Network


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

Hotspot


Dive into the research topics where Yi Si is active.

Publication


Featured researches published by Yi Si.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2012

Protein Kinase C-Delta Mediates Adventitial Cell Migration Through Regulation of Monocyte Chemoattractant Protein-1 Expression in a Rat Angioplasty Model

Yi Si; Jun Ren; Pu Wang; Debra L. Rateri; Alan Daugherty; Xudong Shi; K. Craig Kent; Bo Liu

Objective—The adventitia is increasingly recognized as an important player during the development of intimal hyperplasia. However, the mechanism of adventitial cell recruitment to the subintimal space remains largely undefined. We have shown previously that gene transfer of protein kinase C-delta (PKC&dgr;) increases apoptosis of smooth muscle cells following balloon injury. In the current study, we investigated a potential role of PKC&dgr; in regulating the recruitment of adventitial cells. Methods and Results—Conditioned media from PKC&dgr;-overexpressing smooth muscle cells stimulated migration and CCR2 expression of adventitial fibroblasts through a MCP-1 dependent mechanism. Following balloon injury of rat carotid arteries, overexpression of PKC&dgr; in smooth muscle cells significantly increased MCP-1 and CCR2 expression and the number of adventitia-originated cells detected in the neointima. Administration of an anti-MCP–1 antibody markedly diminished the recruitment of adventitial cells. Combined PKC&dgr; overexpression and anti-MCP–1 inhibited intimal hyperplasia more effectively than either approach alone. Conclusion—Our data suggest that PKC&dgr; regulates recruitment of adventitial cells to the neointima via a mechanism involving upregulation of the MCP-1/CCR2 signaling axis in injured arteries. Blockage of MCP-1 while enhancing apoptosis may serve as a potential therapeutic strategy to attenuate intimal hyperplasia.


PLOS ONE | 2014

Periadventitial application of rapamycin-loaded nanoparticles produces sustained inhibition of vascular restenosis.

Xudong Shi; Guojun Chen; Lian-Wang Guo; Yi Si; Men Zhu; Srikanth Pilla; Bo Liu; Shaoqin Gong; K. Craig Kent

Open vascular reconstructions frequently fail due to the development of recurrent disease or intimal hyperplasia (IH). This paper reports a novel drug delivery method using a rapamycin-loaded poly(lactide-co-glycolide) (PLGA) nanoparticles (NPs)/pluronic gel system that can be applied periadventitially around the carotid artery immediately following the open surgery. In vitro studies revealed that rapamycin dispersed in pluronic gel was rapidly released over 3 days whereas release of rapamycin from rapamycin-loaded PLGA NPs embedded in pluronic gel was more gradual over 4 weeks. In cultured rat vascular smooth muscle cells (SMCs), rapamycin-loaded NPs produced durable (14 days versus 3 days for free rapamycin) inhibition of phosphorylation of S6 kinase (S6K1), a downstream target in the mTOR pathway. In a rat balloon injury model, periadventitial delivery of rapamycin-loaded NPs produced inhibition of phospho-S6K1 14 days after balloon injury. Immunostaining revealed that rapamycin-loaded NPs reduced SMC proliferation at both 14 and 28 days whereas rapamycin alone suppressed proliferation at day 14 only. Moreover, rapamycin-loaded NPs sustainably suppressed IH for at least 28 days following treatment, whereas rapamycin alone produced suppression on day 14 with rebound of IH by day 28. Since rapamycin, PLGA, and pluronic gel have all been approved by the FDA for other human therapies, this drug delivery method could potentially be translated into human use quickly to prevent failure of open vascular reconstructions.


Journal of Biological Chemistry | 2014

Protein Kinase C-δ (PKCδ) Regulates Proinflammatory Chemokine Expression through Cytosolic Interaction with the NF-κB Subunit p65 in Vascular Smooth Muscle Cells

Jun Ren; Qiwei Wang; Stephanie Morgan; Yi Si; Aarthi Ravichander; Changlin Dou; K. Craig Kent; Bo Liu

Background: Proinflammatory chemokines released by vascular smooth muscle cells (VSMCs) play a critical role in vascular inflammation. Results: Promoting protein kinase C-δ (PKCδ) translocation or inhibition of NF-κB pathway diminishes proinflammatory chemokine production. Conclusion: PKCδ regulates proinflammatory chemokine expression through cytosolic interaction with the NF-κB subunit p65, thus modulating inflammation. Significance: Learning how PKCδ regulates proinflammatory chemokine expression is crucial for understanding vascular inflammation. Proinflammatory chemokines released by vascular smooth muscle cells (VSMCs) play a critical role in vascular inflammation. Protein kinase C-δ (PKCδ) has been shown to be up-regulated in VSMCs of injured arteries. PKCδ knock-out (Prkcd−/−) mice are resistant to inflammation as well as apoptosis in models of abdominal aortic aneurysm. However, the precise mechanism by which PKCδ modulates inflammation remains incompletely understood. In this study, we identified four inflammatory chemokines (Ccl2/Mcp-1, Ccl7, Cxcl16, and Cx3cl1) of over 45 PKCδ-regulated genes associated with inflammatory response by microarray analysis. Using CCL2 as a prototype, we demonstrated that PKCδ stimulated chemokine expression at the transcriptional level. Inhibition of the NF-κB pathway or siRNA knockdown of subunit p65, but not p50, eliminated the effect of PKCδ on Ccl2 expression. Overexpressing PKCδ followed by incubation with phorbol 12-myristate 13-acetate resulted in an increase in p65 Ser-536 phosphorylation and enhanced DNA binding affinity without affecting IκB degradation or p65 nuclear translocation. Prkcd gene deficiency impaired p65 Ser-536 phosphorylation and DNA binding affinity in response to TNFα. Results from in situ proximity ligation analysis and co-immunoprecipitation performed on cultured VSMCs and aneurysmal aorta demonstrated physical interaction between PKCδ and p65 that took place largely outside the nucleus. Promoting nuclear translocation of PKCδ with peptide ψδRACK diminished Ccl2 production, whereas inhibition of PKCδ translocation with peptide δV1-1 enhanced Ccl2 expression. Together, these results suggest that PKCδ modulates inflammation at least in part through the NF-κB-mediated chemokines. Mechanistically, PKCδ activates NF-κB through an IκB-independent cytosolic interaction, which subsequently leads to enhanced p65 phosphorylation and DNA binding affinity.


American Journal of Physiology-heart and Circulatory Physiology | 2012

TGF-β and Smad3 modulate PI3K/Akt signaling pathway in vascular smooth muscle cells

Pasithorn A. Suwanabol; Stephen Seedial; Fan Zhang; Xudong Shi; Yi Si; Bo Liu; K. Craig Kent

Transforming growth factor-β (TGF-β) is upregulated at the time of arterial injury; however, the mechanism through which TGF-β enhances the development of intimal hyperplasia is not clear. Recent studies from our laboratory suggest that in the presence of elevated levels of Smad3, TGF-β stimulates smooth muscle cell (SMC) proliferation. This is a novel phenomenon in that TGF-β has traditionally been known as a potent inhibitor of cellular proliferation. In these studies we explore the signaling pathways through which TGF-β mediates its proliferative effect in vascular SMCs. We found that TGF-β phosphorylates and activates Akt in a time-dependent manner, and this effect is significantly enhanced by overexpression of Smad3. Furthermore, both chemical and molecular inhibition of Smad3 can reverse the effect of TGF-β on Akt. Although we found numerous signaling pathways that might function as intermediates between Smad3 and Akt, p38 appeared the most promising. Overexpression of Smad3 enhanced p38 phosphorylation and inhibition of p38 with a chemical inhibitor or a small interfering RNA blocked TGF-β-induced Akt phosphorylation. Moreover, TGF-β/Smad3 enhancement of SMC proliferation was blocked by inhibition of p38. Phosphorylation of Akt by TGF-β/Smad3 was not dependent on gene expression or protein synthesis, and immunoprecipitation studies revealed a physical association among p38, Akt, and Smad3 suggesting that activation requires a direct protein-protein interaction. Our findings were confirmed in vivo where overexpression of Smad3 in a rat carotid injury model led to enhancement of p-p38, p-Akt, as well as SMC proliferation. Furthermore, inhibition of p38 in vivo led to decreased Akt phosphorylation and SMC proliferation. In summary, our studies reveal a novel pathway whereby TGF-β/Smad3 stimulates SMC proliferation through p38 and Akt. These findings provide a potential mechanism for the substantial effect of TGF-β on intimal hyperplasia and suggest new targets for chemical or molecular prevention of vascular restenosis.


Cell Death and Disease | 2014

TGF-β/Smad3 inhibit vascular smooth muscle cell apoptosis through an autocrine signaling mechanism involving VEGF-A

Xu Dong Shi; Lian-Wang Guo; Stephen Seedial; Yi Si; Bowen Wang; Toshio Takayama; Pasithorn A. Suwanabol; S. Ghosh; Daniel DiRenzo; Bo Liu; K.C. Kent

We have previously shown that in the presence of elevated Smad3, transforming growth factor-β (TGF-β) transforms from an inhibitor to a stimulant of vascular smooth muscle cell (SMC) proliferation and intimal hyperplasia (IH). Here we identify a novel mechanism through which TGF-β/Smad3 also exacerbates IH by inhibiting SMC apoptosis. We found that TGF-β treatment led to inhibition of apoptosis in rat SMCs following viral expression of Smad3. Conditioned media from these cells when applied to naive SMCs recapitulated this effect, suggesting an autocrine pathway through a secreted factor. Gene array of TGF-β/Smad3-treated cells revealed enhanced expression of vascular endothelial growth factor (VEGF), a known inhibitor of endothelial cell apoptosis. We then evaluated whether VEGF is the secreted mediator responsible for TGF-β/Smad3 inhibition of SMC apoptosis. In TGF-β/Smad3-treated cells, VEGF mRNA and protein as well as VEGF secretion were increased. Moreover, recombinant VEGF-A inhibited SMC apoptosis and a VEGF-A-neutralizing antibody reversed the inhibitory effect of conditioned media on SMC apoptosis. Stimulation of SMCs with TGF-β led to the formation of a complex of Smad3 and hypoxia-inducible factor-1α (HIF-1α) that in turn activated the VEGF-A promoter and transcription. In rat carotid arteries following arterial injury, Smad3 and VEGF-A expression were upregulated. Moreover, Smad3 gene transfer further enhanced VEGF expression as well as inhibited SMC apoptosis. Finally, blocking either the VEGF receptor or Smad3 signaling in injured carotid arteries abrogated the inhibitory effect of Smad3 on vascular SMC apoptosis. Taken together, our study reveals that following angioplasty, elevation of both TGF-β and Smad3 leads to SMC secretion of VEGF-A that functions as an autocrine inhibitor of SMC apoptosis. This novel pathway provides further insights into the role of TGF-β in the development of IH.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2012

Elevated Protein Kinase C-δ Contributes to Aneurysm Pathogenesis Through Stimulation of Apoptosis and Inflammatory Signaling

Stephanie Morgan; Dai Yamanouchi; Calvin Harberg; Qiwei Wang; Melissa R. Keller; Yi Si; William J. Burlingham; Stephen Seedial; Justin Lengfeld; Bo Liu

Objective—Apoptosis of smooth muscle cells (SMCs) is a prominent pathological characteristic of abdominal aortic aneurysm (AAA). We have previously shown that SMC apoptosis stimulates proinflammatory signaling in a mouse model of AAA. Here, we test whether protein kinase C-&dgr; (PKC&dgr;), an apoptotic mediator, participates in the pathogenesis of AAA by regulating apoptosis and proinflammatory signals. Methods and Results—Mouse experimental AAA is induced by perivascular administration of CaCl2. Mice deficient in PKC&dgr; exhibit a profound reduction in aneurysmal expansion, SMC apoptosis, and transmural inflammation as compared with wild-type littermates. Delivery of PKC&dgr; to the aortic wall of PKC&dgr;–/– mice restores aneurysm, whereas overexpression of a dominant negative PKC&dgr; mutant in the aorta of wild-type mice attenuates aneurysm. In vitro, PKC&dgr;–/– aortic SMCs exhibit significantly impaired monocyte chemoattractant protein-1 production. Ectopic administration of recombinant monocyte chemoattractant protein-1 to the arterial wall of PKC&dgr;–/– mice restores inflammatory response and aneurysm development. Conclusion—PKC&dgr; is an important signaling mediator for SMC apoptosis and inflammation in a mouse model of AAA. By stimulating monocyte chemoattractant protein-1 expression in aortic SMCs, upregulated PKC&dgr; exacerbates the inflammatory process, in turn perpetuating elastin degradation and aneurysmal dilatation. Inhibition of PKC&dgr; may serve as a potential therapeutic strategy for AAA.


Biomacromolecules | 2015

Polymer Multilayers that Promote the Rapid Release and Contact Transfer of DNA

Yan Yu; Yi Si; Shane L. Bechler; Bo Liu; David M. Lynn

We report a layer-by-layer approach to the fabrication of thin polymer-based multilayers that release DNA rapidly in physiologically relevant environments. This approach exploits the properties of a weak anionic polyelectrolyte [poly(acrylic acid); PAA] to disrupt ionic interactions and promote disassembly in coatings that otherwise erode slowly. We investigated this approach using multilayers fabricated from plasmid DNA and linear poly(ethylenimine) (LPEI), a model synthetic cationic polymer used widely for DNA delivery. LPEI/DNA multilayers erode and release DNA slowly over ∼4 days when incubated in PBS buffer. In contrast, substitution of every other layer of DNA with PAA lead to thin films that released DNA rapidly, with >60% being released in the first 5 min. These quick-release coatings release bioactive DNA and can be used to fabricate uniform coatings on a variety of objects, including the tips of inflatable balloon catheters. We demonstrate that these coatings can promote high levels of cell transfection in vitro and the robust contact transfer and expression of DNA in vascular tissue in vivo using a rat model of vascular injury. These materials provide useful alternatives to multilayers and other coatings that promote the prolonged release of DNA. More broadly, approaches that depart from the use of degradable polymers to promote film erosion create opportunities to design new gene delivery coatings using a broader range of polymer-based building blocks designed for other gene delivery applications. With further development, this approach could thus provide a new and useful platform for the rapid contact transfer of DNA to cells and tissues of interest in a range of fundamental and applied contexts.


Stem Cells | 2016

Local CXCR4 Upregulation in the Injured Arterial Wall Contributes to Intimal Hyperplasia

Xudong Shi; Lian-Wang Guo; Stephen Seedial; Toshio Takayama; Bowen Wang; Mengxue Zhang; Sarah Franco; Yi Si; Mirnal A. Chaudhary; Bo Liu; K. Craig Kent

CXCR4 is a stem/progenitor cell surface receptor specific for the cytokine stromal cell‐derived factor‐1 (SDF‐1α). There is evidence that bone marrow‐derived CXCR4‐expressing cells contribute to intimal hyperplasia (IH) by homing to the arterial subintima which is enriched with SDF‐1α. We have previously found that transforming growth factor‐β (TGFβ) and its signaling protein Smad3 are both upregulated following arterial injury and that TGFβ/Smad3 enhances the expression of CXCR4 in vascular smooth muscle cells (SMCs). It remains unknown, however, whether locally induced CXCR4 expression in SM22 expressing vascular SMCs plays a role in neointima formation. Here, we investigated whether elevated TGFβ/Smad3 signaling leads to the induction of CXCR4 expression locally in the injured arterial wall, thereby contributing to IH. We found prominent CXCR4 upregulation (mRNA, 60‐fold; protein, 4‐fold) in TGFβ‐treated, Smad3‐expressing SMCs. Chromatin immunoprecipitation assays revealed a specific association of the transcription factor Smad3 with the CXCR4 promoter. TGFβ/Smad3 treatment also markedly enhanced SDF‐1α‐induced ERK1/2 phosphorylation as well as SMC migration in a CXCR4‐dependent manner. Adenoviral expression of Smad3 in balloon‐injured rat carotid arteries increased local CXCR4 levels and enhanced IH, whereas SMC‐specific depletion of CXCR4 in the wire‐injured mouse femoral arterial wall produced a 60% reduction in IH. Our results provide the first evidence that upregulation of TGFβ/Smad3 in injured arteries induces local SMC CXCR4 expression and cell migration, and consequently IH. The Smad3/CXCR4 pathway may provide a potential target for therapeutic interventions to prevent restenosis. Stem Cells 2016;34:2744–2757


Biomaterials | 2013

Reduction of intimal hyperplasia in injured rat arteries promoted by catheter balloons coated with polyelectrolyte multilayers that contain plasmid DNA encoding PKCδ.

Shane L. Bechler; Yi Si; Yan Yu; Jun Ren; Bo Liu; David M. Lynn


Arteriosclerosis, Thrombosis, and Vascular Biology | 2014

Abstract 160: Perivascular Administration of Rapamycin Through Nanoparticles Prolongs Inhibition of Arterial Restenosis

Guojun Chen; Xudong Shi; Lian-Wang Guo; Yi Si; Men Zhu; Srikanth Pilla; Bo Liu; Shaoqin Gong; K. Craig Kent

Collaboration


Dive into the Yi Si's collaboration.

Top Co-Authors

Avatar

Bo Liu

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

K. Craig Kent

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Xudong Shi

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Jun Ren

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Stephen Seedial

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

K.C. Kent

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Lian-Wang Guo

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Dai Yamanouchi

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Justin Lengfeld

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Pasithorn A. Suwanabol

University of Wisconsin-Madison

View shared research outputs
Researchain Logo
Decentralizing Knowledge