Network


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

Hotspot


Dive into the research topics where Yuting Sun is active.

Publication


Featured researches published by Yuting Sun.


Animal | 2014

Inhibition of FASN reduces the synthesis of medium-chain fatty acids in goat mammary gland.

Jiangjiang Zhu; Jun Luo; Wang W; K. Yu; Hui Wang; Huaiping Shi; Yuting Sun; Xianzi Lin; Jun Li

Fatty acid synthase (FASN) is known as a crucial enzyme of cellular de novo fatty acid synthesis in mammary gland which has been proved as the main source of short and medium-chain fatty acids of milk. However, the regulatory role of FASN in goat-specific milk fatty acids composition remains unclear. We cloned and analyzed the full-length of FASN gene from the mammary gland of Capra hircus (Xinong Saanen dairy goat) (DQ 915966). Comparative gene expression analysis suggested that FASN is predominantly expressed in fat, small intestine and mammary gland tissues, and expresses higher level at lactation period. Inhibition of FASN activity by different concentrations (0, 5, 15, 25 and 35 μM) of orlistat, a natural inhibitor of FASN, resulted in decreased expression of acetyl-CoA carboxylase α (ACCα), lipoprotein lipase and heart-type fatty acid binding protein (H-FABP) in a concentration-dependent manner in goat mammary gland epithelial cells (GMEC). Similar results were also obtained by silencing of FASN. Additionally, reduction of FASN expression also led to apparent decline of the relative content of decanoic acid (C10:0) and lauric acid (C12:0) in GMEC. Our study provides a direct evidence for inhibition of FASN reduces cellular medium-chain fatty acids synthesis in GMEC.


Journal of Dairy Science | 2014

Peroxisome proliferator-activated receptor γ1 and γ2 isoforms alter lipogenic gene networks in goat mammary epithelial cells to different extents.

Huaiping Shi; W.S. Zhao; Jun Luo; Dawei Yao; Yuting Sun; Jun Li; Hengbo Shi; Juan J. Loor

In nonruminants, the alternative splicing of peroxisome proliferator-activated receptor γ (PPARG) generates PPARG1 and PPARG2 isoforms. Although transcriptional control differences between isoforms have been reported in human adipose tissue, their roles in ruminant mammary cells are not well known. To assess which of these isoforms is more closely associated with the regulation of mammary lipogenic pathways, their tissue distribution was analyzed and the expression of key genes regulating lipogenic gene networks was measured after overexpression of the 2 isoforms in goat mammary epithelial cells (GMEC). The expression of PPARG2 was markedly greater in adipose tissue, whereas PPARG1 is the main isoform in goat mammary tissue (ratio of PPARG1:PPARG2 was close to 37:1). As was reported in previous work, PPARG1 upregulated the transcription regulators SREBF1 and PPARG and the lipogenic genes FASN, ACACA, and SCD. Along with a tendency for greater expression of AGPAT6, DGAT1, and PLIN2, these data suggest that PPARG1 is the isoform controlling lipogenesis in mammary cells. Addition of the PPARG ligand rosiglitazone (ROSI) to GMEC overexpressing both isoforms upregulated the expression of LPL and CD36, which help control uptake of long-chain fatty acids into mammary cells. Other responses to ROSI addition to GMEC overexpressing PPARG1 and PPARG2 included upregulation of AGPAT6, DGAT1, INSIG1, SREBF1, and NR1H3. Although the data suggest that both PPARG1 and PPARG2 could affect mammary lipogenesis via control of gene expression when stimulated (e.g., by ROSI), the fact that PPARG1 is more abundant in mammary tissue and that its overexpression alone upregulated key lipogenic gene networks suggest that it is the more important isoform in goat mammary cells.


Journal of Dairy Science | 2015

Short communication: Effect of inhibition of fatty acid synthase on triglyceride accumulation and effect on lipid metabolism genes in goat mammary epithelial cells

Jiangjiang Zhu; Jun Luo; Yuting Sun; Huaiping Shi; Jun Li; M. Wu; K. Yu; Abiel Berhane Haile; Juan J. Loor

The role of fatty acid synthase (FASN) on de novo fatty acid synthesis has been well established. In monogastrics, unlike acetyl-coenzyme A carboxylase, FASN is primarily controlled at the transcriptional level. However, no data exist on ruminant mammary cells evaluating effects of FASN knockdown on mRNA expression of lipogenic genes. Inhibition of FASN in mammary cells by C75-mediated interference, a synthetic inhibitor of FASN activity, and short hairpin RNA-mediated interference markedly reduced cellular triglyceride content at least in part by decreasing the expression of genes related to triglyceride synthesis (GPAT, AGPAT6, and DGAT2) and enhancing the expression of lipolysis-related genes (ATGL and HSL). Consistent with the markedly lower expression of genes related to lipid droplet formation and secretion (TIP47, ADFP, BTN1A1, and XDH), cellular lipid droplets also were reduced sharply after incubation with C75 or adenovirus-short-hairpin-RNA. The results underscored the essential role of FASN in the overall process of milk-fat formation in goat mammary epithelial cells.


PLOS ONE | 2015

MiR130b-Regulation of PPARγ Coactivator- 1α Suppresses Fat Metabolism in Goat Mammary Epithelial Cells

Zhi Chen; Jun Luo; Liuan Ma; Hui Wang; Wenting Cao; HuiFei Xu; Jiangjiang Zhu; Yuting Sun; Jun Li; Dawei Yao; Kang Kang; Deming Gou

Fat metabolism is a complicated process regulated by a series of factors. microRNAs (miRNAs) are a class of negative regulator of proteins and play crucial roles in many biological processes; including fat metabolism. Although there have been some researches indicating that miRNAs could influence the milk fat metabolism through targeting some factors, little is known about the effect of miRNAs on goat milk fat metabolism. Here we utilized an improved miRNA detection assay, S-Poly-(T), to profile the expression of miRNAs in the goat mammary gland in different periods, and found that miR-130b was abundantly and differentially expressed in goat mammary gland. Additionally, overexpressing miR-130b impaired adipogenesis while inhibiting miR-130b enhanced adipogenesis in goat mammary epithelial cells. Utilizing 3’-UTR assay and Western Blot analusis, the protein peroxisome proliferator-activated receptor coactivator-1α (PGC1α), a major regulator of fat metabolism, was demonstrated to be a potential target of miR-130b. Interestingly, miR-130b potently repressed PGC1α expression by targeting both the PGC1α mRNA coding and 3’ untranslated regions. These findings have some insight of miR-130b in mediating adipocyte differentiation by repressing PGC1α expression and this contributes to further understanding about the functional significance of miRNAs in milk fat synthesis.


Comparative Biochemistry and Physiology B | 2015

Regulation of the fatty acid synthase promoter by liver X receptor α through direct and indirect mechanisms in goat mammary epithelial cells.

Jun Li; Jun Luo; Jiangjiang Zhu; Yuting Sun; Dawei Yao; Hengbo Shi; Wei Wang

Fatty acid synthase (FASN) is a central enzyme of milk fat synthesis in the ruminant mammary gland. However, the mechanisms regulating goat FASN transcription remain elusive. The objective of this study was to investigate the mechanisms by which liver X receptor α (LXRα) regulates the FASN promoter in goat mammary epithelial cells (GMECs). In this study, T0901317 (T09), an agonist for LXRα, significantly enhanced the mRNA expression and promoter activity of FASN. Cloning of the dairy goat FASN promoter revealed the presence of one LXR response element (LXRE) and two sterol regulatory elements (SREs). Deletion or mutation of the FASN promoter LXRE reduced, but did not eliminate the transcriptional response of FASN to T09. While the LXRE and the SREs were both disrupted, basal transcription was severely reduced and there was no response to T09 treatment. This suggested that a complete response required one LXRE and two SREs. Knockdown of LXRα by siRNA did not alter the basal or T09-induced transcriptional activity of FASN. However, when sterol regulatory binding protein 1 (SREBP1) was knocked down, T09 significantly increased FASN transcription by wild-type GMECs, but had no effect on cells with LXRE-mutant promoters. The results suggested that LXR regulates FASN promoter activity through direct interaction with the LXRE as well as through increasing SREBP1 abundance. The present study provides insight into the transcriptional regulatory mechanisms controlling de novo fatty acid synthesis in GMECs.


Journal of Dairy Science | 2015

Adipocyte differentiation-related protein promotes lipid accumulation in goat mammary epithelial cells

Huaiping Shi; K. Yu; Jun Luo; Jun Li; H.B. Tian; Jiangjiang Zhu; Yuting Sun; Dawei Yao; Huifen Xu; Hengbo Shi; Juan J. Loor

Milk fat originates from the secretion of cytosolic lipid droplets (CLD) synthesized within mammary epithelial cells. Adipocyte differentiation-related protein (ADRP; gene symbol PLIN2) is a CLD-binding protein that is crucial for synthesis of mature CLD. Our hypothesis was that ADRP regulates CLD production and metabolism in goat mammary epithelial cells (GMEC) and thus plays a role in determining milk fat content. To understand the role of ADRP in ruminant milk fat metabolism, ADRP (PLIN2) was overexpressed or knocked down in GMEC using an adenovirus system. Immunocytochemical staining revealed that ADRP localized to the surface of CLD. Supplementation with oleic acid (OA) enhanced its colocalization with CLD surface and enhanced lipid accumulation. Overexpression of ADRP increased lipid accumulation and the concentration of triacylglycerol in GMEC. In contrast, morphological examination revealed that knockdown of ADRP decreased lipid accumulation even when OA was supplemented. This response was confirmed by the reduction in mass of cellular TG when ADRP was knocked down. The fact that knockdown of ADRP did not completely eliminate lipid accumulation at a morphological level in GMEC without OA suggests that some other compensatory factors may also aid in the process of CLD formation. The ADRP reversed the decrease of CLD accumulation induced by adipose triglyceride lipase. This is highly suggestive of ADRP promoting triacylglycerol stability within CLD by preventing access to adipose triglyceride lipase. Collectively, these data provide direct in vitro evidence that ADRP plays a key role in CLD formation and stability in GMEC.


Gene | 2015

Adipose triglyceride lipase regulates lipid metabolism in dairy goat mammary epithelial cells.

Jun Li; Jun Luo; Hui Wang; Hengbo Shi; Jiangjiang Zhu; Yuting Sun; Kang Yu; Dawei Yao

Adipose triglyceride lipase (ATGL) catalyzes the initial step in the lipid lipolysis process, hydrolyzing triglyceride (TG) to produce diacylglycerol (DG) and free fatty acids (FFA). In addition, ATGL regulates lipid storage and release in adipocyte cells. However, its role in mammary gland tissue remains unclear. To assess the role of the ATGL gene in the goat mammary gland, this study analyzed the tissue distribution and expression of key genes together with lipid accumulation after knockdown of the ATGL gene. The mRNA of ATGL was highly expressed in subcutaneous adipose tissue, the lung and the mammary gland with a significant increase in expression during the lactation period compared with the dry period of the mammary gland. Knockdown of the ATGL gene in goat mammary epithelial cells (GMECs) using siRNA resulted in a significant decrease in both ATGL mRNA and protein levels. Silencing of the ATGL gene markedly increased lipid droplet accumulation and intracellular TG concentration (P<0.05), while it reduced FFA levels in GMECs (P<0.05). Additionally, the expression of HSL for lipolysis, FABP3 for fatty acid transport, PPARα for fatty acid oxidation, ADFP, BTN1A1, and XDH for milk fat formation and secretion was down-regulated (P<0.05) after knockdown of the ATGL gene, with increased expression of CD36 for fatty acid uptake (P<0.05). In conclusion, these data suggest that the ATGL gene plays an important role in triglyceride lipolysis in GMECs and provides the first experimental evidence that ATGL may be involved in lipid metabolism during lactation.


Animal Science Journal | 2016

Effect of short-chain fatty acids on triacylglycerol accumulation, lipid droplet formation and lipogenic gene expression in goat mammary epithelial cells

Yuting Sun; Jun Luo; Jiangjiang Zhu; Hengbo Shi; Jun Li; Siyuan Qiu; Ping Wang; Juan J. Loor

Short-chain fatty acids (SCFAs) are the major energy sources for ruminants and are known to regulate various physiological functions in other species. However, their roles in ruminant milk fat metabolism are still unclear. In this study, goat mammary gland epithelial cells (GMECs) were treated with 3 mmol/L acetate, propionate or butyrate for 24 h to assess their effects on lipogenesis. Data revealed that the content of triacylglycerol (TAG) and lipid droplet formation were significantly stimulated by propionate and butyrate. The expression of FABP3, SCD1, PPARG, SREBP1, DGAT1, AGPAT6 and ADRP were upregulated by propionate and butyrate treatment. In contrast, the messenger RNA (mRNA) expression of FASN and LXRα was not affected by propionate, but reduced by butyrate. Acetate had no obvious effect on the content of TAG and lipid droplets but increased the mRNA expression of SCD1 and FABP3 in GMECs. Additionally, it was observed that propionate significantly increased the relative content of mono-unsaturated fatty acids (C18:1 and C16:1) at the expense of decreased saturated fatty acids (C16:0 and C18:0). Butyrate and acetate had no significant effect on fatty acid composition. Overall, the results from this work help enhance our understanding of the regulatory role of SCFAs on goat mammary cell lipid metabolism.


Animal Production Science | 2017

Inhibitions of FASN suppress triglyceride synthesis via the control of malonyl-CoA in goat mammary epithelial cells

Jun Luo; Jiangjiang Zhu; Yuting Sun; Huaiping Shi; Jun Li

Fatty acid synthase (FASN) is the key enzyme for de novo fatty acid synthesis from acetyl-CoA and malonyl-CoA. All the steps involved in fatty acid synthesis by FASN have been clearly defined in monogastrics and ruminants. However, there are no data on the mechanism of how FASN affects triglyceride synthesis. Inhibition of FASN in goat mammary epithelial cells by C75, a synthetic inhibitor of FASN activity, and shRNA markedly suppressed the accumulation of triglyceride in goat mammary epithelial cells. Meanwhile, C75 treatment significantly reduced the relative content of monounsaturated fatty acids (C16:1 and C18:1). Corresponding to the suppression of lipid accumulation, both of C75 and shRNA also decreased the mRNA expression of GPAM, AGPAT6 and DGAT2, all of which are related to triglyceride synthesis. The fact that treatment of malonyl-CoA decreased the expression of these genes is consistent with the results of shRNA treatment. Furthermore, the supplement of malonyl-CoA enhanced the suppression on GPAM, AGPAT6, LPIN1, DGAT1 and DGAT2. The results underscore the role of malonyl-CoA in inhibition of FASN in regulating triglyceride synthesis in goat mammary epithelial cells.


Gene | 2012

Goat liver X receptor α, molecular cloning, functional characterization and regulating fatty acid synthesis in epithelial cells of goat mammary glands.

Wei Wang; Jun Luo; Yu Zhong; Xianzi Lin; Hengbo Shi; Jiangjiang Zhu; Jun Li; Yuting Sun; Wangsheng Zhao

Collaboration


Dive into the Yuting Sun's collaboration.

Top Co-Authors

Avatar

Jun Li

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Jun Luo

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Jiangjiang Zhu

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Hengbo Shi

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Dawei Yao

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Huaiping Shi

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Hui Wang

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Huifen Xu

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

K. Yu

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Wei Wang

Laboratory of Molecular Biology

View shared research outputs
Researchain Logo
Decentralizing Knowledge