded into a 24-well plate at a density Prism 8.0 (GraphPad Software, San Diego, USA).
of 2 × 105 cells/well and cultured until they Data were in accordance with the normal distribu­
reached ~80% confluence. The operation was con­ tion by Shapiro-Wilk test [27], and comparisons
ducted using the TUNEL kit (Beyotime were conducted using one-way ANOVA [28] fol­
Biotechnology) according to the manufacturer’s lowed by Tukey’s post hoc test [29]. P < 0.05 was
instructions. Following the cover glass was sealed, considered to indicate a statistically significant
the results were observed under a fluorescence difference.
microscope (magnification, x200; Olympus
Corporation, Japan) [24].
Results
In the present study, CTRP6 and PPARγ were
Wound healing assay
found to be upregulated in high glucose-induced
Cells were seeded into a 6-well plate at a density of cells. And CTRP6 knockdown could significantly
5 × 105 cells/well until reaching ~80% confluence. elevate the abilities of cell viability, migration and
A gentle scrape on the well surface was performed invasion, and avoid cell apoptosis. In addition,
with a 200-µl pipette tip. The wells were washed PPARγ overexpression was found to restrain the
with PBS to remove free cells. Following culture in protective effects of CTRP6 knockdown on the
serum-free RPMI-1640 medium at 37°C for 24 h, above aspects, indicating CTRP6 played a role in
images of the wound area were captured at 0/24 h trophoblast cells via inhibiting PPARγ signaling.
under an inverted microscope (magnification,
x100; Nikon Corporation, Japan) [25].
Expression levels of CTRP6 and PPARγ in high
glucose induced-trophoblast cells
Transwell assay
CTRP6 was found to be expressed in gestation
The Matrigel (BD Biosciences) was taken out from trophoblast from the analysis result of HPA web­
−20°C and melted on ice, and quickly diluted with site (Figure 1a). Human chorionic trophoblast cells
serum-free RPMI-1640 medium at a ratio of 1:4. (HTR-8/SVneo cells) were firstly divided into the
50 μl of the diluted Matrigel was added to the control group and high glucose groups (10, 20,
upper chamber of the Transwell (Corning, Merck 30 mM). Cell viability in each group was assessed
KGaA, Germany) for dry at 37°C for 10 h. The using a CCK-8 assay. Following induction with
cells were resuspended in serum-free RPMI-1640, glucose, cell viability was found to be decreased
and the cell density was adjusted to 4 × 104 cells/ in a concentration-dependent manner (Figure 1b).
ml. 100 µl cell suspension was added into the upper Next, the expression levels of CTRP6 in each
chamber, and 500 µl RPMI-1640 containing 20% group of cells were determined using RT-qPCR
FBS was added into lower chamber. Following incu­ and Western blotting. The results revealed that
bation at 37°C for 24 h, the non-invaded cells in the CTRP6 expression level was upregulated accom­
upper chamber were gently wiped away with a wet panied with the increased concentration of glucose
cotton swab. The cells on the submembrane surface (Figure 1c-d). In addition, the expression levels of
were fixed with 4% paraformaldehyde for 20 min PPARγ were also upregulated depending on the
and stained with 0.1% crystal violet for 10 min at elevated concentration of glucose (Figure 1e-f).
210 J. ZHANG AND W.-P. BAI

CTRP6 knockdown improves the viability, inva­ were divided into four groups: ) the control; )
sion and migration of high glucose induced- high glucose (30 mM); ) high glucose (30 mM) +
trophoblast cells shRNA-NC; ) high glucose (30 mM) + shRNA-
In order to explore the role of CTRP6 in high CTRP6. Cell viabilities of these groups of cells
glucose induced-trophoblast cells, its expression were determined using the CCK8 assay, and the
was knocked down by transfection with shRNAs. results demonstrated that CTRP6 knockdown
The expression levels of CTRP6 in the control and could upregulate the viability of high glucose
the transfected cells were determined using RT- induced-cells (Figure 2c). Meanwhile, cell apopto­
qPCR and Western blotting. According to the sis was determined using TUNEL assay and
results, the expression level was lower in the Western blotting. The fluorescence intensity was
shRNA-CTRP6-1 group compared with the enhanced in the high glucose group compared
shRNA-CTRP6-2 group (Figure 2a-b), hence, with the control group, and that was downregu­
these cells transfected with shRNA-CTRP6-1were lated in the CTRP6 knockdown group to a certain
used for the subsequent assays. Afterward, cells extent (Figure 2d-e). In addition, the Western blot

Figure 2. Expression levels of CTRP6 in the control and the transfected cells were determined using (a) Western blotting and (b) RT-
qPCR. (c) Cell viability was determined using a CCK8 assay. Cell apoptosis was determined using (d–e) TUNEL assay and (f) Western
blotting, magnification, x200. *p < 0.05, **P < 0.01, ***P < 0.001.
BIOENGINEERED 211

result revealed that the expression levels of Bax Subsequently, PPARγ overexpression was estab­
and cleaved caspase 3 were downregulated, lished by transfection, and the efficacy was verified
whereas Bcl2 expression was upregulated arisen using RT-qPCR and Western blotting. The results
from CTRP6 knockdown (figure 2f). These indi­ of these two assays both revealed that PPARγ was
cated that CTRP6 knockdown could protect the highly expressed in the transfected cells
aliveness of trophoblast cells from the injury (Figure 4b-c). Following cells were divided into
caused by high glucose. five groups: ) the control; ) high glucose
Moreover, the abilities of cell migration and inva­ (30 mM); ) high glucose (30 mM) + shRNA-
sion were assessed using wound healing and Transwell CTRP6; ) high glucose (30 mM) + shRNA-
assays. The result of wound healing assay displayed CTRP6 + Ov-NC; ) high glucose (30 mM) +
that high glucose caused the inhibited cell migration shRNA-CTRP6 + Ov-PPARγ, cell viability was
and CTRP6 knockdown promoted it (Figure 3a). determined in these five groups using the CCK8
Coincidentally, CTRP6 knockdown likewise recov­ assay. The result indicated that PPARγ overexpres­
ered the inhibiting effect of high glucose on cell inva­ sion downregulated high glucose-induced cell via­
sion from the result of Transwell assay (Figure 3b). To bility, breaking the effect of CTRP6 knockdown on
sum up, CTRP6 knockdown improved cell survival, the viability (Figure 4d). Furthermore, cell apop­
invasion and migration of trophoblast cells which tosis was determined similarly as aforementioned.
were subjected to high glucose. Focusing on the results of the fifth group, PPARγ
CTRP6 knockdown inhibits PPARγ expression and overexpression significantly promoted cell apopto­
improves the viability, invasion and migration of high sis, which is equivalent to reversing the impact of
glucose induced-trophoblast cells by inhibiting PPARγ CTRP6 knockout on cell survival (Figure 4e-g).
The expression levels of PPARγ in the four Finally, cell migration and invasion was also
groups of cells mentioned above were determined assessed using wound healing and Transwell
using Western blotting. Its expression was upre­ assay. The results suggested that PPARγ over­
gulated in high glucose group and downregulated expression could not only suppress cell migra­
in the CTRP6 knockdown group, indicating insuf­ tion but also inhibit the invasion (Figure 5a-d).
ficient CTRP6 expression may also cause a decline These results indicated that CTRP6 knockdown
in the expression of PPARγ (Figure 4a). improved the invasion and migration of high

Figure 3. The ability of cell migration and invasion was assessed using (a) wound healing and (b) Transwell assay, magnification,
x100. ***P < 0.001.
212 J. ZHANG AND W.-P. BAI

Figure 4. (a) Expression levels of PPARγ in four groups of cells were determined using Western blotting. The efficacy of PPARγ overexpression
was verified using (b) RT-qPCR and (c) Western blotting. (d) Cell viability was determined in five groups using a CCK8 assay. Cell apoptosis was
determined using (e-f) TUNEL assay and (g) Western blotting, magnification, x200. *p < 0.05, **P < 0.01, ***P < 0.001.

glucose induced-trophoblast cells by inhibiting and fetal development. It is also one of the predictors
PPARγ expression. of long-term type 2 diabetes in pregnant women and
their offspring [5]. Those with severe GDM or poor
control of blood glucose may ultimately suffer from
Discussion some adverse pregnancy outcomes, such as hyper­
GDM is a familiar perinatal comorbidity that influ­ tension of pregnancy, premature delivery, stillbirth,
ences maternal transformation, placenta formation fetal development and giant babies [30]. Therefore,
BIOENGINEERED 213

Figure 5. The ability of cell migration and invasion was assessed using (a) wound healing and (b) Transwell assay, magnification,
x100. (c) Quantitative result of wound healing assay and (d) Transwell assay. *p < 0.05, ***P < 0.001.

reasonable control of the condition and blood glu­ Therefore, this study selected trophoblast cells
cose can greatly reduce the impact on the health of as a model to study the regulatory functions of
mothers and children. During pregnancy, the pla­ CTRP6. As aforementioned, the phenotype of
centa is a highly specific organ, it not only has the trophoblast cells is intimately relevant to fetal
functions of supplying nutrients to the fetus, exchan­ development [32]. Therefore, following confirm­
ging gas and removing metabolites, but also has ing that the expression level of CTRP6 was upre­
immune and endocrine functions [31]. The forma­ gulated in high glucose-induced cells, its effects
tion of placenta principally depends on the extra­ on cell proliferation, migration and invasion were
chorionic trophoblast to invade the membrane, and studied through silencing its expression. The
trophoblast cells serve as the first barrier between the results demonstrated that CTRP6 knockdown
mother and the fetus; its proliferation, differentia­ could maintain the viability of cells stimulated
tion, invasion, and apoptosis are associated with the by high glucose, inhibit their apoptosis, and alle­
normality of the placenta and the development of viate the inhibitory effects of high glucose on cell
fetus [32]. Moreover, based on the direct correlation migration and invasion. Hence, these findings
between trophoblast cells and the mother, the mater­ indicate that inhibiting CTRP6 expression may
nal blood glucose alterations directly influence the reduce the adverse effects on fetal development
function of trophoblast cells. The trophoblast cells under high glucose condition. A clinical report
are the first to be involved and undergo various revealed that compared with healthy individuals,
pathological changes to cope with the high glucose serum CTRP6 levels in obese individuals were
stress environment [33,34]. significantly upregulated. More importantly, the
214 J. ZHANG AND W.-P. BAI

receiver operating characteristic curve results sug­ Funding


gested that CTRP6 levels were associated with IR
This study was supported by Beijing Haidian District health
[35]. In addition, a previous study found that development scientific research and cultivation plan
CTRP6 knockdown could suppress the inflamma­ (HP2021-05-50602); Youth fund of Beijing Shijitan Hospital
tion, oxidative stress and extracellular matrix Affiliated to Capital Medical University (2019-q20).
accumulation in high glucose-induced glomerular
mesangial cells, which provides a basis for CTRP6
to be applied in the treatment of diabetic nephro­ Availability of data and materials
pathy [36]. The datasets used and/or analysed during the present study
Furthermore, CTRP6 overexpression was reported are available from the corresponding author on reasonable
to activate the PPARγ signal to relieve hypertension request.
and vascular endothelial dysfunction in spontaneously
hypertensive rats [37]. And its knockdown was
Authors’ contributions
deemed to inhibit fat production in 3T3-L1 adipocytes
through PPARγ signaling [38]. These indicated that JZ and WB made contributions to content design and parti­
cipated in the experiments; JZ analyzed the data and draft the
PPARγ might act as the downstream target of CTRP6.
manuscript; WB examined the data and critically revised the
Especially, a previous study revealed that PPARγ manuscript for important intellectual content. Both of them
expression was upregulated in GDM placental tissue confirmed the authenticity of all the raw data and approved
and trophoblast cells under high glucose environment the final manuscript.
[39]. Herein, following the determination of upregu­
lated PPARγ expression level in high glucose-induced
trophoblast cells, the effects of PPARγ on cell pheno­ References
type were studied by urging it to overexpress. The [1] American College of Obstetricians and Gynecologists
results in this study suggested that PPARγ overexpres­ Committee on Practice Bulletins. ACOG practice bul­
sion reversed the influences of CTRP6 knockdown on letin no. 190: gestational diabetes mellitus. Obstet
cell phenotype. In other words, PPARγ overexpres­ Gynecol. 2018 Feb;131(2):e49–e64.
sion could suppress cell viability, migration and inva­ [2] de Gennaro G, Palla G, Battini L, et al. The role of
adipokines in the pathogenesis of gestational diabetes
sion, and promote cell apoptosis, indicating CTRP6
mellitus. Gynecol Endocrinol. 2019 Sep;35(9):737–751.
play a role in altering the phenotype of high glucose- [3] Chiefari E, Arcidiacono B, Foti D, et al. Gestational
induced trophoblast cells through regulating PPARγ diabetes mellitus: an updated overview. J Endocrinol
expression. This article proposes for the first time that Invest. 2017 Sep;40(9):899–909.
CTRP6 affects GDM through PPARγ signaling. [4] Szmuilowicz ED, Josefson JL, Metzger BE. Gestational
However, this study is limited to the cellular level, diabetes mellitus. Endocrinol Metab Clin North Am.
2019 Sep;48(3):479–493.
clinical samples will be collected for in-depth research
[5] Johns EC, Denison FC, Norman JE, et al. Gestational
in the future. diabetes mellitus: mechanisms, treatment, and
complications. Trends Endocrinol Metab. 2018 Nov;29
(11):743–754.
Conclusion [6] Plows JF, Stanley JL, Baker PN, et al. The pathophy­
siology of gestational diabetes mellitus. Int J Mol Sci.
To sum up, the present study has found that
2018 Oct 26;19(11):11.
CTRP6 knockdown promotes the viability, inva­ [7] Pérez-Pérez A, Vilariño-García T, Guadix P, et al.
sion and migration of high glucose induced- Leptin and nutrition in gestational diabetes.
trophoblast cells via inhibiting PPARγ expression. Nutrients. 2020 Jul 2;12(7):1970.
The findings of the present study may promote [8] Radzicka S, Pietryga M, Iciek R, et al. The role of
CTRP6 as a biomarker or therapeutic target for the visfatin in pathogenesis of gestational diabetes
(GDM). Ginekol Pol. 2018;89(9):518–521.
treatment of GDM.
[9] Si Y, Fan W, Sun L. A review of the relationship
between CTRP family and coronary artery disease.
Curr Atheroscler Rep. 2020 May 28;22(6):22.
Disclosure statement
[10] Wong GW, Krawczyk SA, Kitidis-Mitrokostas C, et al.
No potential conflict of interest was reported by the authors. Molecular, biochemical and functional characterizations
BIOENGINEERED 215

of C1q/TNF family members: adipose-tissue-selective [23] Wang Y, Zhang L, Zhou X. Activation of Nrf2 signaling
expression patterns, regulation by PPAR-gamma agonist, protects hypoxia-induced HTR-8/SVneo cells against
cysteine-mediated oligomerizations, combinatorial asso­ ferroptosis. J Obstet Gynaecol Res. 2021 Nov;47
ciations and metabolic functions. Biochem J. 2008 Dec (11):3797–3806.
1;416(2):161–177. [24] Li J, Tong C, Xu P, et al. QSOX1 regulates trophoblas­
[11] Sadeghi A, Fadaei R, Moradi N, et al. Circulating levels tic apoptosis in preeclampsia through hydrogen per­
of C1q/TNF-α-related protein 6 (CTRP6) in polycystic oxide production. J Matern Fetal Neonatal Med. 2019
ovary syndrome. IUBMB Life. 2020 Jul;72 Nov;32(22):3708–3715.
(7):1449–1459. [25] Zhang X, Li BY, Fu LJ, et al. Stomatin-like protein 2
[12] Hano K, Hatano K, Saigo C, et al. An adiponectin (SLP2) regulates the proliferation and invasion of tro­
paralog protein, CTRP6 decreased the proliferation phoblast cells by modulating mitochondrial functions.
and invasion activity of oral squamous cell carci­ Placenta. 2020 Oct;100:13–23.
noma cells: possible interaction with laminin recep­ [26] Fang YN, Huang ZL, Li H, et al. Highly expressed
tor pathway. Mol Biol Rep. 2019 Oct;46 miR-182-5p can promote preeclampsia progression by
(5):4967–4973. degrading RND3 and inhibiting HTR-8/SVneo cell
[13] Dong X, Hu H, Fang Z, et al. CTRP6 inhibits invasion. Eur Rev Med Pharmacol Sci. 2018 Oct;22
PDGF-BB-induced vascular smooth muscle cell prolif­ (20):6583–6590.
eration and migration. Biomed Pharmacothe. 2018 [27] Vetter TR. Fundamentals of research data and vari­
Jul;103:844–850. ables: the devil is in the details. Anesth Analg. 2017
[14] Wu W, Zhang J, Zhao C, et al. Lentivirus-mediated Oct;125(4):1375–1380.
CTRP6 silencing ameliorates diet-induced obesity in [28] Wei P, Ru D, Li X, et al. Exposure to environmental
mice. Exp Cell Res. 2018 Jun 1;367(1):15–23. bisphenol A inhibits HTR-8/SVneo cell migration and
[15] Yen IW, Lee CN, Lin MW, et al. Overweight and invasion. J Biomed Res. 2020 Jun 30;34(5):369–378.
obesity are associated with clustering of metabolic [29] Hazra A, Gogtay N. Biostatistics series module 3: com­
risk factors in early pregnancy and the risk of GDM. paring groups: numerical variables. Indian J Dermatol.
PloS One. 2019;14(12):e0225978. 2016 May-Jun;61(3):251–260.
[16] Knöfler M, Haider S, Saleh L, et al. Human placenta [30] Pan JP, Liang SS, Huang MY, et al. Obstetric and neonatal
and trophoblast development: key molecular mechan­ outcomes after frozen-thawed embryos transfer with
isms and model systems. Cell Mol Life Sci. 2019 Sep;76 laser-assisted hatching: a retrospective cohort study. Arch
(18):3479–3496. Gynecol Obstet. 2021 Aug 14. DOI:10.1007/s00404-021-
[17] Thul PJ, Åkesson L, Wiking M, et al. A subcellular map 06153-0.
of the human proteome. Science (New York, NY). 2017 [31] Rolfo A, Nuzzo AM, De Amicis R, et al. Fetal-maternal
May 26;356(6340):6340. exposure to endocrine disruptors: correlation with diet
[18] Msheik H, Azar J, El Sabeh M, et al. HTR-8/SVneo: intake and pregnancy outcomes. Nutrients. 2020 Jun
a model for epithelial to mesenchymal transition in the 11;12(6). DOI:10.3390/nu12061744.
human placenta. Placenta. 2020 Jan 15;90:90–97. [32] West RC, Ming H, Logsdon DM, et al. Dynamics of
[19] Liu L, Zhang J, Liu Y. MicroRNA-1323 serves as trophoblast differentiation in peri-implantation-stage
a biomarker in gestational diabetes mellitus and aggra­ human embryos. Proc Natl Acad Sci U S A. 2019
vates high glucose-induced inhibition of trophoblast Nov 5;116(45):22635–22644.
cell viability by suppressing TP53INP1. Exp Ther [33] Lorenzon AR, Moreli JB, de Macedo Melo R, et al.
Med. 2021 Mar;21(3):230. Stromal cell-derived factor (SDF) 2 and the endoplasmic
[20] Wang Q, Lu X, Li C, et al. Down-regulated long reticulum stress response of trophoblast cells in gesta­
non-coding RNA PVT1 contributes to gestational dia­ tional diabetes mellitus and in vitro hyperglycaemic
betes mellitus and preeclampsia via regulation of condition. Curr Vasc Pharmacol. 2021;19(2):201–209.
human trophoblast cells. Biomed Pharmacothe. 2019 [34] Blundell C, Tess ER, Schanzer AS, et al.
Dec;120:109501. A microphysiological model of the human placental
[21] Zhang L, Zhao F, Li C, et al. Hypomethylation of DNA barrier. Lab Chip. 2016 Aug 2;16(16):3065–3073.
promoter upregulates ADAMTS7 and contributes to [35] Liao X, Liu S, Tang X, et al. Circulating CTRP6 levels are
HTR-8/SVneo and JEG-3 cells abnormalities in increased in overweight or obese Chinese individuals and
pre-eclampsia. Placenta. 2020 Apr;93:26–33. associated with insulin resistance parameters: a pilot study.
[22] Livak KJ, Schmittgen TD. Analysis of relative gene Exp Clin Endocrinol Diabetes. 2021 Jul;129(7):535–541.
expression data using real-time quantitative PCR and [36] Xu E, Yin C, Yi X, et al. Knockdown of CTRP6 inhibits
the 2(-Delta Delta C(T)) method. Methods. 2001 high glucose-induced oxidative stress, inflammation and
Dec;25(4):402–408. extracellular matrix accumulation in mesangial cells
216 J. ZHANG AND W.-P. BAI

through regulating the Akt/NF-κB pathway. Clin Exp [38] Wu WJ, Mo DL, Zhao CZ, et al. Knockdown of CTRP6
Pharmacol Physiol. 2020 Jul;47(7):1203–1211. inhibits adipogenesis via lipogenic marker genes and Erk1/
[37] Chi L, Hu X, Zhang W, et al. Adipokine CTRP6 2 signalling pathway. Cell Biol Int. 2015 May;39
improves PPARγ activation to alleviate angiotensin (5):554–562.
II-induced hypertension and vascular endothelial [39] Suwaki N, Masuyama H, Masumoto A, et al. Expression
dysfunction in spontaneously hypertensive rats. and potential role of peroxisome proliferator-activated
Biochem Biophys Res Commun. 2017 Jan 22;482 receptor gamma in the placenta of diabetic pregnancy.
(4):727–734. Placenta. 2007 Apr;28(4):315–323.

You might also like