Network


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

Hotspot


Dive into the research topics where Hengbo Shi is active.

Publication


Featured researches published by Hengbo Shi.


Ppar Research | 2013

PPARγ Regulates Genes Involved in Triacylglycerol Synthesis and Secretion in Mammary Gland Epithelial Cells of Dairy Goats

Hengbo Shi; Jun Luo; Jiangjiang Zhu; Jun Li; Yuting Sun; Xianzi Lin; Liping Zhang; Dawei Yao; Huaiping Shi

To explore the function of PPARγ in the goat mammary gland, we cloned the whole cDNA of the PPARγ gene. Homology alignments revealed that the goat PPARγ gene is conserved among goat, bovine, mouse, and human. Luciferase assays revealed that rosiglitazone enhanced the activity of the PPARγ response element (PPRE) in goat mammary epithelial cells (GMECs). After rosiglitazone (ROSI) treatment of GMECs, there was a significant (P < 0.05) increase in the expression of genes related to triacylglycerol synthesis and secretion: LPL, FASN, ACACA, PLIN3, FABP3, PLIN2, PNPLA2, NR1H3, SREBF1, and SCD. The decreases in expression observed after knockdown of PPARγ relative to the control group (Ad-NC) averaged 65%, 52%, 67%, 55%, 65%, 58%, 85%, 43%, 50%, and 24% for SCD, DGAT1, AGPAT6, SREBF1, ACACA, FASN, FABP3, SCAP, ATGL, and PLIN3, respectively. These results provide direct evidence that PPARγ plays a crucial role in regulating the triacylglycerol synthesis and secretion in goat mammary cells and underscore the functional importance of PPARγ in mammary gland tissue during lactation.


Gene | 2013

MiR-27a suppresses triglyceride accumulation and affects gene mRNA expression associated with fat metabolism in dairy goat mammary gland epithelial cells.

Xianzi Lin; Jun Luo; Liping Zhang; Wei Wang; Hengbo Shi; Jiangjiang Zhu

MicroRNAs (miRNAs), a well-defined group of small RNAs containing about 22 nucleotides, participate in various biological metabolic processes. miR-27a is a miRNA that is known to regulate fat synthesis and differentiation in preadipocyte cells. However, little is known regarding the role that miR-27a plays in regulating goat milk fat synthesis. In this study, we determined the miR-27a expression profile in goat mammary gland and found that miR-27a expression was correlated with the lactation cycle. Additionally, prolactin promoted miR-27a expression in goat mammary gland epithelial cells. Further functional analysis showed that over-expression of miR-27a down-regulated triglyceride accumulation and decreased the ratio of unsaturated/saturated fatty acid in mammary gland epithelial cells. miR-27a also significantly affected mRNA expression related to milk fat metabolism. Specifically, over-expression of miR-27a reduced gene mRNA expression associated with triglyceride synthesis by suppressing PPARγ protein levels. This study provides the first experimental evidence that miR-27a regulates triglyceride synthesis in goat mammary gland epithelial cells and improves our understanding about the importance of miRNAs in milk fat synthesis.


Journal of Dairy Science | 2013

Peroxisome proliferator-activated receptor-γ stimulates the synthesis of monounsaturated fatty acids in dairy goat mammary epithelial cells via the control of stearoyl-coenzyme A desaturase.

Huaiping Shi; Jun Luo; Dawei Yao; Jiangjiang Zhu; Huifen Xu; Hengbo Shi; Juan J. Loor

In rodents, peroxisome proliferator-activated receptor-γ (PPARG) plays a crucial role in fatty acid (FA) metabolism through regulation of gene expression, including stearoyl-coenzyme A desaturase (SCD), which is the rate-limiting enzyme for the biosynthesis of monounsaturated FA. However, whether or how PPARG regulates the activity of mammary SCD in ruminants is unknown. This study explored the potential role of PPARG isoforms in regulating SCD mRNA expression in lactating goat mammary epithelial cells (GMEC). Using quantitative real-time PCR, we observed a positive correlation between PPARG and SCD expression in the goat mammary gland at peak lactation. Overexpression of both PPARG1 and PPARG2 in GMEC increased markedly the expression of SCD, the concentration of 16:1 and 18:1, and the desaturation indices of 16:1 and 18:1. The PPARG ligand rosiglitazone further increased SCD expression and desaturation indices in GMEC, overexpressing PPARG1 and PPARG2. Incubation with rosiglitazone alone increased the expression of SCD, but did not alter the concentration of 16- to 18-carbon FA or their desaturation indices. The results provide evidence that PPARG regulates the expression and activity of SCD in GMEC. As such, PPARG may contribute to regulation of SCD and monounsaturated FA synthesis during lactation.


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.


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.


Journal of Dairy Science | 2018

trans-10,cis-12 conjugated linoleic acid alters lipid metabolism of goat mammary epithelial cells by regulation of de novo synthesis and the AMPK signaling pathway

Tianying Zhang; Jiangtao Huang; H.B. Tian; Yue Ma; Zhi Chen; J.J. Wang; Hengbo Shi; Jun Luo

The trans-10,cis-12 isomer of conjugated linoleic acid (t10c12-CLA) is a biohydrogenation intermediate in the rumen and has been shown to cause milk fat depression in dairy goats. However, few studies have focused on the in vitro molecular mechanisms involved in the response of the goat mammary gland to t10c12-CLA. In the present study, RNA sequencing technology was used to investigate the effects of t10c12-CLA on goat mammary epithelial cells. From the data, 25,153 annotated transcripts were obtained, and differentially expressed genes were selected based on a false discovery rate <0.05. Candidate genes and potent cellular signaling pathways were identified through Gene Ontology (GO) and pathway analysis. Next, real-time quantitative PCR and Western blot analyses were used to verify the results of the RNA sequencing data. The results indicated that t10c12-CLA inhibits fatty acid synthesis through downregulation of genes involved in de novo fatty acid synthesis, and this process is likely correlated with the activation of the AMP-activated protein kinase signaling pathways.


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 Hengbo Shi's collaboration.

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

Jun Li

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Yuting Sun

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

Huifen Xu

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Wei Wang

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Xianzi Lin

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

H.B. Tian

Laboratory of Molecular Biology

View shared research outputs
Researchain Logo
Decentralizing Knowledge