Journal Pre-proof TBX3 deficiency accelerates apoptosis in cardiomyoblasts through regulation of P21 expression Meiling Cao, Binlu Zhu, Yuanyuan Sun, Xueqi Zhao, Guangrong Qiu, Weineng Fu, Hongkun Jiang PII:
S0024-3205(19)30967-1
DOI:
https://doi.org/10.1016/j.lfs.2019.117040
Reference:
LFS 117040
To appear in:
Life Sciences
Received Date: 22 July 2019 Revised Date:
23 October 2019
Accepted Date: 31 October 2019
Please cite this article as: M. Cao, B. Zhu, Y. Sun, X. Zhao, G. Qiu, W. Fu, H. Jiang, TBX3 deficiency accelerates apoptosis in cardiomyoblasts through regulation of P21 expression, Life Sciences, https:// doi.org/10.1016/j.lfs.2019.117040. This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. Please note that, during the production process, errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain. © 2019 Published by Elsevier Inc.
1
TBX3 deficiency accelerates apoptosis in cardiomyoblasts through regulation of
2
P21 expression
3
Meiling Cao1, Binlu Zhu2, Yuanyuan Sun3, Xueqi Zhao2, Guangrong Qiu3, Weineng
4
Fu3, Hongkun Jiang2, *
5
1
6
University, Shenyang 110001, People’s Republic of China
7
2
8
Shenyang 110001, People’s Republic of China
9
3
Department of Neonatology, The First Affiliated Hospital of China Medical
Department of Pediatrics, The First Affiliated Hospital of China Medical University,
Department of Medical Genetics, College of Basic Medical Sciences, China Medical
10
University, Shenyang 110122, People’s Republic of China
11
*
12
Affiliated Hospital of China Medical University, 155 North Nanjing Street, Shenyang
13
110001, People’s Republic of China
14
Tel/Fax: +86-24-83282527
15
16
17
18
19
20
Corresponding author: Dr. Hongkun Jiang, Department of Pediatrics, The First
21
Abstract
22
Congenital heart disease (CHD) is the most common birth defect in newborns.
23
There is increasing evidence that apoptosis and remodeling of the cardiomyoblasts are
24
the major pathology of CHD. Previous research found that T-box transcription factor
25
3 (TBX3) was compulsory for the regulation of proliferation, cell cycle arrest and
26
apoptosis in various cells. Hence, TBX3 might be involved in the treatment of CHD.
27
The primary aim of this study was to study the effects of TBX3 on apoptosis in aged
28
cardiomyoblasts and investigate the latent mechanism. In the present study, we found
29
TBX3 knockdown induced proliferation inhibition, cell cycle arrest and apoptosis
30
accompanied by mitochondrial dysfunction in cardiomyoblasts at passage 10 to 15.
31
Apoptosis-inducing effects of TBX3 silence could be neutralized by silencing P21
32
using specific siRNA. In addition, the mRNA and protein expression levels of TBX3
33
in the heart tissues of sporadic type CHD donors were obviously down-regulated. In
34
conclusion, we demonstrated that TBX3 deficiency accelerated apoptosis via directly
35
regulating P21 expression in senescent cardiomyoblasts.
36
Keywords: Congenital heart disease; TBX3; Cell cycle arrest; Apoptosis
37
Abbreviations: ATPase, adenosine triphosphatase; CDK, cyclin dependent kinase;
38
CHD, congenital heart disease; ROS, reactive oxygen species; TBX3, T-box
39
transcription factor 3; UMS, ulnar-mammary syndrome.
40
1. Introduction
41
Congenital heart disease (CHD), the leading non-infectious cause of morbidity
42
and mortality in newborn children in the world, is a life-threatening congenital
43
malformation associated with genetic backgrounds and environmental risk factors.
44
According to the clinical manifestation, CHD can be classified into two types,
45
including acyanotic congenital heart disease and cyanotic congenital heart disease
46
[1-2]. An increasing number of epidemiological investigations found that the
47
incidence of CHD was 19 to 75 of every 1000 living newborns [3]. If the infants who
48
did not survive were calculated, CHD incidence would be higher [4-5]. Despite
49
simple CHD can be completely remitted at present, early surgical management that
50
has high risk is the necessary therapeutic strategy for complex CHD. As known,
51
although multi-gene abnormalities have been confirmed in the occurrence of CHD,
52
the accurate mechanism of CHD is remained to be investigated. Programmed cell
53
death plays a crucial part in maintaining normal cardiac function under physiological
54
conditions and apoptosis-related protein is expressed at the basal level in the postnatal
55
hearts [6-7]. A clinical assessment indicated that aggravated apoptosis could be found
56
in the cardiac tissue samples from CHD patients compared with the normal heart
57
tissues from the aged-matched subjects [8]. In addition, promoting proliferation and
58
suppressing apoptosis exerted protective effects in H9C2 cardiomyoblasts,
59
highlighting that apoptosis inhibition might be a potential therapeutic strategy for
60
CHD [9-10]. Hence, we believe that inhibition of apoptosis in cardiomyoblasts might
61
be a promising therapeutic strategy to inhibit CHD development.
62
T-box transcription factor 3 (TBX3) belongs to the T-box factor family that is
63
composed of evolutionary conserved transcription factors regulating early embryonic
64
development [11]. TBX3 has established role in the development of the heart,
65
mammary glands, limbs and genitalia in the vertebrates [12]. In addition, human
66
TBX3 gene mutation leads to ulnar-mammary syndrome (UMS), which is
67
characterized by upper limb malformation, mammary hypoplasia, as well as
68
deficiency in hair, tooth and reproductive system [13-14]. On the contrary, the
69
oncogenic effects of TBX3 overexpression have emerged in a wide variety of cancer,
70
including gastric cancer, breast cancer and pancreatic cancer [15-17]. A recent study
71
showed that the occurrence and development of conotruncal heart defect could be
72
attributed to the variants of TBX3 [18]. Hence, the effects of TBX3 in
73
cardiomyoblasts are worth to be emphasized in CHD. Except for the definite
74
regulation role on the development of multiple organs, TBX3 expression possesses
75
the function on modulating cell cycle. The previous experiments have shown that the
76
protein expression of TBX3 is elevated at S phase and TBX3 silence results in S
77
phase arrest, indicating the regulation of TBX3 in cell cycle [19]. Moreover, the
78
previous study also has illustrated that the function of TBX3 on overriding cell cycle
79
to inhibit apoptosis and enhance proliferation is achieved by suppressing cell cycle
80
regulatory protein P21 (also named as P21 waf1/cip1 and P21/CDKN1A) [19]. As the
81
downstream signaling molecule of P53, P21 serves as the inhibitor of CDK family in
82
both P53-dependent and P53-independent manners, which is necessary for adjusting
83
cell cycle and proliferation [20]. Cell cycle transitions (G1 phase to S phase and G2
84
phase to mitosis) is depended on continuous activation and inactivation of CDK
85
proteins that are attached to serine/threonine protein kinases [21]. For instance, the
86
activation of CDK4 and CDK6 by cyclin D1 senses the mitogenic signals and serves
87
as the growth factor sensors in G1 phase, cyclin E1 and E2-induced CDK activation
88
triggers the transition from G1 phase to S phase [22]. Plus, the first identified CKI
89
protein P21 suppresses diverse cell cycle progression through disrupting the
90
interactions between CDKs and cyclin proteins [23]. Several lines of evidence
91
demonstrate that the mitochondrion involves in regulation of apoptosis in mammals.
92
Mitochondria are the cellular bioenergy centers in a physiological manner [24]. In
93
addition to participating in regulation of energy generation, intercellular
94
communication and metabolism, the importance of mitochondria in apoptosis has
95
been
96
inter-membrane space to the cytoplasm and triggers the apoptosis-related events at
97
molecular levels [26]. Cytosolic cytochrome C interacts with Apaf-1, recruits
98
procaspase-9, assembles cytochrome c/Apaf-1/caspase-9 apoptosomes, and thereby
99
initiating downstream apoptotic cascade [27]. These events above suggested that
100
TBX3 might be the important bond between the pathogenesis of CHD and
101
pathological apoptosis as well as cell cycle arrest in cardiomyoblasts. Therefore, the
102
effects of TBX3 in the pathogenesis of CHD are worthy to be investigated.
well
established
[25].
Cytochrome
C
migrates
from
mitochondrial
103
Here we aimed to investigate the effects of TBX3 on mitochondrial function,
104
apoptosis and cell cycle arrest in aged H9C2 cardiomyoblasts and explore the
105
potential mechanism of CHD occurrence.
106
2. Materials and methods
107
2.1 Cell treatment
108
2.1.1 Cell culture
109
Rat cardiomyoblast H9C2 cell line was obtained from Zhong Qiao Xin Zhou
110
Biotechnology Co., Ltd. (Shanghai, China). H9C2 cardiomyoblasts at passage 10 to
111
15 were employed. H9C2 cardiomyoblasts were grown in Dulbecco’s modifed
112
Eagle’s medium (DMEM) (Gibco, USA) supplemented with 10% fetal bovine serum
113
(FBS) (Hyclone, USA) at 37 °C with 5% CO2 in a humidified incubator.
114
2.1.2 Transfection
115
H9C2 cardiomyoblasts at passage 10 to 15 were transfected with TBX3-specific
116
small interfering RNA (siRNA) 1-5 or control siRNA (Wanlei Biological Technology
117
Co., Ltd., Shenyang, China) according to the instructions. Then, the transfected H9C2
118
cardiomyoblasts were maintained in fresh culture medium for 48 h before the
119
transfection efficiency detection.
120 121 122 123 124 125 126 127 128 129
The sequences for TBX3 siRNA1-5 and control siRNA were listed as follows: TBX3 siRNA-1: 5’-CCAUGAGAGAUCCGGUUAUTT-3’ 5’-AUAACCGGAUCUCUCAUGGTT-3’ TBX3 siRNA-2: 5’-GCGAAUGUUCCCUCCAUUUTT-3’ 5’-AAAUGGAGGGAACAUUCGCTT-3’ TBX3 siRNA-3: 5’-GCUGAUGACUGUCGAUAUATT-3’ 5’-UAUAUCGACAGUCAUCAGCTT-3’ TBX3 siRNA-4: 5’-CCAACAACAUUUCGGAUAATT-3’ 5’-UUAUCCGAAAUGUUGUUGGTT-3’ TBX3 siRNA-5: 5’-GCAGUCCAUGAGGGUGUUUTT-3’
5’-AAACACCCUCAUGGACUGCTT-3’
130
Control siRNA: 5’-UUCUCCGAACGUGUCACGUTT-3’
131
5’-ACGUGACACGUUCGGAGAATT-3’
132
133
2.1.3 Transduction
134
H9C2 cardiomyoblasts at passage 10 to 15 were transduced with TBX3-expressing
135
lentivirus or negative control lentivirus (Wanlei Biological Technology Co., Ltd.,
136
Shenyang, China) and all the transduction protocols were carried out according to the
137
manufacturer’s instruction. Afterwards, the transduced H9C2 cardiomyoblasts were
138
maintained in fresh culture medium for 48 h before harvest.
139
2.2 Real-time polymerase chain reaction (RT-PCR)
140
After the corresponding treatment, the total RNA of aforementioned H9C2
141
cardiomyoblasts or heart samples was extracted by using Trizol reagent. After TBX3
142
cDNA was synthesized by using the M-MLV reverse transcriptase (TianGEN, China),
143
RT-PCR analysis was conducted with SYBR Green master mix kits in the Exicycler
144
96 System (Bioneer, Korea). TBX3 mRNA level was normalized to that of GAPDH
145
and calculated by 2 -△△CT method.
146
The primers for RT-PCR were as follows:
147
Rat
148
5’-GTTGGCTCTGACGATGTGG-3’
149
Rat
150
5’-TAGGAGCCAGGGCAGTAA-3’
151
Human
152
5’-TCGCCTTCCCGACTTGGT-3’
153
Human
TBX3
forward:
GAPDH
TBX3
GAPDH
5’-TCGGATAAACACGGATTTACTT-3’;
forward:
forward:
forward:
5’-ACGTTGACATCCGTAAAGAC-3’;
5’-GGCATCCCGTTCTCCTCCCT-3’;
5’-GACCTGACCTGCCGTCTAG-3’;
reverse:
reverse:
reverse:
reverse:
154
5’-AGGAGTGGGTGTCGCTGT-3’
155
2.3 Cell counting kit-8 (CCK-8) assay
156
To assess the viability of aged H9C2 cardiomyoblasts following treatment, H9C2
157
cardiomyoblasts at passage 10 to 15 were seeded in plates at a density of 5×103
158
cells/well. After the transfection or transduction described above, the H9C2
159
cardiomyoblasts were incubated for another 0, 24 or 48 h before the exposure to
160
CCK8 solution (KeyGEN BioTECH, China) according to the manufacturer’s
161
instructions. The absorbance at 450 nm was measured by a microplate reader.
162
2.4 Cell cycle analysis
163
Cell cycle distribution of the aforementioned H9C2 cardiomyoblasts was
164
analyzed by using flow cytometry. To be brief, H9C2 cardiomyoblasts at passage 10
165
to 15 were harvested 48 h after the respective treatment, fixed in 70% ethanol at 4˚C
166
for 2 h, treated with propidium iodide (PI) dye (Beyotime Institute of Biotechnology,
167
China) at 37˚C for 30 min, and subsequently analyzed by using a flow cytometer
168
(NovoCyte, United States).
169
2.5 Hochest 33342 staining
170
Hoechst 33342 staining was implemented to observe the apoptotic morphological
171
characteristics of aged H9C2 cardiomyoblasts. Simply, H9C2 cardiomyoblasts at
172
passage 10 to 15 were plated in a 6-well plate at a density of 1×105 cells/well.
173
Following the transfection or transduction protocol, the H9C2 cardiomyoblasts were
174
incubated for another 48 h, washed with PBS, and stained by Hoechst 33342
175
according to the manufacturer’s instruction. Finally, apoptotic morphological
176
alterations in the stained H9C2 cardiomyoblasts were observed under a fluorescence
177
microscope at 400 × magnification.
178
2.6 Apoptosis analysis
179
The influence of TBX3 expression on apoptosis in aged H9C2 cardiomyoblast
180
was examined using flow cytometry. Simply, H9C2 cardiomyoblasts at passage 10 to
181
15 were plated in a 6-well plate at a density of 1×105 cells/well. Following the
182
transfection or transduction protocol, the H9C2 cardiomyoblasts were stained by
183
adding Annexin V-fluorescein isothiocyanate (FITC) and PI in dark at room
184
temperature for 10-20 min. finally, apoptosis rate of the treated H9C2
185
cardiomyoblasts was detected by using a flow cytometer (NovoCyte, United States).
186
2.7 Mitochondrial membrane potential detection
187
The changes of mitochondrial membrane potential in aforementioned H9C2
188
cardiomyoblasts were detected by using specific fluorescent probe JC-1 according to
189
the requirement of mitochondrial membrane potential detection kit (Beyotime
190
Institute of Biotechnology, China). The relative fluorescence intensity (excitation and
191
emission wavelengths of 485 and 525 nm, excitation and emission wavelengths of
192
525 and 590 nm, respectively) represented the change of mitochondrial membrane
193
potential.
194
2.8 Adenosine triphosphatase (ATPase) activity
195
Treated H9C2 cardiomyoblasts were lysed with PBS and the protein
196
concentrations of lysis were determined by BCA protein quantification kit (Beyotime
197
Institute of Biotechnology, China). In short, the activity of ATPase in H9C2
198
cardiomyoblasts was quantified by corresponding commercial kits (Nanjing Jiancheng
199
Bioengineering Institute, China). The activity of ATPase was expressed as U per
200
milligram protein (U/mg prot).
201
2.9 Mitochondrial reactive oxygen species (ROS) production
202
Mitochondria of the H9C2 cardiomyoblasts at passage 10 to 15 were separated by
203
centrifugation as recommended by the manufacturer (Beyotime Institute of
204
Biotechnology, China). The separated mitochondria were incubated with 10 µM
205
fluorescence probe DCFH-DA for 30 min at 37 °C followed by fluorescence intensity
206
detection according to the instructions of ROS detection kit (Beyotime Institute of
207
Biotechnology, China).
208
2.10 Immunofluorescence
209
H9C2 cardiomyoblasts at passage 10 to 15 were treated as described in cell
210
treatment, fixed with 4% paraformaldehyde for 15 min, and then incubated with 0.1%
211
tritonX-100 (Beyotime Institute of Biotechnology, China) for 30 min at room
212
temperature. After wash with PBS for three times, the treated H9C2 cardiomyoblasts
213
were incubated with primary Ki67 antibody (Cell Signaling Technology, United States,
214
dilution: 1:100) overnight at 4 °C followed by Cy3-labeled goat anti-rabbit IgG (H +
215
L) (Solarbio, China). Afterwards, 4’, 6-diamidino-2-phenylindole (DAPI, Beyotime
216
Institute of Biotechnology, China) incubation was utilized to stain the nuclei. Finally,
217
immune-positive H9C2 cardiomyoblasts were observed under a fluorescence
218
microscope at 400 × magnification.
219
2.11 Chromatin immunoprecipitation (ChIP) assay
220
ChIP assay was implemented to detect the targeted relationship between TBX3
221
and P21. Briefly, the aforementioned H9C2 cardiomyoblasts were fixed in 1%
222
formaldehyde solution at room temperature for 10 min, washed with PBS, and
223
harvested by centrifugation at 2000rpm for 5 min. The chromatin was isolated and
224
exposed to sonication to obtain DNA fragments (fractionated size: 500-1000 bp) that
225
were in turn immunoprecipitated with TBX3 antibody. Afterwards, the TBX3
226
protein-bound DNA was purified by a DNA purifcation kit (Wanlei Biological
227
Technology Co., Ltd., Shenyang, China). Finally, the purified DNA was utilized to
228
analyze the combination between TBX3 and P21.
229
2.12 Western blot
230
The mitochondrial and total protein samples were isolated from the
231
aforementioned H9C2 cardiomyoblasts. Briefly, the same amount of protein samples
232
were separated by 8%, 10% or 14% sodium dodecyl sulfate polyacrylamide gel
233
electrophoresis (SDS-PAGE) and transferred onto PVDF membranes (Millipore, USA)
234
that were subsequently blocked by 5% non-fat milk for 1 h at the room temperature.
235
Afterwards, the PVDF membranes were subjected to the relevant primary antibody at
236
4 °C overnight, respective horseradish peroxidase (HRP) labeled secondary antibody
237
(Solarbio, China) for 45 min at 37 °C, and ultimately the ECL kit (Solarbio, China).
238
The quantification of the protein expression was implemented by Gel-Pro-Analyzer
239
software.
240
The primary antibodies were as follows: TBX3 (Abcam, UK), cyclin A (Abcam,
241
UK), cyclin D1 (Cell Signaling Technology, United States), cyclin B1 (Cell Signaling
242
Technology, United States), caspase-3 (Boster, China), cleaved caspase-3 (Cell
243
Signaling Technology, United States), caspase-9 (Abcam, UK), cleaved caspase-9
244
(Cell Signaling Technology, United States), Bcl-2 (proteintech, China), Bax
245
(proteintech, China), cleaved-PARP (Cell Signaling Technology, United States),
246
cytochrome C (Cell Signaling Technology, United States), P21 (Abclonal, China).
247
Patient samples
248
Eight heart samples from CHD fetuses (24-25 weeks) were recruited in the First
249
Affiliated Hospital of China Medical University. The sporadic types of CHD included
250
ventricular septal defect, atrioventricular septal defect and atrial septal defect. Eight
251
healthy hearts came from fetuses (24-25 weeks) with other congenital defects. Heart
252
tissue collection was implemented after abortion. RT-PCR and TBX3 ELISA (USCN,
253
China) were used to detect heart TBX3 mRNA and protein levels. The procedures
254
were in accordance with the Declaration of Helsinki and the protocols were
255
accordance with the Ethics Committee of the First Affiliated Hospital of China
256
Medical University.
257
2.13 Statistical analysis
258
Data were presented as mean ± standard derivations (SD) and calculated by
259
GraphPad Prism 7.0 (GraphPad Software, United States). Significant differences
260
between two groups were analyzed by t-test and data from three or more groups was
261
determined using one way analysis of variance (ANOVA) followed by a Tukey’s
262
multiple comparison test. Value of p less than 0.05 was considered statistically
263
significant. All in vitro experiments were performed three times (n=3).
264
3. Results
265
3.1 Effect of TBX3 on proliferation in H9C2 cardiomyoblasts
266
CCK-8 assay was utilized to inspect the influence of TBX3 expression on
267
proliferation of H9C2 cardiomyoblasts. We used TBX3 specific siRNA to silence
268
CXCR2 gene in H9C2 cardiomyoblasts, meanwhile, TBX3-expressing lentivirus was
269
used to induce TBX3 overexpression (Supplement Fig. S1-2). As shown in Fig. 1A,
270
TBX3 knockdown significantly decreased H9C2 cardiomyoblast viability in a time
271
dependent manner. On the contrary, H9C2 cardiomyoblast proliferation could be
272
elevated with TBX3 overexpression. Similar to the results of CCK8, the fluorescence
273
targeting the proliferation markers Ki67 was whittled with TBX3 silence while
274
enhanced with TBX3 overexpression in H9C2 cardiomyoblasts (Fig. 1B). The above
275
results indicated that TBX3 silence accelerated proliferation deficiency in H9C2
276
cardiomyoblasts.
277
3.2 Effect of TBX3 on cell cycle arrest in H9C2 cardiomyoblasts
278
To investigate whether TBX3 expression was involved in cell cycle arrest, we
279
analyzed the distribution of H9C2 cardiomyoblast cell cycle following TBX3
280
expression interference. As exhibited in Fig. 2A, compared with the control group,
281
there was a significant increase in the percentage of H9C2 cardiomyoblasts at G1
282
phase (p < 0.05) while an obvious decrease at S phase (p < 0.05) after TBX3 specific
283
siRNA treatment. However, TBX3 overexpression did not markedly affected cell
284
cycle distribution in H9C2 cardiomyoblasts. In addition, TBX3 silence inhibited the
285
expression of cyclin protein family, including cyclin D1, cyclin A and cyclin B1 (Fig.
286
2B-D, p < 0.05). Interestingly, TBX3 overexpression could aggrandize cyclin protein
287
expression while did not regulate cell cycle in H9C2 cardiomyoblasts. The data
288
indicated that TBX3 knockdown accelerated cell cycle arrest at S phase in H9C2
289
cardiomyoblasts.
290
3.3 Effect of TBX3 on apoptosis in H9C2 cardiomyoblasts
291
Since apoptosis is a consequence of cell cycle arrest, we employed Hoechst
292
33342 staining to observe apoptosis morphology and Annexin V FITC/PI double
293
staining to quantify cell apoptosis rate in H9C2 cardiomyoblasts after TBX3
294
expression interference. As shown in Fig. 3A, TBX3 siRNA-treated H9C2
295
cardiomyoblasts exhibited typical morphological characteristics of apoptosis,
296
including nuclear fragmentation and chromatin condensation. Afterwards, the
297
percentage of apoptotic H9C2 cardiomyoblasts was assessed by Annexin V FITC/PI
298
double staining and the data showed that TBX3 silence dramatically induced
299
apoptosis in H9C2 cardiomyoblasts (Fig. 3B-C, p < 0.05). Subsequently, western blot
300
was also conducted to investigate the levels of Apoptosis related proteins, such as
301
cleaved caspase-9, cleaved caspase-3, cleaved PARP, Bcl-2 and Bax. The results
302
demonstrated
303
PARP and Bax, and a decrease in Bcl-2 after TBX3 knockdown in H9C2
304
cardiomyoblasts (Fig. 3D-H, p < 0.05). However, there was no significant change in
305
apoptotic state of TBX3-overexpressing H9C2 cardiomyoblasts. The results indicated
306
that TBX3 knockdown was involved in H9C2 cardiomyoblast apoptosis.
307
3.4 Effect of TBX3 on mitochondrial function in H9C2 cardiomyoblasts
an increase in the levels of cleaved caspase-9, cleaved caspase-3, cleaved
308
To further examine the effects of TBX3 expression on mitochondrial function in
309
H9C2 cardiomyoblasts, we first detected the alterations of mitochondrial membrane
310
potential by utilizing the JC-1 fluorescent dye. As shown in Fig. 4A, the ratio between
311
the red and green fluorescence in H9C2 cardiomyoblasts loaded with JC-1 staining
312
was significantly down-regulated after TBX3 knockdown (p < 0.05). Then
313
mitochondrial ATPase activity and ROS generation were also analyzed. The results
314
showed that mitochondrial ATPase activity was down-regulated (Fig. 4B, p < 0.05)
315
and ROS generation was up-regulated (Fig. 4C, p < 0.05) in H9C2 cardiomyoblasts
316
with TBX3 knockdown, which indicated the existence of mitochondrial dysfunction.
317
Besides, as exhibited in Fig. 4D-E, the cytochrome C content in the cytoplasm was
318
prominently increased while decreased in the mitochondria after TBX3 siRNA
319
treatment, indicating TBX3 knockdown accelerated cytochrome C release from the
320
mitochondria into the cytoplasm in H9C2 cardiomyoblasts. The results indicated that
321
TBX3 knockdown induced mitochondrial disorder in in H9C2 cardiomyoblasts.
322
3.5 Effect of TBX3 on p21 expression in H9C2 cardiomyoblasts
323
To investigate the underlying mechanism of TBX3 expression on cell cycle and
324
apoptosis, we detected cell cycle regulatory protein P21 levels in H9C2
325
cardiomyoblasts after TBX3 expression interference. As shown in Fig. 5A-B, both the
326
mRNA and protein levels of P21 were increased in H9C2 cardiomyoblasts with TBX3
327
knockdown (p < 0.05). Moreover, the interaction between TBX3 protein and P21
328
DNA was verified by ChIP assay (Fig. 5C). The results indicated that P21 gene
329
expression was directly regulated by TBX3 protein.
330
3.6 Effect of TBX3/p21 pathway on proliferation in H9C2 cardiomyoblasts
331
To verify the effect of TBX3/p21 pathway on proliferation in H9C2
332
cardiomyoblasts, TBX3 siRNA and p21 siRNA were co-transfected into H9C2
333
cardiomyoblasts. As shown in Fig. 6A, P21 expression was obviously suppressed
334
after the co-transfection compared with TBX3 siRNA transfection alone (p < 0.05).
335
Additionally, the decreased proliferation capacity induced by TBX3 silence could be
336
observably promoted by TBX3 siRNA and p21 siRNA co-transfection in H9C2
337
cardiomyoblasts (Fig. 6B), manifesting that TBX3/p21 pathway might participate in
338
the regulation of proliferation in H9C2 cardiomyoblasts.
339
3.7 TBX3 expression in CHD samples
340
Finally, we examined the mRNA and protein expression of TBX3 in the hearts of
341
CHD donors to verify its potential in CHD treatment. As shown in Fig. 7A and B, the
342
mRNA and protein expression levels of TBX3 were obviously decreased in donors
343
with sporadic type CHD compared with the control ones (p < 0.05), indicating the
344
importance of TBX3 in CHD treatment.
345
4. Discussion
346
Pathological cardiomyoblast loss is a major histopathological hallmark of CHD,
347
but the accurate molecular mechanisms still need future investigation. In the present
348
study, we investigated whether TBX3 expression deficiency was the key point of
349
accelerating mitochondrial dysfunction, cell cycle arrest and apoptosis in aged H9C2
350
cardiomyoblasts. We found that TBX3 silence by specific siRNA inhibited
351
proliferation and blocked G1/S phase transition in aged H9C2 cardiomyoblasts. TBX3
352
knockdown up-regulated aged H9C2 cardiomyoblast apoptosis accompanied by
353
mitochondrial dysfunction. Besides, the apoptosis induction of TBX3 deletion in aged
354
H9C2 cardiomyoblasts could be abolished by P21 knockdown. Interestingly,
355
exorbitant proliferate was not observed in H9C2 cardiomyoblast with TBX3
356
overexpression. Hence, our results manifested that TBX3 was an important mediator
357
that regulated proliferation and apoptosis by targeting P21 in aged H9C2
358
cardiomyoblasts.
359
TBX3 is a critical member of the highly conserved T-box family of transcription
360
factors that possess explicit regulatory role on development in vertebrates [28].
361
Moreover, TBX3 involves in cell fate decision, cell cycle regulation, organ
362
development and even oncogenesis [29]. In addition, a strong relationship between
363
TBX3 insufficiency and congenital developmental disorders has been reported in the
364
literature [30]. As mentioned in the aforementioned literature, TBX3 variants could be
365
observed in patients with conotruncal heart defects, a heterogeneous congenital heart
366
malformation [18]. In our study, we found TBX3 deficiency in the hearts of donors
367
with sporadic types of CHD, which indicated its clinical importance in CHD
368
treatment. Afterwards, in order to further verify the effects of TBX3 expression on the
369
cell proliferative potential in aged H9C2 cardiomyoblasts, we employed TBX3
370
specific siRNA to silence TBX3 and TBX3-expressing lentivirus to increase TBX3 in
371
H9C2 cells at passage 10 to 15. After the corresponding treatment, the mRNA and
372
protein levels of TBX3 were respectively up-regulated and down-regulated in H9C2
373
cardiomyoblasts (Supplement Fig. S1-2). Besides, the inhibiting effects of TBX3
374
silence did not appear until 48 h after TBX3 specific siRNA employment. Similarly,
375
the effects of promoting growth were detected 48 h after TBX3-expressing lentivirus
376
transduction. It is well understood that Ki67 protein expressed at all stages of the cell
377
cycle other than resting phase is an approved cellular marker labeling proliferative
378
potential in several cell types [31]. Hence, we utilized immunofluorescence targeting
379
Ki67 to verify cellular proliferation in aged H9C2 cardiomyoblasts 48 h after
380
treatment and the results were in line with CCK8 analysis. Based on these data, we
381
confirmed that the abnormal expression of TBX3 was involved in the regulation of
382
proliferation in aged H9C2 cardiomyoblasts.
383
It is well accepted that transition from G1 phase to S phase and G2 phase to M
384
phase are the two critical stage of the cell cycle. In addition, cell growth can be
385
inhibited, if the cell cycle is blocked at any phase. Prior studies that have suggested
386
the importance of TBX3 expression on inducing cell cycle arrest through blocking
387
G1/S transition [19]. Subsequently, the effects of TBX3 expression on the distribution
388
of cell cycle in H9C2 cardiomyoblasts were investigated by using flow cytometry. We
389
also observed that the percentage of aged H9C2 cardiomyoblast at G1 phase was
390
increased while decreased in S phase, indicating the diminished DNA synthesis after
391
TBX3 knockdown. Since cell cycle was triggered by CDKs and their regulatory
392
subunit cyclins, we found cyclin D1, cyclin A and cyclin B1 levels were decreased in
393
TBX3 siRNA-treated H9C2 cardiomyoblasts. Wondrously, TBX3 knockdown
394
inhibited cell proliferation and triggered cell cycle arrest, but TBX3 over-expression
395
is ineffective for the cell cycle in aged H9C2 cardiomyoblasts, the mechanism of
396
which needed further investigation.
397
Since cell cycle arrest is an induction factor of apoptosis, which is considered to
398
participate in pathological changes of diverse cardiac disorders, such as myocarditis
399
and cardiomyopathy [7]. Although apoptosis is occurred at base levels under the
400
physiological circumstance, excessive cardiomyoblast apoptosis can be detected in
401
CHD patients with volume overload [32]. Recently, inhibiting apoptosis and elevating
402
survival of cardiomyoblasts have been proposed as the promising treatment strategies
403
targeting CHD. Based on the above, we tried to evaluate changes in apoptosis
404
morphology, apoptosis index and apoptosis-related protein expression in modified
405
H9C2 cardiomyoblasts. The data of current study demonstrated that apoptosis was
406
promoted after TBX3 silence in aged H9C2 cardiomyoblasts. Moreover,
407
mitochondrial dysfunction characterized by decreased mitochondrial membrane
408
potential, suppressed ATPase activity and elevated ROS generation was also detected
409
in TBX3-silenced H9C2 cardiomyoblasts. Since apoptosis could be affected by
410
multifarious intracellular transduction cascades, we only explored the pathway
411
associated with cell cycle arrest. As an inhibitor of CDKs, P21 is known to possess
412
multiple biological functions, such as regulating cell cycle, differentiation and
413
apoptosis [33]. Nonetheless, the effect of P21 on apoptosis is bidirectional. This is
414
mainly because P21 expression induces cell cycle blockage to repair DNA damage
415
when the cell suffers mild DNA lesions, whereas P21 triggers apoptotic response once
416
the DNA damage exceeds the repair ability [34]. In order to investigate the role of P21
417
in modified H9C2 cardiomyoblasts, we co-transfected TBX3 siRNA and P21 siRNA
418
into aged H9C2 cardiomyoblasts. In our study, P21 silence abolished the
419
apoptosis-inducing effects of TBX3 deficiency. Combined the CHIP assay, we found
420
P21 was the direct downstream signaling molecule of TBX3. Based on the data above,
421
we summarized TBX3/P21 signaling was involved in apoptotic occurrence in aged
422
H9C2 cardiomyoblasts.
423
H9C2 cardiomyoblasts at passage 10 to 15 were employed in the present study.
424
Prior studies have suggested that morphology changes and sensitivity to toxicity are
425
increased with the number of passages in H9C2 cardiomyoblasts [35]. However, the
426
literature results also indicated that the influence of cell line passage number on the
427
reliability of study results was intricate and depended on multi-factor, such as culture
428
conditions [35]. Hence, we will investigate the effect of TBX3 deficiency on
429
apoptosis in cardiomyoblasts at different passage in the future.
430
In conclusion, the present study showed that TBX3 expression deficiency
431
accelerated apoptosis in aged cardiomyoblasts, which was accompanied by
432
mitochondrial dysfunction, cell cycle arrest, growth suppression and apoptosis.
433
Although more research should be implemented to verify the accurate mechanism of
434
TBX3 expression on cardiomyoblast function, our work suggests TBX3 maybe one of
435
the potential therapeutic targets of CHD.
436
Acknowledgement
437
This study was supported by grants from the National Natural Science
438
Foundation of China (No. 81300130) and the Basic Scientific Research Projects of
439
Liaoning Higher Education Institutions (No. LFWK201701).
440
References
441
1.
442
BJM. The changing epidemiology of congenital heart disease. Nature Reviews
443
Cardiology. 2011;8(1):50–60.
444
2.
445
Epidemiological study of congenital heart defects in children and adolescents:
446
analysis of 4,538 cases. Arq Bras Cardiol. 2003;80(3): 269-278.
Van Der Bom T, Zomer AC, Zwinderman AH, Meijboom FJ, Bouma BJ, Mulder
Miyague NI, Cardoso SM, Meyer F, Ultramari FT, Araújo FH, Rozkowisk I, et al.
447
3.
Zhou Y, Jia WK, Jian Z, Zhao L, Liu CC, Wang Y, et al. Downregulation of
448
microRNA-199a-5p protects cardiomyoblasts in cyanotic congenital heart disease by
449
attenuating endoplasmic reticulum stress. Mol Med Rep. 2017;16(3):2992–3000.
450
4.
451
Pediatr Cardiol. 1995;16(4):155–165.
452
5.
453
Cardiol. 2002;39(12):1890–900.
454
6.
455
of graded hypoxia on the embryonic chick heart. Teratology. 1985;32(3):463–472.
456
7.
457
2000;45(3):603–620.
458
8.
459
might play a role in the onset of congenital heart disease by inducing cardiomyocyte
460
apoptosis, Circ. J. 74 (2010) 2410–2418.
461
9.
462
congenital heart disease by suppressing HES1, Eur. J. Pharmacol. 836 (2018) 18–24.
463
10. L. Wang, D. Tian, J. Hu, H. Xing, M. Sun, J. Wang, Q. Jian, H. Yang,
464
MiRNA-145 Regulates the Development of Congenital Heart Disease Through
465
Targeting FXN, Pediatr. Cardiol. 37 (2016) 629–636.
466
11. Sarangapani KK. A, Asbury CL. Trends Genet. Trends Genet. 2012;37(1):62–70.
467
12. Carlson H, Ota S, Song Y, Chen Y, Hurlin PJ. Tbx3 impinges on the p53 pathway
468
to suppress apoptosis, facilitate cell transformation and block
469
differentiation. Oncogene. 2002;21(24):3827–3835.
Hoffman JIE. Incidence of Congenital Heart Disease: II. Prenatal Incidence J.I.E.
Hoffman JIE, Kaplan S. The incidence of congenital heart disease. J Am Coll
Ruckman RN, Rosenquist GC, Rademaker DA, Morse DE, Getson PR. The effect
van den Hoff M. Programmed cell death in the developing heart. Cardiovasc Res.
H.K. Jiang, G.R. Qiu, J. Li-Ling, N. Xin, K.L. Sun, Reduced ACTC1 expression
Y. Zhang, B. Peng, Y. Han, MiR-182 alleviates the development of cyanotic
myogenic
470
13. Willmer T, Cooper A, Peres J, Omar R, Prince S. The T-Box transcription factor 3
471
in development and cancer. BioScience Trends. 2017;11(3):254–266.
472
14. Renard CA, Labalette C, Armengol C, Cougot D, Wei Y, Cairo S, et al. Tbx3 is a
473
downstream target of the Wnt/β-catenin pathway and a critical mediator of β-catenin
474
survival functions in liver cancer. Cancer Res. 2007;67(3):901–910.
475
15. Miao ZF, Liu XY, Xu HM, Wang ZN, Zhao TT, Song YX, et al. Tbx3
476
overexpression in human gastric cancer is correlated with advanced tumor stage and
477
nodal status and promotes cancer cell growth and invasion. Virchows Arch.
478
2016;469(5):505–513.
479
16. Aliwaini S, Lubbad A, Shourfa A, Hamada H, Ayesh B, Abu Tayem H, et al.
480
Overexpression of TBX3 transcription factor as a potential diagnostic marker for
481
breast cancer. Mol Clin Oncol. 2019; 10(1): 105-112.
482
17. Perkhofer L, Walter K, Costa IG, Carrasco MCR, Eiseler T, Hafner S, et al. Tbx3
483
fosters
484
activin/nodal-dependent induction of stemness. Stem Cell Res. 2016;17(2):367–378.
485
18. Xie H, Zhang E, Hong N, Fu Q, Li F, Chen S, et al. Identification of TBX2 and
486
TBX3 variants in patients with conotruncal heart defects by target sequencing. Hum
487
Genomics. 2018;12(1):44.
488
19. Knaapen MWM, Davies MJ, De Bie M, Haven AJ, Martinet W, Kockx MM.
489
Apoptotic versus autophagic cell death in heart failure. Cardiovasc Res.
490
2001;51(2):304–312.
491
20. Roninson IB. Oncogenic functions of tumour suppressor p21Waf1/Cip1/Sdi1:
492
Association with cell senescence and tumour-promoting activities of stromal
493
fibroblasts. Cancer Letters. 2002;179(1):1–14.
pancreatic
cancer
growth
by
increased
angiogenesis
and
494
21. Yagi A, Hasegawa Y, Xiao H, Haneda M, Kojima E, Nishikimi A, et al. GADD34
495
Induces p53 Phosphorylation and p21/WAF1 Transcription. J Cell Biochem.
496
2003;90(6):1242–1249.
497
22. Lee EW, Lee MS, Camus S, Ghim J, Yang MR, Oh W, et al. Differential
498
regulation of p53 and p21 by MKRN1 E3 ligase controls cell cycle arrest and
499
apoptosis. EMBO J. 2009;28(14):2100–2113.
500
23. Wang Y, Fisher JC, Mathew R, Ou L, Otieno S, Sublet J, et al. Intrinsic disorder
501
mediates the diverse regulatory functions of the Cdk inhibitor p21. Nat Chem Biol.
502
2011;7(4):214–221.
503
24. Zhang H, Menzies KJ, Auwerx J. The role of mitochondria in stem cell fate and
504
aging. Development. 2018;145(8):dev143420.
505
25. Clavier A, Rincheval-Arnold A, Colin J, Mignotte B, Guénal I. Apoptosis in
506
Drosophila: Which role for mitochondria? Apoptosis. 2016;21(3):239–251.
507
26. Liang S, Sun K, Wang Y, Dong S, Wang C, Liu LX, et al. Role of
508
Cyt-C/caspases-9,3, Bax/Bcl-2 and the FAS death receptor pathway in apoptosis
509
induced by zinc oxide nanoparticles in human aortic endothelial cells and the
510
protective effect by alpha-lipoic acid. Chem Biol Interact. 2016;258:40–51.
511
27. Ruffolo SC, Breckenridge DG, Nguyen M, Goping IS, Gross A, Korsmeyer SJ, et
512
al. BID-dependent and BID-independent pathways for BAX insertion into
513
mitochondria. Cell Death Differ. 2000;7(11):1101–1108.
514
28. Papaioannou VE. T-box genes in development: From hydra to humans. Int Rev
515
Cytol. 2001;207:1–70.
516
29. Wansleben S, Peres J, Hare S, Goding CR, Prince S. T-box transcription factors in
517
cancer
biology.
Biochimica
et
Biophysica
Acta
-
Reviews
on
Cancer.
518
2014;1846(2):380–391.
519
30. Packham
520
2003;12(90001):37R–44.
521
31. Sundara Rajan S, Hanby AM, Horgan K, Thygesen HH, Speirs V. The potential
522
utility of geminin as a predictive biomarker in breast cancer. Breast Cancer Res Treat.
523
2014;143(1):91–98.
524
32. Kajstura J, Mansukhani M, Cheng W, Reiss K, Krajewski S, Reed JC, et al.
525
Programmedcell death and expression of the protooncogene bcl-2 in myocytes during
526
postnatal maturation of the heart. Exp Cell Res. 1995;219(1):110–121.
527
33. Blundell RA. The Biology of p21Waf1/Cip1 - Review Paper. Am J Biochem
528
Biotechnol. 2009;2(1):33–40.
529
34. Jackson JG, Pant V, Li Q, Chang LL, Quintás-Cardama A, Garza D, et al.
530
P53-Mediated Senescence Impairs the Apoptotic Response to Chemotherapy and
531
Clinical Outcome in Breast Cancer. Cancer Cell. 2012;21(6):793–806.
532
35. P. Witek, A. Korga, F. Burdan, M. Ostrowska, B. Nosowska, M. Iwan, J. Dudka,
533
The effect of a number of H9C2 rat cardiomyocytes passage on repeatability of
534
cytotoxicity study results, Cytotechnology. 68 (2016) 2407–2415.
535
Disclosure: The authors declared no conflict of interest.
536 537 538 539 540
EA.
T-box
genes
in
human
disorders.
Hum
Mol
Genet.
541
Figures and legends
542
Figure.1
543
544
Fig. 1. Effect of TBX3 on proliferation in H9C2 cardiomyoblasts. (A) CCK-8 was
545
used to evaluate the effect of TBX3 expression on viability in H9C2 cardiomyoblasts.
546
(B) Immunofluorescence targeting Ki67 in treated H9C2 cardiomyoblasts. Data were
547
represented as mean ± SD at least three independent experiments. The data from three
548
or more groups were analyzed by one-way analysis of variance (ANOVA) followed
549
by Tukey’s multiple comparison test.
550
551
552
553
554
555
556
557
558
Figure.2
559
560
Fig. 2. Effect of TBX3 on cell cycle arrest in H9C2 cardiomyoblasts. (A) Flow
561
cytometry was used to analyze cell cycle distribution in H9C2 cardiomyoblasts.
562
Representative western blot for (B) Cyclin D1, (C) Cyclin A and (D) Cyclin B1 in
563
H9C2 cardiomyoblasts. Data were represented as mean ± SD at least three
564
independent experiments. The data from three or more groups were analyzed by
565
one-way analysis of variance (ANOVA) followed by Tukey’s multiple comparison
566
test.
567
568
569
Figure.3
570 571
Fig. 3. Effect of TBX3 on apoptosis in H9C2 cardiomyoblasts. (A) Representative
572
images of Hoechst 33342 staining at 400× magnification in H9C2 cardiomyoblasts.
573
(B) Annexin V FITC/PI double staining and (C) apoptosis rate of H9C2
574
cardiomyoblasts were detected by using a flow cytometer. Representative western blot
575
for (D) cleaved caspase-9, (E) cleaved caspase-3 and (F) Bcl-2, (G) Bax and (H)
576
cleaved PARP in H9C2 cardiomyoblasts. Data were represented as mean ± SD at least
577
three independent experiments. The data from three or more groups were analyzed by
578
one-way analysis of variance (ANOVA) followed by Tukey’s multiple comparison
579
test.
580
581
582
583
584
585
586
587
588
589
590
591
592
593
594
Figure.4
595
596
Fig. 4. Effect of TBX3 on mitochondrial function in H9C2 cardiomyoblasts. (A) The
597
alteration of mitochondrial membrane potential in H9C2 cardiomyoblasts. (B) The
598
activity of ATPase in H9C2 cardiomyoblasts. (C) Fluorescence intensity of ROS in
599
H9C2 cardiomyoblasts. Representative western blot for (D) cytochrome C in the
600
cytoplasm and (E) cytochrome C in the mitochondria of H9C2 cardiomyoblasts. The
601
data from three or more groups were analyzed by one-way analysis of variance
602
(ANOVA) followed by Tukey’s multiple comparison test.
603
604
605
606
Figure.5
607
608
Fig. 5. Effect of TBX3 on p21 expression in H9C2 cardiomyoblasts. The relative (A)
609
mRNA and (B) protein levels of P21 in H9C2 cardiomyoblasts. (C) ChIP was used to
610
verify the targeted combination between TBX3 and p21 in H9C2 cardiomyoblasts.
611
Negative control: control IgG; Positive control: anti-RNA Polymerase II;
612
PCR-negative control: distilled H2O; RT-Input 1: P21; RT-Input 2: GAPDH. The data
613
from three or more groups were analyzed by one-way analysis of variance (ANOVA)
614
followed by Tukey’s multiple comparison test.
615
616
617
618
619
620
621
Figure.6
622
623
Fig. 6. Effect of TBX3/p21 pathway on proliferation in H9C2 cardiomyoblasts. (A)
624
The relative protein levels of P21 in H9C2 cardiomyoblasts. (B) Immunofluorescence
625
targeting Ki67 in H9C2 cardiomyoblasts. The data from three or more groups were
626
analyzed by one-way analysis of variance (ANOVA) followed by Tukey’s multiple
627
comparison test.
628
629
630
631
632
633
634
635
Figure.7
636
637
Fig. 7. TBX3 expression in CHD samples. TBX3 mRNA (A) and protein (B) levels
638
in the heart tissues of CHD donors. The data from two groups was determined by t
639
test.
640
641
642
643
644
645
646
647
648
Supplement Figure. 1
649 650
Supplement Fig. 1. Silencing TBX3 expression by specific siRNA in H9C2
651
cardiomyoblasts. (A) The mRNA expression levels of TBX3 after TBX3 siRNA
652
transfection in H9C2 cardiomyoblasts. (B) The protein expression levels of TBX3
653
after TBX3 siRNA transfection in H9C2 cardiomyoblasts. *p< 0.05 vs. control siRNA
654
group
655
656
657
658
659
660
661
662
Supplement Figure. 2
663
664
Supplement Fig. 2. Elevating TBX3 expression by lentivirus in H9C2
665
cardiomyoblasts. (A) The mRNA expression levels of TBX3 after TBX3-expressing
666
lentivirus transduction in H9C2 cardiomyoblasts. (B) The protein expression levels of
667
TBX3 after TBX3-expressing lentivirus transduction. *p< 0.05 vs. control lentivirus
668
group.
Disclosure: The authors declared no conflict of interest.