Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Keqiong Tang is active.

Publication


Featured researches published by Keqiong Tang.


Reproductive Toxicology | 2017

Apoptosis inducing factor gene depletion inhibits zearalenone-induced cell death in a goat Leydig cell line

Diqi Yang; Tingting Jiang; Pengfei Lin; Huatao Chen; Lei Wang; Nan Wang; Fan Zhao; Keqiong Tang; Dong Zhou; Aihua Wang; Yaping Jin

Zearalenone (ZEA) is a contaminant of human food and animal feedstuffs that causes health hazards. However, the signal pathways underlying ZEA toxicity remain elusive. The aims of this study were to determine which pathways are involved in ZEA-induced cell death and investigate the effect of apoptosis inducing factor (AIF) on cell death during ZEA treatment in the immortalized goat Leydig cell line hTERT-GLC. This study showed that ZEA-induced cell death in hTERT-GLCs works via endoplasmic reticulum (ER) stress, the caspase-dependent pathway, the caspase-independent pathway and autophagy. Recombinant lentiviral vectors were constructed to silence AIF expression in hTERT-GLCs. Flow cytometry results showed that knockdown of AIF diminished ZEA-induced cell apoptosis in hTERT-GLCs. Furthermore, we found AIF depletion down-regulated phosphoIRE1α, GRP78, CHOP and promoted the switch of LC3-I to LC3-II. Therefore, ZEA induces cytotoxicity in hTERT-GLCs via different pathways, while AIF-mediated signaling plays a critical role in ZEA-induced cell death in hTERT-GLCs.


Animal Science Journal | 2016

Establishment and evaluation of a stable steroidogenic goat Leydig cell line.

Jinhua Zhou; Rui Dai; Lanjie Lei; Pengfei Lin; Xiaolong Lu; Xiangguo Wang; Keqiong Tang; Aihua Wang; Yaping Jin

Leydig cells play a key role in synthesizing androgen and regulating spermatogenesis. The dysfunction of Leydig cells may lead to various male diseases. Although primary Leydig cell cultures have been used, their finite lifespan hinders the assessment of long-term effects. In the present study, primary goat Leydig cells (GLCs) were immortalized via the transfection of a plasmid containing the human telomerase reverse transcriptase (hTERT) gene. The expressions of hTERT and telomerase activity were evaluated in transduced GLCs (hTERT-GLCs). These cells steadily expressed the hTERT gene and exhibited longer telomere lengths at passage 55 that were similar to those of HeLa cells. The hTERT-GLCs at passages 30 and 50 expressed genes that encoded key proteins, enzymes and receptors that are inherent to normal Leydig cells, for example, steroidogenic acute regulatory protein (StAR), cytochrome P450 cholesterol side-chain cleavage enzyme (P450scc), 3β-hydroxysteroid dehydrogenase (3β-HSD) and LH-receptor (LH-R). Additionally, the immortalized goat Leydig cells secreted detectable quantities of testosterone in response to hCG stimulation. Furthermore, this cell line appeared to proliferate more quickly than the control cells, although no neoplastic transformation occurred in vitro. We concluded that the GLCs immortalized with hTERT retained their original characteristics and might provide a useful model for the study of Leydig cell function.


Journal of Reproduction and Development | 2017

An immortalized steroidogenic goat granulosa cell line as a model system to study the effect of the endoplasmic reticulum (ER)-stress response on steroidogenesis

Diqi Yang; Lei Wang; Pengfei Lin; Tingting Jiang; Nan Wang; Fan Zhao; Huatao Chen; Keqiong Tang; Dong Zhou; Aihua Wang; Yaping Jin

With granulosa and theca cells, the ovaries are responsible for producing oocytes and secreting sex steroids such as estrogen and progesterone. Endoplasmic reticulum stress (ERS) plays an important role in follicle atresia and embryo implantation. In this study, goat granulosa cells were isolated from medium-sized (4–6 mm) healthy follicles. Primary granulosa cells were immortalized by transfection with human telomerase reverse transcriptase (hTERT) to establish a goat granulosa cell line (hTERT-GGCs). These hTERT-GGCs expressed hTERT and had relatively long telomeres at passage 50. Furthermore, hTERT-GGCs expressed the gonadotropin receptor genes CYP11A1, StAR, and CYP19A1, which are involved in steroidogenesis. Additionally, progesterone was detectable in hTERT-GGCs. Although the proliferation potential of hTERT-GGCs significantly improved, there was no evidence to suggest that the hTERT-GGCs are tumorigenic. In addition, thapsigargin (Tg) treatment led to a significant dose-dependent decrease in progesterone concentration and steroidogenic enzyme expression. In summary, we successfully generated a stable goat granulosa cell line. We found that Tg induced ERS in hTERT-GGCs, which reduced progesterone production and steroidogenic enzyme expression. Future studies may benefit from using this cell line as a model to explore the molecular mechanisms regulating steroidogenesis and apoptosis in goat granulosa cells.


International Journal of Molecular Sciences | 2017

Knockdown of XBP1 by RNAi in Mouse Granulosa Cells Promotes Apoptosis, Inhibits Cell Cycle, and Decreases Estradiol Synthesis

Nan Wang; Fan Zhao; Pengfei Lin; Guangle Zhang; Keqiong Tang; Aihua Wang; Yaping Jin

Granulosa cells are crucial for follicular growth, development, and follicular atresia. X-box binding protein 1 (XBP1), a basic region-leucine zipper protein, is widely involved in cell differentiation, proliferation, apoptosis, cellular stress response, and other signaling pathways. In this study, RNA interference, flow cytometry, western blot, real-time PCR, Cell Counting Kit (CCK8), and ELISA were used to investigate the effect of XBP1 on steroidogenesis, apoptosis, cell cycle, and proliferation of mouse granulosa cells. ELISA analysis showed that XBP1 depletion significantly decreased the concentrations of estradiol (E2). Additionally, the expression of estrogen synthesis enzyme Cyp19a1 was sharply downregulated. Moreover, flow cytometry showed that knockdown of XBP1 increased the apoptosis rate and arrests the cell cycle in S-phase in granulosa cells (GCs). Further study confirmed these results. The expression of CCAAT-enhancer-binding protein homologous protein (CHOP), cysteinyl aspartate specific proteases-3 (caspase-3), cleaved caspase-3, and Cyclin E was upregulated, while that of Bcl-2, Cyclin A1, and Cyclin B1 was downregulated. Simultaneously, CCK8 analysis indicated that XBP1 disruption inhibited cell proliferation. In addition, XBP1 knockdown also alters the expression of Has2 and Ptgs2, two essential genes for folliculogenesis. Collectively, these data reveal a novel critical role of XBP1 in folliculogenesis by regulating the cell cycle, apoptosis, and steroid synthesis of mouse granulosa cells.


Journal of Reproduction and Development | 2016

Herp depletion arrests the S phase of the cell cycle and increases estradiol synthesis in mouse granulosa cells.

Fenglei Chen; Nan Wang; Diqi Yang; Xin Wen; Tagwa Norain Mahmoud; Dong Zhou; Keqiong Tang; Pengfei Lin; Aihua Wang; Yaping Jin

The endoplasmic reticulum (ER) stress response has been implicated in the development, atresia and luteinization of ovarian follicles. However, there have been few reports concerning the role of Herp, an ER stress-induced protein, in follicular development. The present study aims to detect the distribution and cyclic variations of Herp during the estrous cycle and to reveal the roles of Herp in regulating the cell cycle, apoptosis and steroid hormone biosynthesis in mouse granulosa cells. In this study, immunohistochemistry staining showed that Herp expression was primarily in the granulosa cells and oocytes. Furthermore, we constructed recombinant lentiviral vectors for Herp short hairpin interfering RNA (shRNA) expression; immunofluorescence staining, real-time quantitative PCR (RT-qPCR) and western blot analysis revealed that Herp was successfully knocked down. Flow cytometry showed that knockdown of Herp arrested granulosa cells at the S phase of the cell cycle. More importantly, ELISA analysis revealed that Herp knockdown significantly upregulated the concentration of estradiol (E2) in the culture supernatants. RT-qPCR was performed to determine the regulatory mechanism of Herp knockdown in the cell cycle, and in steroid synthesis, RT-qPCR analysis revealed that Herp knockdown upregulated the mRNA expression of steroidogenic enzymes (Cyp19a1) and downregulated metabolic enzymes (Cyp1b1) and cell cycle factors (cyclin A1, cyclin B1 and cyclin D2). These results suggest that Herp may regulate the cell cycle and hormone secretions in mouse granulosa cells. The present study helps to elucidate the physiological functions of Herp as they relate to reproduction.


PLOS ONE | 2016

Knockdown of CREB3/Luman by shRNA in Mouse Granulosa Cells Results in Decreased Estradiol and Progesterone Synthesis and Promotes Cell Proliferation.

Fan Zhao; Nan Wang; Yanglei Yi; Pengfei Lin; Keqiong Tang; Aihua Wang; Yaping Jin

Luman (also known as LZIP or CREB3) is a transcription factor and a member of the cAMP responsive element-binding (CREB) family proteins. Although Luman has been detected in apoptotic granulosa cells and disorganized atretic bodies, the physiological function of Luman in follicular development has not been reported. Our objective is to determine the role of Luman in folliculogenesis by knocking down Luman expression in mouse GCs (granulosa cells) using shRNA. Luman expression was successfully knocked down in mouse GCs at the mRNA and protein level, as confirmed by real-time quantitative PCR, western blot and immunofluorescence staining, respectively. Knockdown of Luman significantly decreased the concentrations of estradiol (E2) and progesterone (P4) in cell culture medium. Furthermore, Luman knockdown promoted cell proliferation but had no effect on cell apoptosis. To elucidate the regulatory mechanism underlying the effects of Luman knockdown on steroid synthesis and cell cycle, we measured the mRNA and protein expression levels of several related genes. The expression of Star, Cyp19a1, and Cyp1b1, which encode steroidogenic enzymes, was down-regulated, while that of Cyp11a1 and Runx2, which also encode steroidogenic enzymes, was up-regulated. The expression of the cell cycle factors Cyclin A1, Cyclin B1, Cyclin D2, and Cyclin E was significantly up-regulated. Among apoptosis-related genes, only Bcl-2 was down-regulated, while Caspase 3, Bax and p53 were not significantly affected, suggesting that Luman knockdown may regulate cell cycle activity and hormone secretion at the transcriptional and translational level in mouse GCs. The expression of two important genes associated with folliculogenesis in mouse GCs, Has2 and Ptgs2, were also significantly altered by Luman knockdown. In conclusion, the findings of this study indicate that Luman regulates mouse GCs modulation of steroid synthesis, cell cycle activity and other regulators of folliculogenesis.


Journal of Cellular Physiology | 2018

Hormone regulates endometrial function via cooperation of endoplasmic reticulum stress and mTOR-autophagy

Diqi Yang; Tingting Jiang; J. Liu; Jin Hong; Pengfei Lin; Huatao Chen; Dong Zhou; Keqiong Tang; Aihua Wang; Yaping Jin

In ruminant, the receptive endometrium and the elongation of the hatched blastocyst are required to complete the process of implantation. However, the mechanisms regulating goat endometrial function during the peri‐implantation period of pregnancy are still unclear. In this study, EECs were treated with progesterone, estradiol, and interferon‐tau (IFNT). We have found that endoplasmic reticulum (ER) stress was activated under hormones treatment. To identify the cellular mechanism of regulation of endometrial function, we investigated the effect of ER stress activator thapsigargin (TG) and inhibitor 4 phenyl butyric acid (4‐PBA) on EECs. We found that TG, which activated the three branches of UPR, increased the expression of genes associated with promoting conceptus elongation and cellular attachment, significantly up‐regulated the spheroid attachment rate and PGE2/PGF2α ratio. 4‐PBA pre‐treatment inhibited UPR and inhibited promoting conceptus elongation and cellular attachment related genes, but the spheroid attachment rate and PGE2/PGF2α ratio were not changed significantly. Moreover, knockdown of ATF6 via shATF6 promoted the conceptus elongation related genes, but increased the dissolution of the corpus luteum. Besides, blocking ATF6 attenuated autophagy by activating mammalian target of rapamycin (mTOR) pathway. Moreover, rapamycin (mTOR inhibitor) pre‐treatment inhibited the expression of promoting conceptus elongation and increased PGE2/PGF2α ratio. Taken together, our study indicated that physiological level of ER stress may contribute to early pregnancy success, and ATF6 signaling pathway cooperated with autophagy to regulate endometrial function by modulating mTOR pathway.


Biology of Reproduction | 2018

CREB3 regulatory factor -mTOR-autophagy regulates goat endometrial function during early pregnancy

Diqi Yang; Tingting Jiang; J. Liu; Beibei Zhang; Pengfei Lin; Huatao Chen; Dong Zhou; Keqiong Tang; Aihua Wang; Yaping Jin

Abstract In domestic ruminants, a receptive endometrium is crucial for successful pregnancy. Although many essential molecular modulators and pathways have been identified during early pregnancy, the precise mechanisms regulating goat endometrial function remains largely unknown. Here, we describe a novel regulator during early pregnancy, whereby hormones increased CREB3 regulatory factor (CREBRF) expression and act as a potential activator of autophagy in endometrial epithelial cells (EECs) via the mTOR pathway. Our results showed that knockdown of CREBRF via shCREBRF hampered EECs proliferation by S-phase cell cycle arrest and significantly inhibited endometrial function. We also reported that CREBRF-mTOR-autophagy pathway plays a vital role in regulating endometrial function, with a blockade of the mTOR by rapamycin demonstrating the regulatory function on prostaglandin (PGs) secretion and cell attachment in EECs. Moreover, chloroquine pretreatment also proved the above conclusion. Collectively, our findings provide new insight into the molecular mechanisms of goat endometrial function and indicate that the CREBRF-mTORautophagy pathway plays a central role in PGs secretion and cell attachment. Summary Sentence The protein CREB3 regulatory factor (CREBRF) acts as a potential activator of autophagy in endometrial epithelial cells via the mTOR pathway. Knockdown of CREBRF via shCREBRF hampered EECs proliferation by S-phase cell cycle arrest and significantly inhibited endometrial function. The CREBRF-mTOR-autophagy pathway plays a vital role in regulating endometrial function, with a blockade of the mTOR by rapamycin demonstrating the regulatory function on prostaglandins (PGs) secretion and cell attachment in EECs.


Reproduction in Domestic Animals | 2017

GDF9 affects the development and tight junction functions of immature bovine Sertoli cells

Keqiong Tang; Lei Wang; Yaping Jin; Wucai Yang; Liguo Yang

Transforming growth factor-β (TGFβ) superfamily are critical regulators of germ cell development that act as extracellular ligands of the signal transduction pathways regulating proliferation, apoptosis and other aspects of cell behaviour. As a member of the TGF-β superfamily, growth differentiation factor 9 (GDF9) plays a critical role in ovarian follicular development and the ovulation rate in females; however, its role in the testis has not been well elucidated. The aim of this study was to investigate the expression of GDF9 and its receptor genes BMPRII and ALK5 in prepuberal bovine Sertoli cells (SCs). In addition, we assessed the effects of GDF9 on immature SCs apoptosis, the cell cycle and tight junction functions. We found that GDF9 and its receptor genes BMPRII and ALK5 were expressed in immature SCs. Exogenous GDF9 significantly promoted SCs proliferation and inhibited the apoptosis of SCs by significantly upregulating Cyclin E (cell cycle) and bcl-2 (anti-apoptosis) mRNA expression and downregulating caspase-3 (pro-apoptosis) mRNA expression. Meanwhile, exogenous GDF9 significantly decreased the value of transepithelial electrical resistance by significantly downregulating claudin-11 mRNA expression and influencing the distribution of occludin. In conclusion, this study reveals that GDF9 is a key regulator of bovine SCs through the modulation of the cell cycle, apoptosis and tight junction functions.


Theriogenology | 2018

Interferon-τ regulates prostaglandin release in goat endometrial stromal cells via JAB1 - unfolded protein response pathway

Diqi Yang; Tingting Jiang; J. Liu; Jin Hong; Pengfei Lin; Huatao Chen; Dong Zhou; Keqiong Tang; Aihua Wang; Yaping Jin

Prostaglandins (PGs) are major products of the uterine endometrium, and they are critical for recognition of pregnancy in ruminants. During the peri-implantation period of pregnancy, interferon tau (IFN-τ) plays an important role in the regulation of endometrial PGs synthesis, but the underlying mechanisms remain poorly understood. In this work, the results demonstrated that IFN-τ increased the PGE2/PGF2α ratio, up-regulated the expression of JAB1 and activated the unfolded protein response (UPR). Knockdown of JAB1 reduced the PGE2/PGF2α ratio and inhibited the expression of UPR markers in endometrial stromal cells (ESCs) under IFN-τ treatment. Pre-treatment with endoplasmic reticulum (ER) stress activator thapsigargin (Tg) activated UPR and restored the PGE2/PGF2α ratio in shJAB1 groups under IFN-τ treatment. In conclusion, our results indicated that IFN-τ regulated the PGE2/PGF2α ratio via cooperation between JAB1 and UPR, and the reduction of JAB1 led to the down-regulation of the PGE2/PGF2α ratio, which inhibits UPR, and thus is harmful to early pregnancy. Activation of UPR could restore JAB1 reduction, resulting in a reduced PGE2/PGF2α ratio. These findings extend our understanding and may provide new insights into the mechanism of IFN-τ regulation of PG secretion in ESCs and the biological functions of JAB1 and UPR.

Collaboration


Dive into the Keqiong Tang's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge