Network


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

Hotspot


Dive into the research topics where Wenjing Wu is active.

Publication


Featured researches published by Wenjing Wu.


Gene | 2014

Identification, stability and expression of Sirt1 antisense long non-coding RNA.

Yu Wang; Weijun Pang; Ning Wei; Yan Xiong; Wenjing Wu; Cunzhen Zhao; Qing-Wu Shen; Gongshe Yang

Natural antisense transcripts (NATs) exist ubiquitously as pivotal molecules to regulate coding gene expression. Sirtuin 1 (Sirt1) is a NAD-dependent deacetylase which is involved in myogenesis. However, whether Sirt1 transcribes NAT during C2C12 differentiation is still unknown. In this study, we identified a Sirt1 NAT which was designated as Sirt1 antisense long non-coding RNA (AS lncRNA) by sequencing and bioinformatic analysis. The level of Sirt1 AS lncRNA was greater in spleen but less in muscle tissue. The expression of both Sirt1 mRNA and Sirt1 AS lncRNA decreased during C2C12 myogenic differentiation, whereas the levels of miR-34a, which targets Sirt1, increased gradually. We further found that the half-life of Sirt1 AS lncRNA was 10h, but that of Sirt1 mRNA was 6h in C2C12 cells treated with 2 μg/ml Actinomycin D. Therefore, compared with Sirt1 mRNA, Sirt1 AS lncRNA was more stable. Overexpression of Sirt1 AS lncRNA increased the levels of Sirt1 protein, whereas overexpression of Sirt1 AS lncRNA mutant did not affect the level of Sirt1 protein in C2C12 cells. Moreover, downregulation of Sirt1 mRNA caused by miR-34a was counteracted by Sirt1 AS lncRNA in C2C12 cells. Taken together, we identified a novel NAT of Sirt1 which implicated in myogenesis through regulating Sirt1 expression.


Cell Biology International | 2015

Knockdown of CTRP6 inhibits adipogenesis via lipogenic marker genes and Erk1/2 signalling pathway.

Wenjing Wu; Delin Mo; Cunzhen Zhao; Chen Zhao; Yaosheng Chen; Weijun Pang; Gongshe Yang

C1q/tumor necrosis factor‐related protein 6 (CTRP6), an adipose‐tissue secretory factor, plays an important role in inflammatory reaction and carcinogenesis. However, the biological function of CTRP6 in adipogenesis remains unclear. In this study, we examined the effects of CTRP6 knockdown on lipogenesis of 3T3‐L1 adipocytes. The results showed that after 3T3‐L1 adipocytes transfected with anti‐CTRP6 small interfering RNA (siRNA), not only levels of secreted CTRP6 protein in the culture medium but also the expression level of the CTRP6 protein in the 3T3‐L1 adipocytes was significantly reduced (P < 0.01). In addition, the number of lipid droplets in the adipocytes was reduced, as well as the OD values reflecting the fat content being significantly decreased (P < 0.01). Meanwhile the levels of adipogenic markers, including peroxisome proliferator activated receptor γ (PPARγ), CCAAT/enhancer‐binding protein α (C/EBPα), CCAAT/enhancer‐binding protein β (C/EBPβ) and adipocyte fatty acid‐binding protein 4 (aP2), were decreased after treatment with anti‐CTRP6 siRNA, whereas the expression of adipose triglyceride lipase (ATGL) and triacylglycerol hydrolase (TGH) were increased. Furthermore, after transfection, activity of phosphorylated Erk1/2 (p‐Erk1/2) was inhibited in the early stage of differentiation, but in terminal differentiation of adipocytes, its activity was activated. Taken together, the results indicate that knockdown of CTRP6 can inhibit adipogenesis of 3T3‐L1 adipocytes through lipogenic marker genes and Erk1/2 signaling pathway.


Journal of Animal Science | 2014

Obese and lean porcine difference of FoxO1 and its regulation through C/EBPβ and PI3K/GSK3β signaling pathway1

Weijun Pang; Ning Wei; Yu Wang; Yan Xiong; F. F. Chen; Wenjing Wu; Cunzhen Zhao; S. D. Sun; Gongshe Yang

Forkhead box O 1 (FoxO1) is an important transcription factor implicated in adipogenesis. In this study, we detected the breed differences in FoxO1 between Bamei pigs (an obese breed) and Large White pigs (a lean breed). Compared with Large White pigs, the BW of Bamei pigs was lower (P < 0.01), but back fat thickness, fat percent, and intramuscular fat content were greater (P < 0.01). The levels of FoxO1 mRNA and protein were lower (P < 0.01) in subcutaneous adipose tissue (SAT) of Bamei pigs at 180 d, adipocytes and stromal-vascular fraction extracted from SAT of Bamei pigs at 1 d compared with Large White pigs. Knockdown of FoxO1 increased triglyceride content (P < 0.01) and upregulated the levels of adipocyte fatty-acid binding protein, PPARγ, and CCAAT enhancer-binding protein α (C/EBPα) at 6 d after porcine preadipocytes were induced. Furthermore, the transcriptional regulation of FoxO1 through C/EBPβ during early porcine preadipocyte differentiation and the effect of insulin on phosphoinositide 3 kinase (PI3K)/glycogen synthase kinase 3β (GSK3β) signal pathway by FoxO1 were examined. The results indicated that FoxO1 inhibited transcription activity of C/EBPβ, whereas C/EBPβ did not affect transcription activity of FoxO1. At 6 and 12 h of early differentiation, knockdown of FoxO1 triggered the transcription activity of C/EBPβ. In addition, FoxO1 protein interacted with C/EBPβ protein in porcine adipocytes at 12 h after induction. Under treatment with 100 nM insulin, knockdown or overexpression of FoxO1 mediated PI3K/GSK3β signaling via upregulating or downregulating the levels of GSK3β and its phosphorylation in adipocytes. Taken together, there is low, but detectable, expression of FoxO1 in SAT of obese pigs and FoxO1 inhibited adipogenesis through C/EBPβ and PI3K/GSK3β signaling pathway. These findings provide useful information to further the understanding of the function of FoxO1 in porcine adipogenesis.


Developmental and Comparative Immunology | 2014

Knockdown of PU.1 mRNA and AS lncRNA regulates expression of immune-related genes in zebrafish Danio rerio.

Ning Wei; Weijun Pang; Yu Wang; Yan Xiong; Ruxiang Xu; Wenjing Wu; Cunzhen Zhao; Gongshe Yang

The transcription factor PU.1 plays a key role in the development of immune system. Recent evidence demonstrated bidirectional transcription and a sense/antisense transcriptional regulatory manner in PU.1 locus. However, the effect of PU.1 mRNA and its antisense long non-coding RNA (AS lncRNA) on adaptive immunity in vivo is still not clear. In this study, we first confirmed the expression of PU.1 AS lncRNA by strand-specific RT-PCR in zebrafish. Additionally, we found that GFP was detected in zebrafish kidney using tissue smears after zebrafish was intraperitoneally injected with pLentiHI-PU.1 shRNA or pLentiHI-PU.1 AS shRNA for 2 days. Moreover, on day 0, 2 and 4, the levels of PU.1 and immune-related genes including TCRAC, Rag2, AID, IgLC-1, mIg, and sIg mRNAs were detected using real-time qPCR. The results showed that the levels of PU.1 and above 6 immune-related gene mRNAs were significantly downregulated on day 2 (P<0.05) and day 4 (P<0.01) by the treatment with the pLentiHI-PU.1 shRNA, whereas these genes were markedly upregulated by the treatment with the pLentiHI-PU.1 AS shRNA. Based on our results, we suggested that the effects of PU.1 transcripts including mRNA and AS lncRNA on immune-related gene expression in zebrafish were opposite. To our knowledge, this was the first report that a novel functional AS lncRNA in adaptive immunity was transcribed from the zebrafish PU.1 locus. Our findings provided novel insight into further exploration on modulating adaptive immunity by regulating PU.1 mRNA and AS lncRNA.


Experimental Cell Research | 2016

MAT2B promotes adipogenesis by modulating SAMe levels and activating AKT/ERK pathway during porcine intramuscular preadipocyte differentiation.

Cunzhen Zhao; Xiaochang Chen; Wenjing Wu; Wusu Wang; Weijun Pang; Gongshe Yang

Intramuscular fat (IMF) has been demonstrated as one of the crucial factors of livestock meat quality. The MAT2B protein with MAT2α catalyzes the formation of methyl donor S- adenosylmethionine (SAMe) to mediate cell metabolism including proliferation and apoptosis. However, the regulatory effect of MAT2B on IMF deposition is still unclear. In this study, the effect of MAT2B on adipogenesis and its potential mechanism during porcine intramuscular preadipocyte differentiation was studied. The results showed that overexpression of MAT2B promoted adipogenesis and significantly up-regulated the mRNA and protein levels of adipogenic marker genes including FASN, PPARγ and aP2, consistently, knockdown of MAT2B inhibited lipid accumulation and down-regulated the mRNA and protein levels of the above genes. Furthermore, flow cytometry and EdU-labeling assay indicated that MAT2B regulate adipogenesis was partly due to influence intracellular SAMe levels and further affect cell clonal expansion. Also, increased expression of MAT2B activated the phosphorylations of AKT and ERK1/2, whereas knockdown of MAT2B blocked AKT signaling and repressed the phosphorylation of ERK1/2. Moreover, the inhibitory effect of LY294002 (a specific PI3K inhibitor) on the activities of AKT and ERK1/2 was partially recovered by overexpression of MAT2B in porcine intramuscular adipocytes. Finally, Co-IP experiments showed that MAT2B can directly interact with AKT. Taken together, our findings suggested that MAT2B acted as a positive regulator through modifying SAMe levels as well as activating AKT/ERK signaling pathway to promote porcine intramuscular adipocyte differentiation.


Acta Biochimica et Biophysica Sinica | 2016

Lipogenesis in myoblasts and its regulation of CTRP6 by AdipoR1/Erk/PPARγ signaling pathway

Wenjing Wu; Yunmei Sun; Chen Zhao; Cunzhen Zhao; Xiaochang Chen; Guoqiang Wang; Weijun Pang; Gongshe Yang

The induced lipogenesis and its regulation in C2C12 myoblasts remain largely unclear. Here, we found that the cocktail method could significantly induce lipogenesis through regulating lipid metabolic genes and Erk1/2 phosphorylation in myoblasts. Meanwhile, the expression and secretion of CTRP6 were increased during ectopic lipogenesis. Moreover, CTRP6 knockdown down-regulated the levels of lipogenic genes and phosphorylated Erk1/2 (p-Erk1/2) in the early lipogenic stage, whereas up-regulated p-Erk1/2 in the terminal differentiation. Interestingly, the effect of CTRP6 siRNA was attenuated by U0126 (a special p-Erk1/2 inhibitor) in myoblasts. Furthermore, AdipoR1, not AdipoR2, was first identified as a receptor of CTRP6 during the process of mitotic clonal expansion. Collectively, we suggest that CTRP6 mediates the ectopic lipogenesis through AdipoR1/Erk/PPARγ signaling pathway in myoblasts. Our findings will shed light on the novel biological function of CTRP6 during myoblast lipogenesis and provide a hopeful direction of improving meat quality of domestic animal by lipogenic regulation in skeletal muscle myoblasts.


Journal of Agricultural and Food Chemistry | 2017

CTRP6 Regulates Porcine Adipocyte Proliferation and Differentiation by the AdipoR1/MAPK Signaling Pathway

Wenjing Wu; Jin Zhang; Chen Zhao; Yunmei Sun; Weijun Pang; Gongshe Yang

Intramuscular fat (IMF) and subcutaneous fat (SCF), which are modulated by adipogenesis of intramuscular and subcutaneous adipocytes, play key roles in pork quality. C1q/tumor necrosis factor-related protein 6 (CTRP6), an adipokine, plays an important role in the differentiation of 3T3-L1 cells. However, the effect and regulatory mechanisms of CTRP6 on porcine adipogenesis, and whether CTRP6 has the same effect on intramuscular and subcutaneous adipocytes, are still unknown. Here, we found that CTRP6 significantly inhibited both adipocyte proliferation assessed by proliferative marker expression, but CTRP6 decreased the proliferation rate of intramuscular adipocytes (IM) to a greater extent than subcutaneous adipocytes (SC). Moreover, CTRP6 promoted the activity of the p38 signaling pathway during the proliferation of both cell types. Nevertheless, in subcutaneous adipocytes, CTRP6 also influenced the phosphorylation of extracellular regulated protein kinases1/2 (p-Erk1/2), but not in intramuscular adipocytes. Additionally, during the differentiation of intramuscular and subcutaneous adipocytes, CTRP6 increased adipogenic genes expression and the level of p-p38, while it decreased the activity of p-Erk1/2. Interestingly, the effect of CTRP6 shRNA or CTRP6 recombinant protein was attenuated by U0126 (a special p-Erk inhibitor) or SB203580 (a special p-p38 inhibitor) in adipocytes. By target gene prediction and experimental validation, we demonstrated that CTRP6 may be a target of miR-29a in porcine adipocytes. Moreover, AdipoR1was identified as a receptor of CTRP6 in intramuscular adipocytes, but not in subcutaneous adipocytes. On the basis of the above findings, we suggest that CTRP6 was the target gene of miR-29a, inhibited intramuscular and subcutaneous adipocyte proliferation, but promoted differentiation by the mitogen-activated protein kinase (MAPK) signaling pathway. These findings indicate that CTRP6 played an essentially regulatory role in fat development.


Cell Proliferation | 2018

Knockdown of ubiquitin D inhibits adipogenesis during the differentiation of porcine intramuscular and subcutaneous preadipocytes

Chen Zhao; X. Yao; Xiaochang Chen; Wenjing Wu; Fengxue Xi; Gongshe Yang; Taiyong Yu

Intramuscular fat (IMF) has a significant influence on porcine meat quality. Ubiquitin D (UBD) is involved in the management of diverse intracellular processes. However, its physiological functions in adipose cell differentiation and proliferation are still poorly defined.


Acta Biochimica et Biophysica Sinica | 2018

Knockdown of LGALS12 inhibits porcine adipocyte adipogenesis via PKA–Erk1/2 signaling pathway

Wenjing Wu; Yajun Yin; Ke Xu; Yongjia Peng; Jin Zhang

Increasing intramuscular (IM) fat while concomitantly decreasing subcutaneous (SC) fat content is one major goal of pig breeding. Identifying genes involved in lipid metabolism is critical for this goal. Galectin-12 (LGALS12) has been proven to be an important regulator of fat deposition in mouse models; however, the effect and regulatory mechanisms of LGALS12 on porcine adipogenesis are still unknown. In this study, the effects of LGALS12 on fat deposition were explored with primary culture of porcine SC and IM adipocytes. Analysis of LGALS12 expression across different tissues revealed that LGALS12 was predominantly expressed in adipose tissue. The LGALS12 expression patterns across stages of adipocyte differentiation were also evaluated, with differences observed between SC and IM fat. Small interfering RNA (siRNA) of LGALS12 was designed and transfected into porcine adipocytes derived from SC and IM fat. After transfection, the expression level of LGALS12 was significantly reduced, and the number of lipid droplets was reduced in adipocytes from both SC and IM fat. Simultaneously, the levels of adipogenic markers, including PPARγ and aP2, were decreased, whereas hydrolysis markers, including adipose triglyceride lipase (ATGL) and hormone-sensitive lipase (HSL), were increased. Furthermore, the activation of lipolysis signals, such as the phosphorylation of PKA and Erk1/2, were observed with LGALS12 knockdown in terminally differentiated adipocytes from both SC and IM sources. Taken together, these results suggest that LGALS12 knockdown can inhibit adipogenesis of porcine adipocytes by downregulating lipogenic genes and activating the PKA-Erk1/2 signaling pathway.


Lipids in Health and Disease | 2017

Cell therapy could be a potential way to improve lipoprotein lipase deficiency

Wenjing Wu; Yajun Yin; Jie Zhong; Yongjia Peng; Shuncai Li; Libin Zheng; Hong Cao; Jin Zhang

Collaboration


Dive into the Wenjing Wu'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

Jin Zhang

Hebei Normal University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge