Network


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

Hotspot


Dive into the research topics where Lingxia Pang is active.

Publication


Featured researches published by Lingxia Pang.


Scientific Reports | 2015

miR-148a is Associated with Obesity and Modulates Adipocyte Differentiation of Mesenchymal Stem Cells through Wnt Signaling

Chunmei Shi; Min Zhang; Mei-Ling Tong; Lei Yang; Lingxia Pang; Ling Chen; Guangfeng Xu; Xia Chi; Qin Hong; Yuhui Ni; Chenbo Ji; Xirong Guo

Obesity results from numerous, interacting genetic, behavioral, and physiological factors. Adipogenesis is partially regulated by several adipocyte-selective microRNAs (miRNAs) and transcription factors that regulate proliferation and differentiation of human adipose-derived mesenchymal stem cells (hMSCs-Ad). In this study, we examined the roles of adipocyte-selective miRNAs in the differentiation of hMSCs-Ad to adipocytes. Results showed that the levels of miR-148a, miR-26b, miR-30, and miR-199a increased in differentiating hMSCs-Ad. Among these miRNAs, miR-148a exhibited significant effects on increasing PPRE luciferase activity (it represents PPAR-dependent transcription, a major factor in adipogenesis) than others. Furthermore, miR-148a expression levels increased in adipose tissues from obese people and mice fed high-fat diet. miR-148a acted by suppressing its target gene, Wnt1, an endogenous inhibitor of adipogenesis. Ectopic expression of miR-148a accelerated differentiation and partially rescued Wnt1-mediated inhibition of adipogenesis. Knockdown of miR-148a also inhibited adipogenesis. Analysis of the upstream region of miR-148a locus identified a 3 kb region containing a functional cAMP-response element-binding protein (CREB) required for miR-148a expression in hMSCs-Ad. The results suggest that miR-148a is a biomarker of obesity in human subjects and mouse model, which represents a CREB-modulated miRNA that acts to repress Wnt1, thereby promoting adipocyte differentiation.


Molecular and Cellular Endocrinology | 2014

MiR-146b is a regulator of human visceral preadipocyte proliferation and differentiation and its expression is altered in human obesity.

Ling Chen; Yong-mei Dai; Chenbo Ji; Lei Yang; Chunmei Shi; Guangfeng Xu; Lingxia Pang; Fangyan Huang; Chun-Mei Zhang; Xirong Guo

Visceral obesity is an independent risk factor for metabolic syndrome, and abnormal fat accumulation is linked to increases in the number and size of adipocytes. MiR-146b was a miRNA highly expressed in mature adipocytes while very lowly expressed in human mesenchymal stem cells (hMSCs) and human visceral preadipocytes (vHPA). In this paper, we mainly focused on the roles of miR-146b in adipogenesis. We found miR-146b could inhibit the proliferation of visceral preadipocytes and promote their differentiation. MiR-146b in human visceral adipocytes inhibited the expression of KLF7, a member of the Kruppel-like transcription factors, as demonstrated by a firefly luciferase reporter assay, indicating that KLF7 is a direct target of the endogenous miR-146b. MiR-146b expression was significantly altered in visceral and subcutaneous adipose tissues in human overweight and obese subjects, and in the epididymal fat tissues and brown fat tissues of diet-induced obese mice. Our data indicates that miR-146b may be a new therapeutic target against human visceral obesity and metabolic dysfunction.


Biochemical and Biophysical Research Communications | 2014

Distinct expression profiles of LncRNAs between brown adipose tissue and skeletal muscle

Jun Zhang; Xianwei Cui; Yahui Shen; Lingxia Pang; Aiqing Zhang; Ziyi Fu; Jiantao Chen; Xirong Guo; Weihua Gan; Chenbo Ji

Both brown adipose tissue and skeletalmuscle have abundant mitochondria and energy consumption capacity. They are similar in origin and gain different potential of energy metabolism after differentiation and maturation. The mechanism that cause the difference is not yet fully understood. Long non-coding RNAs (lncRNAs) which comprise the bulk of the human non-coding transcriptome have been proved to play key roles in various biological processes. Whether they will have a function on the differentiation and energy metabolism between BAT and skeletalmuscle is still unknown. To identify the cellular long noncoding RNAs (lncRNAs) involved in the progress, we used the next generation transcriptome sequencing and microarray techniques, and investigated 704 up-regulated and 896 down-regulated lncRNAs (fold-change >3.0) in BAT by comparing the expression profile. Furthermore, we reported AK003288 associated with junctophilin 2 (Jph2) gene which may affect energy metabolism. This study show distinct expression profiles of LncRNAs between brown adipose tissue and skeletal muscle which provide information for further research on differentiation of adipocyte and transdifferentiation between BAT and skeletalmuscle that will be helpful to find a new therapeutic target for combatting obesity.


Molecular Genetics and Genomics | 2015

Differential lncRNA expression profiles in brown and white adipose tissues

Jiantao Chen; Xianwei Cui; Chunmei Shi; Ling Chen; Lei Yang; Lingxia Pang; Jun Zhang; Xirong Guo; Jiaqin Wang; Chenbo Ji

Long non-coding RNAs (lncRNAs) are an important class of pervasive genes involved in a variety of biological functions. It can serve as key co-activators of proteins involved in transcriptional regulation. Studies have found that white and brown adipocytes both originate from the mesoderm. However, it remains unclear whether lncRNAs function during adipogenesis or in energy metabolism in brown adipose tissue (BAT) and white adipose tissue (WAT). In this study, we used lncRNA microarray technology to evaluate differences in the lncRNA expression profiles of WAT and BAT. We observed 735 up-regulated and 877 down-regulated lncRNAs (fold change >4.0). To reveal the potential functions of these lncRNAs, we applied GO and pathway analyses to study the differentially expressed lncRNAs. We found that AK142386 and AK133540 may affect adipogenesis and metabolism. Our data indicate that AK142386 and AK133540 may be involved in BAT and WAT development through their target genes Hoxa3 and Acad10. Together, we have identified numerous lncRNAs and these lncRNAs can potentially serve as a required component for proper adipogenesis.


Cellular Physiology and Biochemistry | 2014

Insight into the Effects of Adipose Tissue Inflammation Factors on miR-378 Expression and the Underlying Mechanism

Xinye Jiang; Mei Xue; Ziyi Fu; Chenbo Ji; Xirong Guo; Lu Zhu; Lulian Xu; Lingxia Pang; Meiyu Xu; Hongming Qu

Background/Aims: Obesity and the related metabolic syndrome have emerged as major public health issues in modern society. miRNAs have been shown to play key roles in regulating obesity-related metabolic syndrome, and some miRNAs regulated by adiponectin were identified as novel targets for controlling adipose tissue inflammation. miR-378 is a candidate target that was shown to be involved in adipose differentiation, mitochondrial metabolism and systemic energy homeostasis. However, little is known about the regulatory mechanisms of miR-378 expression. To better understand the physiological role of miR-378 in obesity and metabolic syndrome, it is crucial that we understand the regulation of miR-378 gene expression in human adipocytes. Methods: In this study, we investigated the effects of adipokines and inflammatory cytokines on miR-378 expression using Real-time PCR and the potential regulatory mechanisms using luciferase reporter assays and electrophoretic mobility shift assay (EMSA). Results: We found that adipokines and cytokines upregulated miR-378 expression primarily through SREBP and C/EBP binding sites in the miR-378 promoter region. Conclusion: Our findings showed that adipokines induced miR-378 expression and revealed the most likely mechanism of adipokine-induced miR-378 dysregulation in human adipocytes. miRNAs have been shown to function in regulating obesity-related metabolic syndrome, and miR-378 may be a novel target for controlling adipose tissue inflammation. This study offers a theoretical basis for understanding systemic adipose tissue inflammation and may provide new strategies for clinical treatment.


Journal of Molecular Endocrinology | 2016

MiR-1275 inhibits adipogenesis via ELK1 and its expression decreases in obese subjects

Lingxia Pang; Lianghui You; Chenbo Ji; Chunmei Shi; Ling Chen; Lei Yang; Fangyan Huang; Yahui Zhou; Jun Zhang; Xiao-Hui Chen; Xirong Guo

Excessive adipocyte differentiation and proliferation are closely associated with the onset of obesity, which has been partially linked to microRNA expression. In previous studies, using miRNA microarray screening, we found that miR-1275 was significantly decreased in human mature adipocytes. In this study, we examined the role of miR-1275 in adipogenesis. Our results indicated that miR-1275 can inhibit the differentiation of human visceral preadipocytes without affecting their proliferation. ELK1, an E-twenty-six (ETS)-domain transcription factor associated with adipocyte differentiation, was strongly suppressed by miR-1275 in human visceral adipocytes. This was demonstrated via a dual-luciferase reporter assay and pointed to ELK1 as a direct target of miR-1275. Furthermore, miR-1275 expression was significantly diminished in the visceral adipose tissue of overweight and obese human subjects accompanied by a negative correlation with body mass index. These results suggest that miR-1275 could play a future role in the management of obesity, as a novel therapeutic target or biomarker.


Molecular Biology Reports | 2015

The biological effects of hsa-miR-1908 in human adipocytes

Lei Yang; Chunmei Shi; Ling Chen; Lingxia Pang; Guangfeng Xu; Nan Gu; Lijun Zhu; Xirong Guo; Yuhui Ni; Chenbo Ji

MicroRNAs (miRNAs) are small non-coding RNAs involved in the regulation of gene expression. MiR-1908 is a recently identified miRNA that is highly expressed in human adipocytes. However, it is not known what role of miR-1908 is involved in the regulation of human adipocytes. In this study, we demonstrate that the level of miR-1908 increases during the adipogenesis of human multipotent adipose-derived stem (hMADS) cells and human preadipocytes-visceral. Overexpression of miR-1908 in hMADS cells inhibited adipogenic differentiation and increased cell proliferation, suggesting that miR-1908 is involved in the regulation of adipocyte cell differentiation and metabolism, and, thus, may have an effect on human obesity.


Molecular Medicine Reports | 2016

Expression of miR-199a-3p in human adipocytes is regulated by free fatty acids and adipokines

Nan Gu; Lianghui You; Chunmei Shi; Lei Yang; Lingxia Pang; Xianwei Cui; Chenbo Ji; Wen Zheng; Xirong Guo

Obesity is associated with a notable risk for disease, including risk of cardiovascular disorders, type 2 diabetes mellitus (T2DM) and hypertension. Adipose tissue modulates the metabolism by releasing free fatty acids (FFAs) and adipokines, including leptin, resistin, tumor necrosis factor-α (TNF-α) and interleukin 6 (IL-6). Altered secretion patterns of FFAs and adipokines have been demonstrated to result in obesity-associated insulin resistance (IR) and inflammatory responses. MicroRNA-199a-3p (miR)-199a-3p expression is significantly induced in differentiated human adipose-derived mesenchymal stem cells and indicates the association with T2DM. However, the association between miR-199a-3p levels in adipocytes and obesity-associated IR, as well as inflammatory responses remains to be elucidated. The present study observed an elevation of miR-199a-3p expression level in mature human adipocytes (visceral) compared with pre-adipocytes. In addition, miR-199a-3p expression was higher in visceral adipose deposits from obese subjects. FFA, TNF-α, IL-6 and leptin significantly induced miR-199a-3p expression in mature human adipocytes, while resistin had the opposite effect. miR-199a-3p may represent a factor in the modulation of obesity-associated IR and inflammatory responses.


Scientific Reports | 2018

Evaluation and optimization of differentiation conditions for human primary brown adipocytes

Xingyun Wang; Lianghui You; Xianwei Cui; Yun Li; Xing Wang; Pengfei Xu; Lijun Zhu; Juan Wen; Lingxia Pang; Xirong Guo; Chenbo Ji

As an effective way to improve energy expenditure, increasing the mass and activity of brown adipose tissue (BAT) has become a promising treatment for obesity and its associated disorders. Many efforts have been made to promote brown adipogenesis and increase the thermogenic capacity of brown adipose cells (BACs). The present culture schemes for human BAC differentiation are mostly derived from white adipocyte differentiation schemes. To solve this issue, we compared the adipogenic and thermogenic effects of various components on human BAC differentiation and optimized their concentrations as well as the culture time for BAC differentiation. In this study, we found that the induction factors did not show a dose-dependent promotion of brown adipogenesis or thermogenic capacity. The higher differentiation levels did not inevitably result in higher BAT-specific gene expression levels or increased β3-receptor agonist sensitivity. As an important element of culture medium, triiodothyronine was found to be essential for differentiation and metabolic property maintenance. Furthermore, compared with other reported methods, this protocol induced a specific intrinsic differentiation program. Our study provides not only an optimized method for human BAC differentiation but also a cell model with good differentiation and thermogenic capacity for brown adipose research.


Biochimica et Biophysica Acta | 2018

PID1 in adipocytes modulates whole-body glucose homeostasis

Ling Chen; Xingyun Wang; Jingai Zhu; Lianghui You; Xing Wang; Xianwei Cui; Chunmei Shi; Fangyan Huang; Yahui Zhou; Lei Yang; Lingxia Pang; Yao Gao; Chenbo Ji; Xirong Guo

The novel obesity-associated protein Phosphotyrosine Interaction Domain containing 1 (PID1) inhibits insulin-PI3K/Akt signaling pathway and insulin-stimulated glucose uptake in vitro. In this study, we generated fat tissue-specific aP2-PID1 transgenic (aP2-PID1tg) mice and PID1 knockout (PID1-/-) mice to explore how PID1 affects glucose metabolism in vivo. We observed insulin resistance and impaired insulin-PI3K/Akt signaling in aP2-PID1tg mice. Consistent with these data, the PID1-/- mice displayed improved glucose tolerance and insulin sensitivity under chow diet, with increased Akt phosphorylation in white adipose tissue (WAT). We further demonstrated that PID1 could interact with low density lipoprotein receptor-related protein 1 (LRP1) but not the insulin receptor (IR) in adipocytes, and its overexpression could lead to decreased GLUT4 level. Our results thus indentify PID1 as a critical regulator of glucose metabolism in adipocytes.

Collaboration


Dive into the Lingxia Pang's collaboration.

Top Co-Authors

Avatar

Chenbo Ji

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Xirong Guo

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Chunmei Shi

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Lei Yang

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Ling Chen

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Xianwei Cui

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Fangyan Huang

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Lianghui You

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Guangfeng Xu

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Jiantao Chen

Xinxiang Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge