Network


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

Hotspot


Dive into the research topics where Sheng Zhong Duan is active.

Publication


Featured researches published by Sheng Zhong Duan.


Journal of Clinical Investigation | 2010

Myeloid mineralocorticoid receptor controls macrophage polarization and cardiovascular hypertrophy and remodeling in mice.

Michael G. Usher; Sheng Zhong Duan; Christine Y. Ivaschenko; Ryan A. Frieler; Stefan Berger; Günther Schütz; Richard M. Mortensen

Inappropriate excess of the steroid hormone aldosterone, which is a mineralocorticoid receptor (MR) agonist, is associated with increased inflammation and risk of cardiovascular disease. MR antagonists are cardioprotective and antiinflammatory in vivo, and evidence suggests that they mediate these effects in part by aldosterone-independent mechanisms. Here we have shown that MR on myeloid cells is necessary for efficient classical macrophage activation by proinflammatory cytokines. Macrophages from mice lacking MR in myeloid cells (referred to herein as MyMRKO mice) exhibited a transcription profile of alternative activation. In vitro, MR deficiency synergized with inducers of alternatively activated macrophages (for example, IL-4 and agonists of PPARgamma and the glucocorticoid receptor) to enhance alternative activation. In vivo, MR deficiency in macrophages mimicked the effects of MR antagonists and protected against cardiac hypertrophy, fibrosis, and vascular damage caused by L-NAME/Ang II. Increased blood pressure and heart rates and decreased circadian variation were observed during treatment of MyMRKO mice with L-NAME/Ang II. We conclude that myeloid MR is an important control point in macrophage polarization and that the function of MR on myeloid cells likely represents a conserved ancestral MR function that is integrated in a transcriptional network with PPARgamma and glucocorticoid receptor. Furthermore, myeloid MR is critical for blood pressure control and for hypertrophic and fibrotic responses in the mouse heart and aorta.


Circulation Research | 2008

Peroxisome Proliferator-Activated Receptor-γ–Mediated Effects in the Vasculature

Sheng Zhong Duan; Michael G. Usher; Richard M. Mortensen

Peroxisome proliferator-activated receptor (PPAR)-γ is a nuclear receptor and transcription factor in the steroid superfamily. PPAR-γ agonists, the thiazolidinediones, are clinically used to treat type 2 diabetes. In addition to its function in adipogenesis and increasing insulin sensitivity, PPAR-γ also plays critical roles in the vasculature. In vascular endothelial cells, PPAR-γ activation inhibits endothelial inflammation by suppressing inflammatory gene expression and therefore improves endothelial dysfunction. In vascular smooth muscle cells, PPAR-γ activation inhibits proliferation and migration and promotes apoptosis. In macrophages, PPAR-γ activation suppresses inflammation by regulating gene expression and increases cholesterol uptake and efflux. A recurring theme in many cell types is the modulation of the innate immunity system particularly through altering the activity of the nuclear factor &kgr;B. This system is likely to be even more prominent in modulating disease in vascular cells. The effects of PPAR-γ in the vascular cells translate into the beneficial function of this transcription factor in vascular disorders, including hypertension and atherosclerosis. Both human genetic studies and animal studies using transgenic mice have demonstrated the importance of PPAR-γ in these disorders. However, recent clinical studies have raised significant concerns about the cardiovascular side effects of thiazolidinediones, particularly rosiglitazone. Weighing the potential benefit and harm of PPAR-γ activation and exploring the functional mechanisms may provide a balanced view on the clinical use of these compounds and new approaches to the future therapeutics of vascular disorders associated with diabetes.


Circulation Research | 2005

Cardiomyocyte-Specific Knockout and Agonist of Peroxisome Proliferator–Activated Receptor-γ Both Induce Cardiac Hypertrophy in Mice

Sheng Zhong Duan; Christine Y. Ivashchenko; Mark W. Russell; David S. Milstone; Richard M. Mortensen

Peroxisome proliferator–activated receptor (PPAR)-γ is required for adipogenesis but is also found in the cardiovascular system, where it has been proposed to oppose inflammatory pathways and act as a growth suppressor. PPAR-γ agonists, thiazolidinediones (TZDs), inhibit cardiomyocyte growth in vitro and in pressure overload models. Paradoxically, TZDs also induce cardiac hypertrophy in animal models. To directly determine the role of cardiomyocyte PPAR-γ, we have developed a cardiomyocyte-specific PPAR-γ–knockout (CM-PGKO) mouse model. CM-PGKO mice developed cardiac hypertrophy with preserved systolic cardiac function. Treatment with a TZD, rosiglitazone, induced cardiac hypertrophy in both littermate control mice and CM-PGKO mice and activated distinctly different hypertrophic pathways from CM-PGKO. CM-PGKO mice were found to have increased expression of cardiac embryonic genes (atrial natriuretic peptide and β-myosin heavy chain) and elevated nuclear factor &kgr;B activity in the heart, effects not found by rosiglitazone treatment. Rosiglitazone increased cardiac phosphorylation of p38 mitogen-activated protein kinase independent of PPAR-γ, whereas rosiglitazone induced phosphorylation of extracellular signal–related kinase 1/2 in the heart dependent of PPAR-γ. Phosphorylation of c-Jun N-terminal kinases was not affected by rosiglitazone or CM-PGKO. Surprisingly, despite hypertrophy, Akt phosphorylation was suppressed in CM-PGKO mouse heart. These data show that cardiomyocyte PPAR-γ suppresses cardiac growth and embryonic gene expression and inhibits nuclear factor &kgr;B activity in vivo. Further, rosiglitazone causes cardiac hypertrophy at least partially independent of PPAR-γ in cardiomyocytes and through different mechanisms from CM-PGKO.


Journal of Clinical Investigation | 2007

Hypotension, lipodystrophy, and insulin resistance in generalized PPARγ-deficient mice rescued from embryonic lethality

Sheng Zhong Duan; Christine Y. Ivashchenko; Steven E. Whitesall; Louis G. D’Alecy; Damon Duquaine; Frank C. Brosius; Frank J. Gonzalez; Charles Vinson; Melissa A. Pierre; David S. Milstone; Richard M. Mortensen

We rescued the embryonic lethality of global PPARgamma knockout by breeding Mox2-Cre (MORE) mice with floxed PPARgamma mice to inactivate PPARgamma in the embryo but not in trophoblasts and created a generalized PPARgamma knockout mouse model, MORE-PPARgamma knockout (MORE-PGKO) mice. PPARgamma inactivation caused severe lipodystrophy and insulin resistance; surprisingly, it also caused hypotension. Paradoxically, PPARgamma agonists had the same effect. We showed that another mouse model of lipodystrophy was hypertensive, ruling out the lipodystrophy as a cause. Further, high salt loading did not correct the hypotension in MORE-PGKO mice. In vitro studies showed that the vasculature from MORE-PGKO mice was more sensitive to endothelial-dependent relaxation caused by muscarinic stimulation, but was not associated with changes in eNOS expression or phosphorylation. In addition, vascular smooth muscle had impaired contraction in response to alpha-adrenergic agents. The renin-angiotensin-aldosterone system was mildly activated, consistent with increased vascular capacitance or decreased volume. These effects are likely mechanisms contributing to the hypotension. Our results demonstrated that PPARgamma is required to maintain normal adiposity and insulin sensitivity in adult mice. Surprisingly, genetic loss of PPARgamma function, like activation by agonists, lowered blood pressure, likely through a mechanism involving increased vascular relaxation.


Current Opinion in Nephrology and Hypertension | 2009

Ppars: the vasculature, inflammation and hypertension

Sheng Zhong Duan; Michael G. Usher; Richard M. Mortensen

Purpose of reviewPeroxisome proliferator-activated receptors (PPARs) are a family of nuclear receptors activated by nutrient molecules and their derivatives. Their role has been increasingly recognized to be important in hypertension, metabolic disorders and cardiovascular disease, including atherosclerosis. Control of innate inflammatory processes mostly through alteration of monocyte/macrophage phenotype promises to be a unifying paradigm in understanding the pleiotropic effects of PPAR agonists. Recent findingsAlthough PPAR-γ was the first to be described as an anti-inflammatory agent, both PPAR-α and PPAR-δ are now known to have similar effects as well. Inflammation is an important part of the damage caused by hypertensive diseases. PPARs have now been recognized as important determinants of macrophage polarization. Monocyte precursors of classical and alternatively activated macrophages are being defined as important changes in progression of cardiovascular disease associated with metabolic syndrome including hypertension, hyperlipidemia and obesity. SummaryA major unifying role for PPARs in hypertension and its complications through modification of the innate immune system and inflammation is increasingly likely. PPAR agonists may be beneficial, alone or in combination with other drugs that modify the inflammatory response, in treating hypertension, atherosclerosis and metabolic derangements associated with obesity.


Stroke | 2011

Myeloid-Specific Deletion of the Mineralocorticoid Receptor Reduces Infarct Volume and Alters Inflammation During Cerebral Ischemia

Ryan A. Frieler; He Meng; Sheng Zhong Duan; Stefan Berger; Günther Schütz; Yangdong He; Guohua Xi; Michael M. Wang; Richard M. Mortensen

Background and Purpose— Mineralocorticoid receptor (MR) antagonists have protective effects in rodent models of ischemic stroke, but the cell type-specific actions of these drugs are unknown. In the present study, we examined the contribution of myeloid cell MR during focal cerebral ischemia using myeloid-specific MR knockout mice. Methods— Myeloid-specific MR knockout mice were subjected to transient (90 minutes) middle cerebral artery occlusion followed by 24 hours reperfusion (n=5 to 7 per group). Ischemic cerebral infarcts were identified by hematoxylin and eosin staining and quantified with image analysis software. Immunohistochemical localization of microglia and macrophages was performed using Iba1 staining, and the expression of inflammatory markers was measured after 24 hours of reperfusion by quantitative reverse transcription-polymerase chain reaction. Results— Myeloid-specific MR knockout resulted in a 65% reduction in infarct volume (P=0.005) after middle cerebral artery occlusion. This was accompanied by a significant reduction in activated microglia and macrophages in the ischemic core. Furthermore, myeloid-specific MR knockout suppressed classically activated M1 macrophage markers tumor necrosis factor-&agr;, interleukin-1&bgr;, monocyte chemoattractant protein-1, macrophage inflammatory protein-1&agr;, and interleukin-6 at the same time as partially preserving the induction of alternatively activated, M2, markers Arg1, and Ym1. Conclusions— These data demonstrate that myeloid MR activation exacerbates stroke and identify myeloid MR as a critical target for MR antagonists. Furthermore, these data indicate that MR activation has an important role in controlling immune cell function during the inflammatory response to stroke.


Journal of Biological Chemistry | 2008

In Vivo and in Vitro Studies of a Functional Peroxisome Proliferator-activated Receptor γ Response Element in the Mouse pdx-1 Promoter

Dhananjay Gupta; Thomas L. Jetton; Richard M. Mortensen; Sheng Zhong Duan; Mina Peshavaria; Jack L. Leahy

We reported that peroxisome proliferator-activated receptor γ (PPARγ) transcriptionally regulates the β-cell differentiation factor pancreatic duodenal homeobox (PDX)-1 based on in vitro RNA interference studies. We have now studied mice depleted of PPARγ within the pancreas (PANC PPARγ-/-) created by a Cre/loxP recombinase system, with Cre driven by the pdx-1 promoter. Male PANC PPARγ-/- mice were hyperglycemic at 8 weeks of age (8.1 ± 0.2 mm versus 6.4 ± 0.3 mm, p = 0.009) with islet cytoarchitecture and pancreatic mass of islet β-cells that were indistinguishable from the controls. Islet PDX-1 mRNA (p = 0.001) and protein levels (p = 0.003) were lowered 60 and 40%, respectively, in tandem with impaired glucose-induced insulin secretion and loss of thiazolidinedione-induced increase in PDX-1 expression. We next identified a putative PPAR-response element (PPRE) in the mouse pdx-1 promoter with substantial homology to the corresponding region of the human PDX-1 promoter. Electrophoretic mobility supershift assays with nuclear extracts from β-cell lines and mouse islets, also in vitro translated PPARγ and retinoid X receptor, and chromatin immunoprecipitation analysis demonstrated specific binding of PPARγ and retinoid X receptor to the human and mouse pdx-1 × PPREs. Transient transfection assays of β-cells with reporter constructs of mutated PPREs showed dramatically reduced pdx-1 promoter activity. In summary, we have presented in vivo and in vitro evidence showing PPARγ regulation of pdx-1 transcription in β-cells, plus our results support an important regulatory role for PPARγ in β-cell physiology and thiazolidinedione pharmacology of type 2 diabetes.


Ppar Research | 2008

PPAR-gamma in the Cardiovascular System.

Sheng Zhong Duan; Christine Y. Ivashchenko; Michael G. Usher; Richard M. Mortensen

Peroxisome proliferator-activated receptor-γ (PPAR-γ), an essential transcriptional mediator of adipogenesis, lipid metabolism, insulin sensitivity, and glucose homeostasis, is increasingly recognized as a key player in inflammatory cells and in cardiovascular diseases (CVD) such as hypertension, cardiac hypertrophy, congestive heart failure, and atherosclerosis. PPAR-γ agonists, the thiazolidinediones (TZDs), increase insulin sensitivity, lower blood glucose, decrease circulating free fatty acids and triglycerides, lower blood pressure, reduce inflammatory markers, and reduce atherosclerosis in insulin-resistant patients and animal models. Human genetic studies on PPAR-γ have revealed that functional changes in this nuclear receptor are associated with CVD. Recent controversial clinical studies raise the question of deleterious action of PPAR-γ agonists on the cardiovascular system. These complex interactions of metabolic responsive factors and cardiovascular disease promise to be important areas of focus for the future.


Journal of Clinical Investigation | 2015

EP3 receptor deficiency attenuates pulmonary hypertension through suppression of Rho/TGF-β1 signaling

Ankang Lu; Caojian Zuo; Yuhu He; Guilin Chen; Lingjuan Piao; Jian Zhang; Bing Xiao; Yujun Shen; Juan Tang; Deping Kong; Sara Alberti; Di Chen; Shenkai Zuo; Qianqian Zhang; Shuai Yan; Xiaochun Fei; Fei Yuan; Bin Zhou; Sheng Zhong Duan; Yu Yu; Michael Lazarus; Yunchao Su; Richard M. Breyer; Colin D. Funk; Ying Yu

Pulmonary arterial hypertension (PAH) is commonly associated with chronic hypoxemia in disorders such as chronic obstructive pulmonary disease (COPD). Prostacyclin analogs are widely used in the management of PAH patients; however, clinical efficacy and long-term tolerability of some prostacyclin analogs may be compromised by concomitant activation of the E-prostanoid 3 (EP3) receptor. Here, we found that EP3 expression is upregulated in pulmonary arterial smooth muscle cells (PASMCs) and human distal pulmonary arteries (PAs) in response to hypoxia. Either pharmacological inhibition of EP3 or Ep3 deletion attenuated both hypoxia and monocrotaline-induced pulmonary hypertension and restrained extracellular matrix accumulation in PAs in rodent models. In a murine PAH model, Ep3 deletion in SMCs, but not endothelial cells, retarded PA medial thickness. Knockdown of EP3α and EP3β, but not EP3γ, isoforms diminished hypoxia-induced TGF-β1 activation. Expression of either EP3α or EP3β in EP3-deficient PASMCs restored TGF-β1 activation in response to hypoxia. EP3α/β activation in PASMCs increased RhoA-dependent membrane type 1 extracellular matrix metalloproteinase (MMP) translocation to the cell surface, subsequently activating pro-MMP-2 and promoting TGF-β1 signaling. Activation or disruption of EP3 did not influence PASMC proliferation. Together, our results indicate that EP3 activation facilitates hypoxia-induced vascular remodeling and pulmonary hypertension in mice and suggest EP3 inhibition as a potential therapeutic strategy for pulmonary hypertension.


Journal of Biological Chemistry | 2015

Inhibition of Glutathione Production Induces Macrophage CD36 Expression and Enhances Cellular-oxidized Low Density Lipoprotein (oxLDL) Uptake

Xiaoxiao Yang; Hui Yao; Yuanli Chen; Lei Sun; Yan Li; Xingzhe Ma; Sheng Zhong Duan; Xiaoju Li; Rong Xiang; Jihong Han; Yajun Duan

Background: The GSH-dependent antioxidant system reduces atherosclerosis. Results: Inhibition of GSH production by BSO enhanced CD36 translational efficiency to induce CD36 protein expression and lipid accumulation that was blocked by antioxidant (enzyme). Conclusion: Alterations of cellular GSH and GSH/GSSG status regulate macrophage CD36 expression and cellular oxLDL uptake. Significance: Our study demonstrates an important anti-atherogenic function of the GSH-dependent antioxidant system. The glutathione (GSH)-dependent antioxidant system has been demonstrated to inhibit atherosclerosis. Macrophage CD36 uptakes oxidized low density lipoprotein (oxLDL) thereby facilitating foam cell formation and development of atherosclerosis. It remains unknown if GSH can influence macrophage CD36 expression and cellular oxLDL uptake directly. Herein we report that treatment of macrophages with l-buthionine-S,R-sulfoximine (BSO) decreased cellular GSH production and ratios of GSH to glutathione disulfide (GSH/GSSG) while increasing production of reactive oxygen species. Associated with decreased GSH levels, macrophage CD36 expression was increased, which resulted in enhanced cellular oxLDL uptake. In contrast, N-acetyl cysteine and antioxidant enzyme (catalase or superoxide dismutase) blocked BSO-induced CD36 expression as well as oxLDL uptake. In vivo, administration of mice with BSO increased CD36 expression in peritoneal macrophages and kidneys. BSO had no effect on CD36 mRNA expression and promoter activity but still induced CD36 protein expression in macrophages lacking peroxisome proliferator-activated receptor γ expression, suggesting it induced CD36 expression at the translational level. Indeed, we determined that BSO enhanced CD36 translational efficiency. Taken together, our study demonstrates that cellular GSH levels and GSH/GSSG status can regulate macrophage CD36 expression and cellular oxLDL uptake and demonstrate an important anti-atherogenic function of the GSH-dependent antioxidant system by providing a novel molecular mechanism.

Collaboration


Dive into the Sheng Zhong Duan's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yan Liu

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Ying Yu

Tianjin Medical University

View shared research outputs
Top Co-Authors

Avatar

Bin Zhou

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Chao Li

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Lin-Juan Du

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Xue-Nan Sun

Shanghai Jiao Tong University

View shared research outputs
Researchain Logo
Decentralizing Knowledge