Network


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

Hotspot


Dive into the research topics where Ping-Ping He is active.

Publication


Featured researches published by Ping-Ping He.


Atherosclerosis | 2014

Lipoprotein lipase: From gene to atherosclerosis

Yuan Li; Ping-Ping He; Da-Wei Zhang; Xi-Long Zheng; Fracisco S. Cayabyab; Wei-Dong Yin; Chao-Ke Tang

Lipoprotein lipase (LPL) is a key enzyme in lipid metabolism and responsible for catalyzing lipolysis of triglycerides in lipoproteins. LPL is produced mainly in adipose tissue, skeletal and heart muscle, as well as in macrophage and other tissues. After synthesized, it is secreted and translocated to the vascular lumen. LPL expression and activity are regulated by a variety of factors, such as transcription factors, interactive proteins and nutritional state through complicated mechanisms. LPL with different distributions may exert distinct functions and have diverse roles in human health and disease with close association with atherosclerosis. It may pose a pro-atherogenic or an anti-atherogenic effect depending on its locations. In this review, we will discuss its gene, protein, synthesis, transportation and biological functions, and then focus on its regulation and relationship with atherosclerosis and potential underlying mechanisms. The goal of this review is to provide basic information and novel insight for further studies and therapeutic targets.


PLOS ONE | 2013

Antagonism of betulinic acid on LPS-mediated inhibition of ABCA1 and cholesterol efflux through inhibiting nuclear factor-kappaB signaling pathway and miR-33 expression.

Guo-Jun Zhao; Shi-Lin Tang; Yun-Cheng Lv; Xin-Ping Ouyang; Ping-Ping He; Feng Yao; Wu-Jun Chen; Qian Lu; Yan-Yan Tang; Min Zhang; Yuchang Fu; Da-Wei Zhang; Kai Yin; Chao-Ke Tang

ATP-binding cassette transporter A1 (ABCA1) is critical in exporting cholesterol from macrophages and plays a protective role in the development of atherosclerosis. The purpose of this study was to investigate the effects of betulinic acid (BA), a pentacyclic triterpenoid, on ABCA1 expression and cholesterol efflux, and to further determine the underlying mechanism. BA promoted ABCA1 expression and cholesterol efflux, decreased cellular cholesterol and cholesterol ester content in LPS-treated macrophages. Furthermore, we found that BA promoted ABCA1 expression via down-regulation of miR-33s. The inhibition of LPS-induced NF-κB activation further decreased miR-33s expression and enhanced ABCA1 expression and cholesterol efflux when compared with BA only treatment. In addition, BA suppressed IκB phosphorylation, p65 phosphorylation and nuclear translocation, and the transcription of NF-κB-dependent related gene. Moreover, BA reduced atherosclerotic lesion size, miR-33s levels and NF-κB activation, and promoted ABCA1 expression in apoE−/− mice. Taken together, these results reveal a novel mechanism for the BA-mediated ABCA1 expression, which may provide new insights for developing strategies for modulating vascular inflammation and atherosclerosis.


Atherosclerosis | 2013

Apelin-13 increases expression of ATP-binding cassette transporter A1 via activating protein kinase C α signaling in THP-1 macrophage-derived foam cells.

Xiao-Yan Liu; Qian Lu; Xin-Ping Ouyang; Shi-Lin Tang; Guo-Jun Zhao; Yun-Cheng Lv; Ping-Ping He; Hai-Jun Kuang; Yan-Yan Tang; Yuchang Fu; Da-Wei Zhang; Chao-Ke Tang

Apelin has an antiatherogenic function through activating protein kinase C (PKC) to initiate a series of cellular signaling pathways. PKC phosphorylates and stabilizes ATP-binding cassette transporter A1 (ABCA1) through inhibiting its degradation mediated by calpain. Thus, in the present study, we investigated whether apelin-13 affects expression of ABCA1 through PKC signaling. The results showed that apelin-13 dramatically increased cholesterol efflux from THP-1 macrophage-derived foam cells and reduced cellular cholesterol levels. ABCA1 protein but not mRNA levels were dramatically increased by apelin-13, and calpain-induced degradation of ABCA1 and calpain activity were suppressed with treatment of apelin-13. However, the effects of apelin-13 on ABCA1 protein expression, cellular cholesterol efflux and calpain activity were abolished by depletion of PKCα, suggesting the potential important role of PKCα. In addition, apelin-13 was shown to phosphorylate serine residues in ABCA1 through the PKCα pathway. Thus, apelin-13 appears to activate PKCα, phosphorylate ABCA1 and inhibit calpain-mediated proteolysis, thereby promoting cholesterol efflux and reducing foam cell formation. Our study herein described a possible mechanism for understanding the antiatherogenic effects of apelin on attenuating the progression of atherosclerosis.


Biochimie | 2012

MicroRNA-467b targets LPL gene in RAW 264.7 macrophages and attenuates lipid accumulation and proinflammatory cytokine secretion

Guo-Ping Tian; Wu-Jun Chen; Ping-Ping He; Shi-Lin Tang; Guo-Jun Zhao; Yun-Cheng Lv; Xin-Ping Ouyang; Kai Yin; Ping-Ping Wang; Hong Cheng; Yuan Chen; Su-Lan Huang; Yuchang Fu; Da-Wei Zhang; Wei-Dong Yin; Chao-Ke Tang

LPL (lipoprotein lipase) is a rate-limiting enzyme involved in the hydrolysis of triglycerides. Previous studies have shown that microRNA (miR)-467b regulates hepatic LPL expression and plays a role in the progression of steatosis or abnormal lipid retention in obese mice. Macrophage-derived LPL has been shown to promote atherosclerosis. However, if miR-476b influences macrophage LPL expression and the subsequent effects are unknown. Here, we utilized oxLDL-treatment RAW 264.7 macrophages that were transfected with miR-467b mimics or inhibitors to investigate the potential roles of macrophage miR-476b. We found that miR-467b significantly decreased lipid accumulation and IL-6, IL-1β, TNF-α and MCP-1 secretions. Furthermore, our studies suggested an additional explanation for the regulatory mechanism of miR-467b on its functional target, LPL in RAW 264.7 macrophages. Thus, our findings indicate that miR-467b may regulate lipid accumulation and proinflammatory cytokine secretion in oxLDL-stimulated RAW 264.7 macrophages by targeting the LPL gene.


International Journal of Cardiology | 2014

NF-κB suppresses the expression of ATP-binding cassette transporter A1/G1 by regulating SREBP-2 and miR-33a in mice.

Guo-Jun Zhao; Shi-Lin Tang; Yun-Cheng Lv; Xin-Ping Ouyang; Ping-Ping He; Feng Yao; Yan-Yan Tang; Min Zhang; Ya-Ling Tang; Deng-Pei Tang; Francisco S. Cayabyab; Guo-Ping Tian; Chao-Ke Tang

a Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Life Science Research Center, University of South China, Hengyang, Hunan 421001, China b Department of Histology and Embryology, Guilin Medical University, Guilin, Guangxi 541004, China c Fourth year student in department of biochemistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada d Department of Physiology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada e Department of Cardiovascular Medicine, Second Affiliated Hospital of University of South China, Hengyang 421001, Hunan, China


PLOS ONE | 2016

MicroRNA-27 Prevents Atherosclerosis by Suppressing Lipoprotein Lipase-Induced Lipid Accumulation and Inflammatory Response in Apolipoprotein E Knockout Mice

Wei Xie; Liang Li; Min Zhang; Hai-Peng Cheng; Duo Gong; Yun-Cheng Lv; Feng Yao; Ping-Ping He; Xin-Ping Ouyang; Gang Lan; Dan Liu; Zhen-Wang Zhao; Yu-Lin Tan; Xi-Long Zheng; Weidong Yin; Chao-Ke Tang

Atherosclerotic lesions are lipometabolic disorder characterized by chronic progressive inflammation in arterial walls. Previous studies have shown that macrophage-derived lipoprotein lipase (LPL) might be a key factor that promotes atherosclerosis by accelerating lipid accumulation and proinflammatory cytokine secretion. Increasing evidence indicates that microRNA-27 (miR-27) has beneficial effects on lipid metabolism and inflammatory response. However, it has not been fully understood whether miR-27 affects the expression of LPL and subsequent development of atherosclerosis in apolipoprotein E knockout (apoE KO) mice. To address these questions and its potential mechanisms, oxidized low-density lipoprotein (ox-LDL)-treated THP-1 macrophages were transfected with the miR-27 mimics/inhibitors and apoE KO mice fed high-fat diet were given a tail vein injection with miR-27 agomir/antagomir, followed by exploring the potential roles of miR-27. MiR-27 agomir significantly down-regulated LPL expression in aorta and peritoneal macrophages by western blot and real-time PCR analyses. We performed LPL activity assay in the culture media and found that miR-27 reduced LPL activity. ELISA showed that miR-27 reduced inflammatory response as analyzed in vitro and in vivo experiments. Our results showed that miR-27 had an inhibitory effect on the levels of lipid both in plasma and in peritoneal macrophages of apoE KO mice as examined by HPLC. Consistently, miR-27 suppressed the expression of scavenger receptors associated with lipid uptake in ox-LDL-treated THP-1 macrophages. In addition, transfection with LPL siRNA inhibited the miR-27 inhibitor-induced lipid accumulation and proinflammatory cytokines secretion in ox-LDL-treated THP-1 macrophages. Finally, systemic treatment revealed that miR-27 decreased aortic plaque size and lipid content in apoE KO mice. The present results provide evidence that a novel antiatherogenic role of miR-27 was closely related to reducing lipid accumulation and inflammatory response via downregulation of LPL gene expression, suggesting a potential strategy to the diagnosis and treatment of atherosclerosis.


Biochemical and Biophysical Research Communications | 2014

The effects of miR-467b on lipoprotein lipase (LPL) expression, pro-inflammatory cytokine, lipid levels and atherosclerotic lesions in apolipoprotein E knockout mice.

Guo-Ping Tian; Yan-Yan Tang; Ping-Ping He; Yun-Cheng Lv; Xin-Pin Ouyang; Guo-Jun Zhao; Shi-Lin Tang; Jian-Feng Wu; Jia-Lin Wang; Juan Peng; Min Zhang; Yuan Li; Francisco S. Cayabyab; Xi-Long Zheng; Da-Wei Zhang; Wei-Dong Yin; Chao-Ke Tang

Atherosclerosis is a lipid disorder disease characterized by chronic blood vessel wall inflammation driven by the subendothelial accumulation of macrophages. Studies have shown that lipoprotein lipase (LPL) participates in lipid metabolism, but it is not yet known whether post-transcriptional regulation of LPL gene expression by microRNAs (miRNAs) occurs in vivo. Here, we tested that miR-467b provides protection against atherosclerosis by regulating the target gene LPL which leads to reductions in LPL expression, lipid accumulation, progression of atherosclerosis and production of inflammatory cytokines in apolipoprotein E knockout (apoE(-/-)) mice. Treatment of apoE(-/-) mice with intra-peritoneal injection of miR-467b agomir led to decreased blood plasma levels of total cholesterol (TC), triglyceride (TG), low density lipoprotein cholesterol (LDL-C), high density lipoprotein cholesterol (HDL-C), tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), IL-1β and monocyte chemotactic protein-1 (MCP-1). Using Western blots and real time PCR, we determined that LPL expression in aorta and abdominal cavity macrophages were significantly down-regulated in the miR-467b agomir group. Furthermore, systemic treatment with miR-467b antagomir accelerated the progression of atherosclerosis in the aorta of apoE(-/-) mice. The present study showed that miR-467b protects apoE(-/-) mice from atherosclerosis by reducing lipid accumulation and inflammatory cytokine secretion via downregulation of LPL expression. Therefore, targeting miR-467b may offer a promising strategy to treat atherosclerotic vascular disease.


PLOS ONE | 2015

MicroRNA-590 Inhibits Lipoprotein Lipase Expression and Prevents Atherosclerosis in apoE Knockout Mice

Ping-Ping He; Xin-Ping Ouyang; Yuan Li; Yun-Cheng Lv; Zongbao Wang; Feng Yao; Wei Xie; Yu-Lin Tan; Liang Li; Min Zhang; Gang Lan; Duo Gong; Hai-Peng Cheng; Hui-Juan Zhong; Dan Liu; Chong Huang; Zhao-Xia Li; Xi-Long Zheng; Weidong Yin; Chao-Ke Tang

Recent studies have suggested that miR-590 may play critical roles in cardiovascular disease. This study was designed to determine the effects of miR-590 on lipoprotein lipase (LPL) expression and development of atherosclerosis in apolipoprotein E knockout (apoE−/−) mice and explore the potential mechanisms. En face analysis of the whole aorta revealed that miR-590 significantly decreased aortic atherosclerotic plaque size and lipid content in apoE−/− mice. Double immunofluorescence staining in cross-sections of the proximal aorta showed that miR-590 agomir reduced CD68 and LPL expression in macrophages in atherosclerotic lesions. MiR-590 agomir down-regulated LPL mRNA and protein expression as analyzed by RT-qPCR and western blotting analyses, respectively. Consistently, miR-590 decreased the expression of CD36 and scavenger receptor A1 (SRA1) mRNA and protein. High-performance liquid chromatography (HPLC)analysis confirmed that treatment with miR-590 agomir reduced lipid levels either in plasma orinabdominal cavity macrophages of apoE−/− mice. ELISA analysis showed that miR-590 agomir decreased plasma levels of pro-inflammatory cytokines, such as tumor necrosis factor-alpha (TNF-α), monocyte chemotactic protein-1 (MCP-1), interleukin-1β (IL-1β)and interleukin-6 (IL-6). In contrast, treatment with miR-590 antagomir prevented or reversed these effects. Taken together, these results reveal a novel mechanism of miR-590 effects, and may provide new insights into the development of strategies for attenuating lipid accumulation and pro-inflammatory cytokine secretion.


Biochemical and Biophysical Research Communications | 2014

Interleukin-27 inhibits foam cell formation by promoting macrophage ABCA1 expression through JAK2/STAT3 pathway.

Hui Fu; Yan-Yan Tang; Xin-Ping Ouyang; Shi-Lin Tang; Hua Su; Xiaotao Li; Li-ping Huang; Miao He; Yun-Cheng Lv; Ping-Ping He; Feng Yao; Yu-Lin Tan; Wei Xie; Min Zhang; Jian-Feng Wu; Yuan Li; Kong Chen; Dan Liu; Gang Lan; Meng-Ya Zeng; Xi-Long Zheng; Chao-Ke Tang

The purpose of this study is to determine whether IL-27 regulates macrophage ABCA1 expression, foam cell formation, and also explore the underlying mechanisms. Here, we revealed that IL-27 decreased lipid accumulation in THP-1 derived macrophages through markedly enhancing cholesterol efflux and increasing ABCA1 expression at both protein and mRNA levels. Our study further demonstrated that IL-27 increased ABCA1 level via activation of signal transducer and activator of transcription 3 (STAT3). Inhibition of Janus kinase 2, (JAK2)/STAT3 suppressed the stimulatory effects of IL-27 on ABCA1 expression. The present study concluded that IL-27 reduces lipid accumulation of foam cell by upregulating ABCA1 expression via JAK2/STAT3. Therefore, targeting IL-27 may offer a promising strategy to treat atherosclerotic vascular disease.


Biochemical and Biophysical Research Communications | 2014

Growth differentiation factor-15 induces expression of ATP-binding cassette transporter A1 through PI3-K/PKCζ/SP1 pathway in THP-1 macrophages.

Jian-Feng Wu; Yan Wang; Min Zhang; Yan-Yan Tang; Bo Wang; Ping-Ping He; Yun-Cheng Lv; Xin-Ping Ouyang; Feng Yao; Yu-Lin Tan; Shi-Lin Tang; Deng-Pei Tang; Francisco S. Cayabyab; Xi-Long Zheng; Da-Wei Zhang; Gao-Feng Zeng; Chao-Ke Tang

OBJECTIVE The aim of this study was to determine whether ATP-binding cassette transporter A1 (ABCA1) was up-regulated by growth differentiation factor-15 (GDF-15) via the phosphoinositide 3-kinase (PI3K)/protein kinase Cζ (PKCζ)/specificity protein 1 (SP1) pathway in THP-1 macrophages. METHODS AND RESULTS We investigated the effects of different concentrations of GDF-15 on ABCA1 expression in THP-1 macrophages. The results showed that GDF-15 dramatically increased cholesterol efflux and decreased cellular cholesterol levels. In addition, GDF15 increased ABCA1 mRNA and protein levels. The effects of GDF-15 on ABCA1 protein expression and cellular cholesterol efflux were abolished by wither inhibition or depletion of PI3K, PKCζ and SP1, respectively, suggesting the potential roles of PI3K, PKCζ and SP1 in ABCA1 expression. Taken together, GDF-15 appears to activate PI3K, PKCζ and SP1 cascade, and then increase ABCA1 expression, thereby promoting cholesterol efflux and reducing foam cell formation. CONCLUSION Our results suggest that GDF-15 has an overall protective effect on the progression of atherosclerosis, likely through inducing ABCA1 expression via the PI3K/PKCζ/SP1 signaling pathway and enhancing cholesterol efflux.

Collaboration


Dive into the Ping-Ping He's collaboration.

Top Co-Authors

Avatar

Chao-Ke Tang

University of South China

View shared research outputs
Top Co-Authors

Avatar

Xin-Ping Ouyang

University of South China

View shared research outputs
Top Co-Authors

Avatar

Yun-Cheng Lv

University of South China

View shared research outputs
Top Co-Authors

Avatar

Min Zhang

University of South China

View shared research outputs
Top Co-Authors

Avatar

Feng Yao

University of South China

View shared research outputs
Top Co-Authors

Avatar

Yan-Yan Tang

University of South China

View shared research outputs
Top Co-Authors

Avatar

Guo-Jun Zhao

University of South China

View shared research outputs
Top Co-Authors

Avatar

Shi-Lin Tang

University of South China

View shared research outputs
Top Co-Authors

Avatar

Wei Xie

University of South China

View shared research outputs
Top Co-Authors

Avatar

Guo-Ping Tian

University of South China

View shared research outputs
Researchain Logo
Decentralizing Knowledge