Try a new search

Format these results:

Searched for:

person:miaoq01

in-biosketch:yes

Total Results:

30


Transcription factor 21 accelerates vascular calcification in mice by activating the IL-6/STAT3 signaling pathway and the interplay between VSMCs and ECs

Zhao, Xiao-Kang; Zhu, Meng-Meng; Wang, Sheng-Nan; Zhang, Ting-Ting; Wei, Xiao-Ning; Wang, Cheng-Yi; Zheng, Juan; Zhu, Wen-Ya; Jiang, Mei-Xiu; Xu, Suo-Wen; Yang, Xiao-Xiao; Duan, Ya-Jun; Zhang, Bu-Chun; Han, Ji-Hong; Miao, Qing R; Hu, Hao; Chen, Yuan-Li
Vascular calcification is caused by the deposition of calcium salts in the intimal or tunica media layer of the aorta, which increases the risk of cardiovascular events and all-cause mortality. However, the mechanisms underlying vascular calcification are not fully clarified. Recently it has been shown that transcription factor 21 (TCF21) is highly expressed in human and mouse atherosclerotic plaques. In this study we investigated the role of TCF21 in vascular calcification and the underlying mechanisms. In carotid artery atherosclerotic plaques collected from 6 patients, we found that TCF21 expression was upregulated in calcific areas. We further demonstrated TCF21 expression was increased in an in vitro vascular smooth muscle cell (VSMC) osteogenesis model. TCF21 overexpression promoted osteogenic differentiation of VSMC, whereas TCF21 knockdown in VSMC attenuated the calcification. Similar results were observed in ex vivo mouse thoracic aorta rings. Previous reports showed that TCF21 bound to myocardin (MYOCD) to inhibit the transcriptional activity of serum response factor (SRF)-MYOCD complex. We found that SRF overexpression significantly attenuated TCF21-induced VSMC and aortic ring calcification. Overexpression of SRF, but not MYOCD, reversed TCF21-inhibited expression of contractile genes SMA and SM22. More importantly, under high inorganic phosphate (3 mM) condition, SRF overexpression reduced TCF21-induced expression of calcification-related genes (BMP2 and RUNX2) as well as vascular calcification. Moreover, TCF21 overexpression enhanced IL-6 expression and downstream STAT3 activation to facilitate vascular calcification. Both LPS and STAT3 could induce TCF21 expression, suggesting that the inflammation and TCF21 might form a positive feedback loop to amplify the activation of IL-6/STAT3 signaling pathway. On the other hand, TCF21 induced production of inflammatory cytokines IL-1β and IL-6 in endothelial cells (ECs) to promote VSMC osteogenesis. In EC-specific TCF21 knockout (TCF21ECKO) mice, VD3 and nicotine-induced vascular calcification was significantly reduced. Our results suggest that TCF21 aggravates vascular calcification by activating IL-6/STAT3 signaling and interplay between VSMC and EC, which provides new insights into the pathogenesis of vascular calcification. TCF21 enhances vascular calcification by activating the IL-6-STAT3 signaling pathway. TCF21 inhibition may be a new potential therapeutic strategy for the prevention and treatment of vascular calcification.
PMCID:10374894
PMID: 36997664
ISSN: 1745-7254
CID: 5594902

Nogo-B receptor increases glycolysis and the paclitaxel resistance of estrogen receptor-positive breast cancer via the HIF-1α-dependent pathway

Liu, Chang; Li, Sijie; Zhang, Xiaoxiao; Jin, Chunxiang; Zhao, Baofeng; Li, Liying; Miao, Qing Robert; Jin, Ying; Fan, Zhimin
Chemotherapy can improve the prognosis and overall survival of breast cancer patients, but chemoresistance continues a major problem in clinical. Most breast cancer is estrogen receptor (ER) positive but responds less to neoadjuvant or adjuvant chemotherapy than ER-negative breast cancer. The Nogo-B receptor (NgBR) increases the chemoresistance of ER-positive breast cancer by facilitating oncogene signaling pathways. Here, we further investigated the potential role of NgBR as a novel target to overcome glycolysis-dependent paclitaxel resistance in ER-positive breast cancer. NgBR knockdown inhibited glycolysis and promoted paclitaxel-induced apoptosis by attenuating HIF-1α expression in ER-positive breast cancer cells via NgBR-mediated estrogen receptor alpha (ERα)/hypoxia-inducible factor-1 alpha (HIF-1α) and nuclear factor-kappa B subunit (NF-κB)/HIF-1α signaling pathways. A ChIP assay further confirmed that NgBR overexpression not only facilitates ERα binding to HIF-1α and GLUT1 genes but also promotes HIF-1α binding to GLUT1, HK2, and LDHA genes, which further promotes glycolysis and induces paclitaxel resistance. In conclusion, our study suggests that NgBR expression is essential for maintaining the metabolism and paclitaxel resistance of ER-positive breast cancer, and the NgBR can be a new therapeutic target for improving chemoresistance in ER-positive breast cancer.
PMID: 36241702
ISSN: 1476-5500
CID: 5352232

Correction to: Nogo-B receptor increases glycolysis and the paclitaxel resistance of estrogen receptor-positive breast cancer via the HIF-1α-dependent pathway

Liu, Chang; Li, Sijie; Zhang, Xiaoxiao; Jin, Chunxiang; Zhao, Baofeng; Li, Liying; Miao, Qing Robert; Jin, Ying; Fan, Zhimin
PMID: 36376422
ISSN: 1476-5500
CID: 5384762

Overexpression of NgBR inhibits high-fat diet-induced atherosclerosis in ApoE-deficiency mice

Gong, Ke; Wang, Mengyao; Wang, Dandan; Gao, Yongyao; Ma, Likun; Yang, Xiaoxiao; Zhu, Xinran; Chen, Shasha; Zhang, Mengxue; Li, Huaxin; Chen, Yuanli; Hu, Wenquan; Miao, Qing R; Iwakiri, Yasuko; Liao, Chenzhong; Duan, Yajun; Han, Jihong
BACKGROUND:Hyperlipidemia (hypercholesterolemia and/or hypertriglyceridemia) is a risk factor for atherosclerosis. Nogo-B receptor (NgBR) plays important roles in hepatic steatosis and cholesterol transport. However, the effect of NgBR overexpression on atherosclerosis remains unknown. MATERIALS AND METHODS:Apolipoprotein E deficient (ApoE-/-) mice infected with adeno-associated virus (AAV)-NgBR expression vector were fed a high-fat diet for 12 weeks, followed by determination of atherosclerosis and the involved mechanisms. RESULTS:We determined that high expression of NgBR by AAV injection mainly occurs in the liver and it can substantially inhibit en face and aortic root sinus lesions. NgBR overexpression also reduced levels of inflammatory factors in the aortic root and serum, and levels of cholesterol, triglyceride, and free fatty acids in the liver and serum. Mechanistically, NgBR overexpression increased the expression of scavenger receptor type BI and the genes for bile acid synthesis, and decreased the expression of cholesterol synthesis genes by reducing sterol regulatory element-binding protein 2 maturation in the liver, thereby reducing hypercholesterolemia. In addition, NgBR overexpression activated AMP-activated protein kinase α via the Ca2+ signaling pathway, which inhibited fat synthesis and improved hypertriglyceridemia. CONCLUSIONS:Taken together, our study demonstrates that overexpression of NgBR enhanced cholesterol metabolism and inhibited cholesterol/fatty acid synthesis to reduce hyperlipidemia, and reduced vascular inflammation, thereby inhibiting atherosclerosis in ApoE-/- mice. Our study indicates that NgBR might be a potential target for atherosclerosis treatment.
PMCID:10069848
PMID: 36996002
ISSN: 2471-254x
CID: 5468402

Overexpression of NgBR inhibits high-fat diet-induced atherosclerosis in ApoE-deficiency mice

Gong, Ke; Wang, Mengyao; Wang, Dandan; Gao, Yongyao; Ma, Likun; Yang, Xiaoxiao; Zhu, Xinran; Chen, Shasha; Zhang, Mengxue; Li, Huaxin; Chen, Yuanli; Hu, Wenquan; Miao, Qing R.; Iwakiri, Yasuko; Liao, Chenzhong; Duan, Yajun; Han, Jihong
ISI:001159566400001
CID: 5636612

NOGOB receptor deficiency increases cerebrovascular permeability and hemorrhage via impairing histone acetylation-mediated CCM1/2 expression

Fang, Zhi; Sun, Xiaoran; Wang, Xiang; Ma, Ji; Palaia, Thomas; Rana, Ujala; Miao, Benjamin; Ragolia, Louis; Hu, Wenquan; Miao, Qing Robert
The loss function of cerebral cavernous malformation (CCM) genes leads to most CCM lesions characterized by enlarged leaking vascular lesions in the brain. Although we previously showed that NOGOB receptor (NGBR) knockout in endothelial cells (ECs) results in cerebrovascular lesions in the mouse embryo, the molecular mechanism by which NGBR regulates CCM1/2 expression has not been elucidated. Here, we show that genetic depletion of Ngbr in ECs at both postnatal and adult stages results in CCM1/2 expression deficiency and cerebrovascular lesions such as enlarged vessels, blood-brain-barrier hyperpermeability, and cerebral hemorrhage. To reveal the molecular mechanism, we used RNA-sequencing analysis to examine changes in the transcriptome. Surprisingly, we found that the acetyltransferase HBO1 and histone acetylation were downregulated in NGBR-deficient ECs. The mechanistic studies elucidated that NGBR is required for maintaining the expression of CCM1/2 in ECs via HBO1-mediated histone acetylation. ChIP-qPCR data further demonstrated that loss of NGBR impairs the binding of HBO1 and acetylated histone H4K5 and H4K12 on the promotor of the CCM1 and CCM2 genes. Our findings on epigenetic regulation of CCM1 and CCM2 that is modulated by NGBR and HBO1-mediated histone H4 acetylation provide a perspective on the pathogenesis of sporadic CCMs.
PMCID:9057619
PMID: 35316220
ISSN: 1558-8238
CID: 5215592

Autoimmunity to Annexin A2 predicts mortality among hospitalised COVID-19 patients [Letter]

Zuniga, Marisol; Gomes, Claudia; Carsons, Steven E; Bender, Michael T; Cotzia, Paolo; Miao, Qing Robert; Lee, David C; Rodriguez, Ana
PMID: 34244321
ISSN: 1399-3003
CID: 4938032

NOGOB receptor-mediated RAS signaling pathway is a target for suppressing proliferating hemangioma

Hu, Wenquan; Liu, Zhong; Salato, Valerie; North, Paula E; Bischoff, Joyce; Kumar, Suresh N; Fang, Zhi; Rajan, Sujith; Hussain, M Mahmood; Miao, Qing R
Infantile hemangioma is a vascular tumor characterized by the rapid growth of disorganized blood vessels followed by slow spontaneous involution. The underlying molecular mechanisms that regulate hemangioma proliferation and involution still are not well elucidated. Our previous studies reported that NOGOB receptor (NGBR), a transmembrane protein, is required for the translocation of prenylated RAS from the cytosol to the plasma membrane and promotes RAS activation. Here, we show that NGBR was highly expressed in the proliferating phase of infantile hemangioma, but its expression decreased in the involuting phase, suggesting that NGBR may have been involved in regulating the growth of proliferating hemangioma. Moreover, we demonstrate that NGBR knockdown in hemangioma stem cells (HemSCs) attenuated growth factor-stimulated RAS activation and diminished the migration and proliferation of HemSCs, which is consistent with the effects of RAS knockdown in HemSCs. In vivo differentiation assay further shows that NGBR knockdown inhibited blood vessel formation and adipocyte differentiation of HemSCs in immunodeficient mice. Our data suggest that NGBR served as a RAS modulator in controlling the growth and differentiation of HemSCs.
PMCID:7934876
PMID: 33400686
ISSN: 2379-3708
CID: 5479752

NGBR is required to ameliorate type 2 diabetes in mice by enhancing insulin sensitivity

Chen, Yi; Hu, Wenquan; Li, Qi; Zhao, Shiwei; Zhao, Dan; Zhang, Shuang; Wei, Zhuo; Yang, Xiaoxiao; Chen, Yuanli; Li, Xiaoju; Liao, Chenzhong; Han, Jihong; Miao, Qing Robert; Duan, Yajun
The reduction of insulin resistance or improvement of insulin sensitivity is the most effective treatment for type 2 diabetes (T2D). We previously reported that Nogo-B receptor (NGBR), encoded by the NUS1 gene, is required for attenuating hepatic lipogenesis by blocking nuclear translocation of liver X receptor alpha, suggesting its important role in regulating hepatic lipid metabolism. Herein, we demonstrate that NGBR expression was decreased in the liver of obesity-associated T2D patients and db/db mice. NGBR knockout in mouse hepatocytes resulted in increased blood glucose, insulin resistance, and beta-cell loss. High-fat diet (HFD)/streptozotocin (STZ)-treated mice presented the T2D phenotype by showing increased nonesterified fatty acid (NEFA) and triglyceride (TG) in the liver and plasma and increased insulin resistance and beta-cell loss. AAV-mediated NGBR overexpression in the liver reduced NEFA and TG in the liver and circulation and improved liver functions. Consequently, HFD/STZ-treated mice with hepatic NGBR overexpression had increased insulin sensitivity and reduced beta-cell loss. Mechanistically, NGBR overexpression restored insulin signaling of AMPKα1-dependent phosphorylation of AKT and GSK3β. NGBR overexpression also reduced expression of endoplasmic reticulum stress-associated genes in the liver and skeletal muscle to improve insulin sensitivity. Together, our results reveal that NGBR is required to ameliorate T2D in mice, providing new insight into the role of hepatic NGBR in insulin sensitivity and T2D treatment.
PMCID:8111265
PMID: 33812996
ISSN: 1083-351x
CID: 5033122

Nogo-B receptor is required for stabilizing TGF-β type I receptor and promotes the TGF-β1-induced epithelial-to-mesenchymal transition of non-small cell lung cancer

Wu, Donghua; Zhao, Baofeng; Song, Yang; Chi, Xinming; Fu, Hailu; Guan, Tiantong; Zhang, Liyuan; Yang, Xueguang; Hu, Ke; Huang, Rong; Jin, Xiaomeng; Miao, Qing Robert; Shao, Shujuan
PMCID:7778533
PMID: 33403029
ISSN: 1837-9664
CID: 5033112