Network


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

Hotspot


Dive into the research topics where Huifen Xu is active.

Publication


Featured researches published by Huifen Xu.


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 | 2016

Overexpression of SREBP1 (sterol regulatory element binding protein 1) promotes de novo fatty acid synthesis and triacylglycerol accumulation in goat mammary epithelial cells

Huifen Xu; Jun Luo; W.S. Zhao; Y.C. Yang; H.B. Tian; Huaiping Shi; Massimo Bionaz

Sterol regulatory element binding protein 1 (SREBP1; gene name SREBF1) is known to be the master regulator of lipid homeostasis in mammals, including milk fat synthesis. The major role of SREBP1 in controlling milk fat synthesis has been demonstrated in bovine mammary epithelial cells. Except for a demonstrated role in controlling the expression of FASN, a regulatory role of SREBP1 on milk fat synthesis is very likely, but has not yet been demonstrated in goat mammary epithelial cells (GMEC). To explore the regulatory function of SREBP1 on de novo fatty acids and triacylglycerol synthesis in GMEC, we overexpressed the mature form of SREBP1 (active NH2-terminal fragment) in GMEC using a recombinant adenovirus vector (Ad-nSREBP1), with Ad-GFP (recombinant adenovirus of green fluorescent protein) as control, and infected the GMEC for 48 h. In infected cells, we assessed the expression of 20 genes related to milk fat synthesis using real time-quantitative PCR, the protein abundance of SREBP1 and FASN by Western blot, the production of triacylglycerol, and the fatty acid profile. Expression of SREBF1 was modest in mammary compared with the other tissues in dairy goats but its expression increased approximately 30-fold from pregnancy to lactation. The overexpression of the mature form of SREBP1 was confirmed by >200-fold higher expression of SREBF1 in Ad-nSREBP1 compared with Ad-GFP. We observed no changes in amount of the precursor form of SREBP1 protein but a >10-fold increase of the mature form of SREBP1 protein with Ad-nSREBP1. Compared with Ad-GFP cells (control), Ad-nSREBP1 cells had a significant increase in expression of genes related to long-chain fatty acid activation (ACSL1), transport (FABP3), desaturation (SCD1), de novo synthesis of fatty acids (ACSS2, ACLY, IDH1, ACACA, FASN, and ELOVL6), and transcriptional factors (NR1H3 and PPARG). We observed a >10-fold increase in expression of INSIG1 but SCAP was downregulated by Ad-nSREBP1. Among genes related to milk fat synthesis and lipid droplet formation, only LPIN1 and DGAT1 were upregulated by Ad-nSREBP1. Compared with the Ad-GFP, the cellular triacylglycerol content was higher and the percentage of C16:0 and C18:1 increased, whereas that of C16:1, C18:0, and C18:2 decreased in Ad-nSREBP1 cells. Overall, the data provide strong support for a central role of SREBP1 in the regulation of milk fat synthesis in goat mammary cells.


Journal of Dairy Science | 2015

MicroRNA-24 can control triacylglycerol synthesis in goat mammary epithelial cells by targeting the fatty acid synthase gene.

Hui Wang; Jun Luo; Zhi Chen; Wenting Cao; Huifen Xu; Deming Gou; Jiangjiang Zhu

In nonruminants it has been demonstrated that microRNA-24 (miR-24) is involved in preadipocyte differentiation, hepatic lipid, and plasma triacylglycerol synthesis. However, its role in ruminant mammary gland remains unclear. In this study we measured miR-24 expression in goat mammary gland tissue at 4 different stages of lactation and observed that it had highest expression at peak lactation when compared with the dry period. Overexpression or downregulation of miR-24 in goat mammary epithelial cells (GMEC) strongly affected fatty acid profiles; in particular, miR-24 enhanced unsaturated fatty acid concentration. Additional effects of miR-24 included changes in triacylglycerol content and the expression of fatty acid synthase, sterol regulatory element binding transcription protein 1, stearoyl-CoA desaturase, glycerol-3-phosphate acyltransferase mitochondrial, and acetyl-CoA carboxylase. Luciferase reporter assay confirmed that fatty acid synthase is a target of miR-24. Taken together, these results not only highlight the physiological importance of miR-24 in fatty acid metabolism in GMEC, but also laid the foundation for further research on regulatory mechanisms among miR-24 and other microRNA expressed in GMEC.


RNA Biology | 2016

MicroRNA-26a/b and their host genes synergistically regulate triacylglycerol synthesis by targeting the INSIG1 gene

Hui Wang; Jun Luo; Tianying Zhang; H.B. Tian; Yue Ma; Huifen Xu; Dawei Yao; Juan J. Loor

ABSTRACT The microRNA-26 (miR-26) family is known to control adipogenesis in non-ruminants. The genomic loci of miR-26a and miR-26b have been localized in the introns of genes encoding for the proteins of the C-terminal domain RNA polymerase II polypeptide A small phosphatase (CTDSP) family. Insulin-induced gene 1 (INSIG1) encodes a protein with a key role in the regulation of lipogenesis in rodent liver. In the present study, we investigated the synergistic function of the miR-26 family and their host genes in goat mammary epithelial cells (GMEC). Downregulation of miR-26a/b and their host genes in GMEC decreased the expression of genes relate to fatty acid synthesis (PPARG, LXRA, SREBF1, FASN, ACACA, GPAM, LPIN1, DGAT1 and SCD1), triacylglycerol accumulation and unsaturated fatty acid synthesis. Luciferase reporter assays confirmed INSIG1 as a direct target of miR-26a/b. Furthermore, inhibition of the CTDSP family also downregulated the expression of INSIG1. Taken together, our findings highlight a functional association of miR-26a/b, their host genes and INSIG1, and provide new insights into the regulatory network controlling milk fat synthesis in GMEC. The data indicate that targeting this network via nutrition might be important for regulating milk fat synthesis in ruminants.


Journal of Cellular Physiology | 2018

Acyl‐CoA synthetase short‐chain family member 2 (ACSS2) is regulated by SREBP‐1 and plays a role in fatty acid synthesis in caprine mammary epithelial cells

Huifen Xu; Jun Luo; Gongzhen Ma; Xueying Zhang; Dawei Yao; Ming Li; Juan J. Loor

Sterol regulatory element binding protein 1 (SREBP‐1) is well‐known as the master regulator of lipogenesis in rodents. Acyl‐CoA synthetase short‐chain family member 2 (ACSS2) plays a key role in lipogenesis by synthesizing acetyl‐CoA from acetate for lipogenesis. ATP citrate lyase (ACLY) catalyzes the conversion of citrate and coenzyme A to acetyl‐CoA, hence, it is also important for lipogenesis. Although ACSS2 function in cancer cells has been elucidated, its essentiality in ruminant mammary lipogenesis is unknown. Furthermore, ACSS2 gene promoter and its regulatory mechanisms have not known. Expression of ACSS2 was high in lipid synthesizing tissues, and its expression increased during lactation compared with non‐lactating period. Simultaneous knockdown of both ACSS2 and ACLY by siRNA in primary goat mammary epithelial cells decreased (p < 0.05) the mRNA abundance of genes associated with de novo fatty acid synthesis (FASN, ACACA, SCD1) and triacylglycerol (TAG) synthesis (DGAT1, DGAT2, GPAM, and AGPAT6). Genes responsible for lipid droplet formation and secretion (PLIN2 and PLIN3) and fatty acid oxidation (ATGL, HSL, ACOX, and CPT1A) all decreased (p < 0.05) after ACSS2 and ACLY knockdown. Total cellular TAG content and lipid droplet formation also decreased. Use of a luciferase reporter assay revealed a direct regulation of ACSS2 by SREBP‐1. Furthermore, SREBP‐1 interacted with an SRE (SREBP response element) spanning at −475 to −483 bp on the ACSS2 promoter. Taken together, our results revealed a novel pathway that SREBP‐1 may regulate fatty acid and TAG synthesis by regulating the expression of ACSS2.


Journal of Cellular Physiology | 2017

SCD1 Alters Long-Chain Fatty Acid (LCFA) Composition and Its Expression Is Directly Regulated by SREBP-1 and PPARγ 1 in Dairy Goat Mammary Cells

Dawei Yao; Jun Luo; Qiuya He; H.B. Shi; Jun Li; Hui Wang; Huifen Xu; Zhi Chen; Yongqing Yi; Juan J. Loor

Stearoyl‐CoA desaturase 1 (SCD1) is a key enzyme for the synthesis of the monounsaturated fatty acids (MUFA) palmitoleic acid and oleic acid. In non‐ruminant species, SCD1 expression is known to be tightly regulated by a variety of transcription factors. Although the role of SCD1 and the transcriptional regulatory mechanism by SREBP‐1 and PPARs in other species is clear, changes in lipid metabolism related to SCD1 and via the regulation of SREBP‐1 or PPARG1 in ruminant mammary tissue remain largely unknown. Here, we demonstrated that SCD1 expression in goat mammary tissue is higher during lactation than the dry period. Overexpression of SCD1 increased the intracellular MUFA content and lipid accumulation, whereas SCD1 silencing resulted in a significant decrease in oleic acid concentration and triacylglycerol (TAG) accumulation. The overexpression of SREBF1 in goat mammary epithelial cells (GMEC) enhanced SCD1 expression and its promoter activity, but that effect was abolished when SREBF1 was silenced. Furthermore, deletion of sterol regulatory element (SRE) and the nuclear factor (NF‐Y)‐binding sites within a −1713 to +65‐base pair region of the SCD1 promoter completely abolished SREBP‐1‐induced SCD1 transcription. Otherwise, PPARG1 overexpression also stimulated the expression of SCD1 and its transcriptional activity directly via a PPAR response element (PPRE) in the SCD1 promoter. Together, these results indicate that SCD1 could markedly affect the fatty acid composition and rate of TAG synthesis through direct regulation via SREBP‐1 and PPARG1, hence, underscoring an important role of the enzyme and this transcription regulator in controlling mammary gland lipid synthesis in the goat. J. Cell. Physiol. 232: 635–649, 2017.


Scientific Reports | 2016

CD36 regulates lipopolysaccharide-induced signaling pathways and mediates the internalization of Escherichia coli in cooperation with TLR4 in goat mammary gland epithelial cells

Duoyao Cao; Jun Luo; Dekun Chen; Huifen Xu; Huaiping Shi; Xiaoqi Jing; Wenjuan Zang

The scavenger receptor CD36 is involved in pathogen recognition, phagocytosis, and pathogen-induced signaling. This study investigated the relationship between CD36 and TLR4 in modifying lipopolysaccharide (LPS)-induced signaling pathways and mediating Escherichia coli (E. coli) endocytosis in primary goat mammary epithelial cells (pGMECs). The manipulation of CD36 expression significantly influenced TLR4 and nuclear factor kappa B (NF-κB) mRNA expression in pGMECs stimulated with LPS for 12 h. NF-κB and activator protein-1 (AP-1) activity was regulated by the manipulation of CD36 expression in LPS-induced pGMECs. However, CD36-mediated AP-1 activation occurred primarily through c-Jun N-terminal kinase (c-JNK). Adaptor proteins and proinflammatory cytokines were also involved in these signaling pathways and acted by regulating CD36 expression in LPS-stimulated cells. Moreover, CD36 cooperated with TLR4 in TLR4-mediated phagocytosis following E. coli simulation, but this complex was not induced by LPS treatment. Our study is the first to illuminate CD36 as a scavenger receptor in ruminants. Additionally, this study indicates that CD36 plays a vital role in the LPS-induced activation of downstream signaling cascades and mediates E. coli phagocytosis via TLR4 in pGMECs, which offers a novel treatment strategy for mastitis.


Journal of Dairy Science | 2016

Thyroid hormone responsive (THRSP) promotes the synthesis of medium-chain fatty acids in goat mammary epithelial cells.

Dawei Yao; Jun Luo; Qiuya He; M. Wu; H.B. Shi; Hui Wang; M. Wang; Huifen Xu; Juan J. Loor

In nonruminants, thyroid hormone responsive (THRSP) is a crucial protein for cellular de novo lipogenesis. However, the role of THRSP in regulating the synthesis of milk fatty acid composition in goat mammary gland remains unknown. In the present study, we compared gene expression of THRSP among different goat tissues. Results revealed that THRSP had the highest expression in subcutaneous fat, and expression was higher during lactation compared with the dry period. Overexpression of THRSP upregulated the expression of fatty acid synthase (FASN), stearoyl-coenzyme A desaturase 1 (SCD1), diacylglycerol acyltransferase 2 (DGAT2), and glycerol-3-phosphate acyltransferase (GPAM) in goat mammary epithelial cells. In contrast, overexpression of THRSP led to downregulation of thrombospondin receptor (CD36) and had no effect on the expression of acetyl-coenzyme A carboxylase α (ACACA) and sterol regulatory element binding transcription factor1 (SREBF1). In addition, overexpressing THRSP in vitro resulted in a significant increase in triacylglycerol (TAG) concentration and the concentrations of C12:0 and C14:0. Taken together, these results highlight an important role of THRSP in regulating lipogenesis in goat mammary epithelial cells.


Experimental Cell Research | 2016

MicroRNA-181b suppresses TAG via target IRS2 and regulating multiple genes in the Hippo pathway.

Zhi Chen; Huaiping Shi; Shuang Sun; Huifen Xu; Duoyao Cao; Jun Luo

Milk fat metabolism is a complex procedure controlled by several factors. MiRNAs (microRNAs) regulate expression of genes and influence a series of biological procedures, such as fatty acid metabolism. Here we screened expression of goat mammary glands miRNA during peak-lactation and late-lactation, and found that miR-181b expresses remarkably. Moreover, we illustrated that the over-expression of miR-181b impaired fat metabolism while the knockdown of miR-181b promoted fat metabolism in GMEC. These findings extend the discovery of miR-181b functioning in mediating adipocyte differentiation, by suggesting its role in impairing fat metabolism, which develops our cognition on the importance of miRNAs in milk fat metabolism and synthesis. In this study, we find that over expressed miR-181b impaired adipogenesis and inhibited miR-181b promoted adipogenesis in GMEC. Using Luciferase reporter assay and Western Blot, IRS2 was illustrated to be a miR-181bs potential target gene. What is interesting is that miR-181b regulates multiple key components in the Hippo pathway, such as LATS1 and YAP1 in GMECs. In conclusion, our findings indicated that miR-181b suppress fat metabolism by means of regulating multiple genes in the Hippo pathway and target IRS2, which promotes further study on the function of miRNAs in milk fat metabolism and synthesis.


Journal of Dairy Science | 2016

Liver X receptor α promotes the synthesis of monounsaturated fatty acids in goat mammary epithelial cells via the control of stearoyl-coenzyme A desaturase 1 in an SREBP-1-dependent manner.

Dawei Yao; Jun Luo; Qiuya He; Huifen Xu; Jun Li; H.B. Shi; Hui Wang; Zhi Chen; Juan J. Loor

Stearoyl-coenzyme A desaturase 1 (SCD1) is a pivotal enzyme in the biosynthesis of monounsaturated fatty acids (MUFA). It is tightly regulated by transcription factors that control lipogenesis. In nonruminants, liver X receptor α (LXRα) is a nuclear receptor and transcription factor that acts as a key sensor of cholesterol and lipid homeostasis. However, the mechanism whereby LXRα regulates the expression and transcriptional activity of SCD1 in ruminant mammary cells remains unknown. In this study with goat mammary epithelial cells (GMEC), the LXRα agonist T 4506585 (T09) markedly enhanced the mRNA expression of SCD1 and sterol regulatory element binding factor 1 (SREBF1). The concentrations of C16:1 and C18:1 and their desaturation indices also were increased by LXRα activation. However, knockdown of LXRα did not alter the mRNA expression of SCD1. Although SCD1 was repressed by SREBF1 knockdown, T09 significantly increased SCD1 expression. Further analysis revealed that the SCD1 promoter activity was activated by LXRα overexpression. The goat SCD1 promoter contains 2 LXR response elements (LXRE), 1 sterol response element (SRE), and 1 nuclear factor Y (NF-Y) binding site. Site-directed mutagenesis of LXRE1, LXRE2, or SRE alone did not eliminate the upregulation of SCD1 when LXRα was overexpressed. In contrast, when NF-Y alone or in combination with SRE was mutated simultaneously, the basal transcriptional activity of the SCD1 promoter was markedly decreased and did not respond to LXRα overexpression. Furthermore, when SREBF1 was knocked down, overexpression of LXRα did not affect the promoter activity of SCD1. Together, these data suggest that LXRα regulates the expression of SCD1 through increasing SREBP-1 abundance to promote interaction with SRE and NF-Y binding sites. The present study provides evidence that LXRα is involved in the synthesis of MUFA in the goat mammary gland through an indirect mechanism.

Collaboration


Dive into the Huifen Xu's collaboration.

Top Co-Authors

Avatar

Jun Luo

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

Hui Wang

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

Jiangjiang Zhu

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Zhi Chen

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

H.B. Tian

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

Qiuya He

Laboratory of Molecular Biology

View shared research outputs
Researchain Logo
Decentralizing Knowledge