Network


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

Hotspot


Dive into the research topics where Yung-Chih Chen is active.

Publication


Featured researches published by Yung-Chih Chen.


Circulation Research | 2009

Dissociation of Pentameric to Monomeric C-Reactive Protein on Activated Platelets Localizes Inflammation to Atherosclerotic Plaques

Steffen U. Eisenhardt; Jonathon Habersberger; Andrew J. Murphy; Yung-Chih Chen; Kevin J. Woollard; Nicole Bassler; Hongwei Qian; Constantin von zur Muhlen; Christoph E. Hagemeyer; Ingo Ahrens; Jaye Chin-Dusting; Alex Bobik; Karlheinz Peter

C-reactive protein (CRP) is a predictor of cardiovascular risk. It circulates as a pentamer (pentameric CRP) in plasma. The in vivo existence of monomeric (m)CRP has been postulated, but its function and source are not clear. We show that mCRP is deposited in human aortic and carotid atherosclerotic plaques but not in healthy vessels. pCRP is found neither in healthy nor in diseased vessels. As source of mCRP, we identify a mechanism of dissociation of pCRP to mCRP. We report that activated platelets, which play a central role in cardiovascular events, mediate this dissociation via lysophosphatidylcholine, which is present on activated but not resting platelets. Furthermore, the dissociation of pCRP to mCRP can also be mediated by apoptotic monocytic THP-1 and Jurkat T cells. The functional consequence is the unmasking of proinflammatory effects of CRP as demonstrated in experimental settings that are pathophysiologically relevant for atherogenesis: compared to pCRP, mCRP induces enhanced monocyte chemotaxis; monocyte activation, as determined by conformational change of integrin Mac-1; generation of reactive oxygen species; and monocyte adhesion under static and physiological flow conditions. In conclusion, we demonstrate mCRP generation via pCRP dissociation on activated platelets and H2O2-treated apoptotic THP-1 and Jurkat T cells, thereby identifying a mechanism of localized unmasking of the proinflammatory properties of CRP. This novel mechanism provides a potential link between the established cardiovascular risk marker, circulating pCRP, and localized platelet-mediated inflammatory and proatherogenic effects.


Circulation Research | 2013

A Novel Mouse Model of Atherosclerotic Plaque Instability for Drug Testing and Mechanistic/Therapeutic Discoveries Using Gene and MicroRNA Expression Profiling

Yung-Chih Chen; Anh Bui; Jeannine Diesch; Richard Manasseh; Christian Hausding; Jennifer Rivera; Izhak Haviv; Alex Agrotis; Nay Min Htun; Jeremy B. M. Jowett; Christoph E. Hagemeyer; Ross D. Hannan; Alex Bobik; Karlheinz Peter

Rationale: The high morbidity/mortality of atherosclerosis is typically precipitated by plaque rupture and consequent thrombosis. However, research on underlying mechanisms and therapeutic approaches is limited by the lack of animal models that reproduce plaque instability observed in humans. Objective: Development and use of a mouse model of plaque rupture that reflects the end stage of human atherosclerosis. Methods and Results: On the basis of flow measurements and computational fluid dynamics, we applied a tandem stenosis to the carotid artery of apolipoprotein E–deficient mice on high-fat diet. At 7 weeks postoperatively, we observed intraplaque hemorrhage in ≈50% of mice, as well as disruption of fibrous caps, intraluminal thrombosis, neovascularization, and further characteristics typically seen in human unstable plaques. Administration of atorvastatin was associated with plaque stabilization and downregulation of monocyte chemoattractant protein-1 and ubiquitin. Microarray profiling of mRNA and microRNA (miR) and, in particular, its combined analysis demonstrated major differences in the hierarchical clustering of genes and miRs among nonatherosclerotic arteries, stable, and unstable plaques and allows the identification of distinct genes/miRs, potentially representing novel therapeutic targets for plaque stabilization. The feasibility of the described animal model as a discovery tool was established in a pilot approach, identifying a disintegrin and metalloprotease with thrombospondin motifs 4 (ADAMTS4) and miR-322 as potential pathogenic factors of plaque instability in mice and validated in human plaques. Conclusions: The newly described mouse model reflects human atherosclerotic plaque instability and represents a discovery tool toward the development and testing of therapeutic strategies aimed at preventing plaque rupture. Distinctly expressed genes and miRs can be linked to plaque instability.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2014

Protective Role for Toll-Like Receptor-9 in the Development of Atherosclerosis in Apolipoprotein E–Deficient Mice

Christine Koulis; Yung-Chih Chen; Christian Hausding; Ingo Ahrens; Tin Kyaw; Christopher Tay; Terri J. Allen; Karin Jandeleit-Dahm; Matthew J. Sweet; Shizuo Akira; Alex Bobik; Karlheinz Peter; Alexander Agrotis

Objective— Atherosclerosis is driven by inflammatory reactions that are shared with the innate immune system. Toll-like receptor-9 (TLR9) is an intracellular pattern recognition receptor of the innate immune system that is currently under clinical investigation as a therapeutic target in inflammatory diseases. Here, we investigated whether TLR9 has a role in the development of atherosclerosis in apolipoprotein E–deficient (ApoE−/−) mice. Approach and Results— Newly generated double-knockout ApoE−/−:TLR9−/− mice and control ApoE−/− mice were fed a high-fat diet from 8 weeks and effects on lesion size, cellular composition, inflammatory status, and plasma lipids were assessed after 8, 12, 15, and 20 weeks. All 4 time points demonstrated exacerbated atherosclerotic lesion severity in ApoE−/−:TLR9−/− mice, with a corresponding increase in lipid deposition and accumulation of macrophages, dendritic cells, and CD4+ T cells. Although ApoE−/−:TLR9−/− mice exhibited an increase in plasma very low-density lipoprotein/low-density-lipoprotein cholesterol, the very low-density lipoprotein/low-density lipoprotein:high-density lipoprotein ratio was unaltered because of a parallel increase in plasma high-density lipoprotein cholesterol. As a potential mechanism accounting for plaque progression in ApoE−/−:TLR9−/− mice, CD4+ T-cell accumulation was further investigated and depletion of these cells in ApoE−/−:TLR9−/− mice significantly reduced lesion severity. As a final translational approach, administration of a TLR9 agonist (type B CpG oligodeoxynucleotide 1668) to ApoE−/− mice resulted in a reduction of lesion severity. Conclusions— Genetic deletion of the innate immune receptor TLR9 exacerbated atherosclerosis in ApoE−/− mice fed a high-fat diet. CD4+ T cells were identified as potential mediators of this effect. A type B CpG oligodeoxynucleotide TLR9 agonist reduced lesion severity, thus identifying a novel therapeutic approach in atherosclerosis.


PLOS ONE | 2011

GPVI and GPIbα Mediate Staphylococcal Superantigen-Like Protein 5 (SSL5) Induced Platelet Activation and Direct toward Glycans as Potential Inhibitors

Houyuan Hu; Paul C. J. Armstrong; Elie Khalil; Yung-Chih Chen; Andreas Straub; Min Li; Juliana Soosairajah; Christoph E. Hagemeyer; Nicole Bassler; Dexing Huang; Ingo Ahrens; Guy Y. Krippner; Elizabeth E. Gardiner; Karlheinz Peter

Background Staphylococcus aureus (S. aureus) is a common pathogen capable of causing life-threatening infections. Staphylococcal superantigen-like protein 5 (SSL5) has recently been shown to bind to platelet glycoproteins and induce platelet activation. This study investigates further the interaction between SSL5 and platelet glycoproteins. Moreover, using a glycan discovery approach, we aim to identify potential glycans to therapeutically target this interaction and prevent SSL5-induced effects. Methodology/Principal Findings In addition to platelet activation experiments, flow cytometry, immunoprecipitation, surface plasmon resonance and a glycan binding array, were used to identify specific SSL5 binding regions and mediators. We independently confirm SSL5 to interact with platelets via GPIbα and identify the sulphated-tyrosine residues as an important region for SSL5 binding. We also identify the novel direct interaction between SSL5 and the platelet collagen receptor GPVI. Together, these receptors offer one mechanistic explanation for the unique functional influences SSL5 exerts on platelets. A role for specific families of platelet glycans in mediating SSL5-platelet interactions was also discovered and used to identify and demonstrate effectiveness of potential glycan based inhibitors in vitro. Conclusions/Significance These findings further elucidate the functional interactions between SSL5 and platelets, including the novel finding of a role for the GPVI receptor. We demonstrate efficacy of possible glycan-based approaches to inhibit the SSL5-induced platelet activation. Our data warrant further work to prove SSL5-platelet effects in vivo.


Brain Pathology | 2012

Amyloid Plaques Dissociate Pentameric to Monomeric C-Reactive Protein: A Novel Pathomechanism Driving Cortical Inflammation in Alzheimer's Disease?

Frederik Strang; Amelie Scheichl; Yung-Chih Chen; Xiaowei Wang; Nay-Min Htun; Nicole Bassler; Steffen U. Eisenhardt; Jonathon Habersberger; Karlheinz Peter

Beta‐amyloid (Aβ) plaques and local inflammation are central to the pathogenesis of Alzheimers disease. Although an association between circulating pentameric C‐reactive protein (pCRP) and Alzheimers disease has been reported no pathomechanistic link has been established. We hypothesized that Aβ plaques induce the dissociation of pCRP to individual monomers (mCRP), which possess strong pro‐inflammatory properties not shared with pCRP and localizing inflammation to Alzheimers plaques. pCRP was incubated with Aβ plaques generated in vitro and with non‐aggregated Aβ42 peptide. pCRP dissociation to mCRP was found only when co‐incubated with Aβ plaques. Furthermore, sections of frontal cortex from brains of patients with and without Alzheimers disease were stained with antibodies specific for mCRP and pCRP. There was significantly more mCRP in the cortex of Alzheimers disease patients (Pu2003≤u20030.01). In contrast, there was no significant difference in pCRP staining. These findings establish that Aβ plaques possess a previously unrecognized potential to dissociate pentameric CRP to monomeric CRP. The existence of mCRP but not pCRP in the brains of Alzheimers disease patients strongly indicates that this newly described biological effect of Aβ plaques is relevant in Alzheimer pathobiology; potentially localizing and amplifying inflammation via the strong pro‐inflammatory effects of locally generated mCRP.


Thrombosis and Haemostasis | 2015

HMGB1 binds to activated platelets via the receptor for advanced glycation end products and is present in platelet rich human coronary artery thrombi

Ingo Ahrens; Yung-Chih Chen; Danijal Topcic; Michael Bode; David Haenel; Christoph E. Hagemeyer; Hannah Seeba; Daniel Duerschmied; Nicole Bassler; Karin Jandeleit-Dahm; Matthew J. Sweet; Alexander Agrotis; Alex Bobik; Karlheinz Peter

High mobility group box 1 (HMGB1) acts as both a nuclear protein that regulates gene expression, as well as a pro-inflammatory alarmin that is released from necrotic or activated cells. Recently, HMGB1-expression in human atherosclerotic plaques was identified. Therapeutic blockade of HMGB1 reduced the development of diet-induced atherosclerosis in ApoE knockout mice. Thus, we hypothesised an interaction between HMGB1 and activated platelets. Binding of recombinant HMGB1 to platelets was assessed by flow cytometry. HMGB1 bound to thrombin-activated human platelets (MFI 2.49 vs 25.01, p=0.0079). Blood from wild-type, TLR4 and RAGE knockout mice was used to determine potential HMGB1 receptors on platelets. HMGB1 bound to platelets from wild type C57Bl6 (MFI 2.64 vs 20.3, p<u20090.05), and TLR4-/- mice (MFI 2.11 vs 25.65, p<u20090.05) but failed to show binding to platelets from RAGE-/- mice (p >u20090.05). RAGE expression on human platelets was detected by RT-PCR with mRNA extracted from highly purified platelets and confirmed by Western blot and immunofluorescence microscopy. Platelet activation increased RAGE surface expression (MFI 4.85 vs 6.74, p<u20090.05). Expression of HMGB1 in human coronary artery thrombi was demonstrated by immunohistochemistry and revealed high expression levels. Platelets bind HMGB1 upon thrombin-induced activation. Platelet specific expression of RAGE could be detected at the mRNA and protein level and is involved in the binding of HMGB1. Furthermore, platelet activation up-regulates platelet surface expression of RAGE. HMGB1 is highly expressed in platelet-rich human coronary artery thrombi pointing towards a central role for HMGB1 in atherothrombosis, thereby suggesting the possibility of platelet targeted anti-inflammatory therapies for atherothrombosis.


Molecular Therapy | 2018

Dual-Targeted Theranostic Delivery of miRs Arrests Abdominal Aortic Aneurysm Development

Xiaowei Wang; Amy Kate Searle; Jan David Hohmann; Ao Leo Liu; Meike-Kristin Abraham; Jathushan Palasubramaniam; Bock Lim; Yu Yao; Maria Wallert; Eefang Yu; Yung-Chih Chen; Karlheinz Peter

Abdominal aortic aneurysm (AAA) is an often deadly disease without medical, non-invasive treatment options. The upregulation of vascular cell adhesion molecule-1 (VCAM-1) on aortic endothelium provides an early target epitope for a novel biotechnological theranostic approach. MicroRNA-126 was used as a therapeutic agent, based on its capability to downregulate VCAM-1 expression in endothelial cells and thereby reduces leukocyte adhesion and exerts anti-inflammatory effects. Ultrasound microbubbles were chosen as carriers, allowing both molecular imaging as well as targeted therapy of AAA. Microbubbles were coupled with a VCAM-1-targeted single-chain antibody (scFvmVCAM-1) and a microRNA-126 mimic (M126) constituting theranostic microbubbles (TargMB-M126). TargMB-M126 downregulates VCAM-1 expression in vitro and in an in vivo acute inflammatory murine model. Most importantly, using TargMB-M126 and ultrasound-guided burst delivery of M126, the development of AAA in an angiotensin-II-induced mouse model can be prevented. Overall, we describe a unique biotechnological theranostic approach with the potential for early diagnosis and long-sought-after medical therapy of AAA.


European Heart Journal | 2018

Myeloperoxidase is a potential molecular imaging and therapeutic target for the identification and stabilization of high-risk atherosclerotic plaque

Imran Rashid; Ghassan J. Maghzal; Yung-Chih Chen; David Cheng; Jihan Talib; Darren Newington; Minqin Ren; Saumitra Vajandar; Amy Kate Searle; Ana Maluenda; Eva-Lotte Lindstedt; Andrew Jabbour; Antony J Kettle; Andre Bongers; Carl A. Power; Erik Michaëlsson; Karlheinz Peter; Roland Stocker

AimsnAs the inflammatory enzyme myeloperoxidase (MPO) is abundant in ruptured human atherosclerotic plaques, we aimed to investigate the role of MPO as a potential diagnostic and therapeutic target for high-risk plaque.nnnMethods and resultsnWe employed the tandem stenosis model of atherosclerotic plaque instability in apolipoprotein E gene knockout (Apoe-/-) mice. To test the role of MPO, we used Mpo-/-Apoe-/- mice and the 2-thioxanthine MPO inhibitor AZM198. In vivo MPO activity was assessed by liquid chromatography-tandem mass spectrometry detection of 2-chloroethidium generation from hydroethidine and by bis-5HT-DTPA-Gd (MPO-Gd) molecular magnetic resonance imaging (MRI), while plaque phenotype was verified histologically. Myeloperoxidase activity was two-fold greater in plaque with unstable compared with stable phenotype. Genetic deletion of MPO significantly increased fibrous cap thickness, and decreased plaque fibrin and haemosiderin content in plaque with unstable phenotype. AZM198 inhibited MPO activity and it also increased fibrous cap thickness and decreased fibrin and haemosiderin in plaque with unstable phenotype, without affecting lesion monocytes and red blood cell markers or circulating leukocytes and lipids. MPO-Gd MRI demonstrated sustained enhancement of plaque with unstable phenotype on T1-weighted imaging that was two-fold greater than stable plaque and was significantly attenuated by both AZM198 treatment and deletion of the Mpo gene.nnnConclusionnOur data implicate MPO in atherosclerotic plaque instability and suggest that non-invasive imaging and pharmacological inhibition of plaque MPO activity hold promise for clinical translation in the management of high-risk coronary artery disease.


Atherosclerosis Supplements | 2018

Targeted Microbubbles Loaded with microRNA for a Theranostic Approach of Abdominal Aortic Aneurysms

Xiaowei Wang; Amy Kate Searle; Yung-Chih Chen; Karlheinz Peter


Atherosclerosis Supplements | 2018

Shining a Light on Plaque Instability: Near Infrared Autofluorescence as Characteristic of Unstable, High Risk Atherosclerotic Plaques

Yung-Chih Chen; Nay Min Htun; Ghassan J. Maghzal; Hans G. Schneider; Roland Stocker; Karlheinz Peter; Xiaowei Wang

Collaboration


Dive into the Yung-Chih Chen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ingo Ahrens

University of Freiburg

View shared research outputs
Top Co-Authors

Avatar

Alex Bobik

Baker IDI Heart and Diabetes Institute

View shared research outputs
Top Co-Authors

Avatar

Nicole Bassler

Baker IDI Heart and Diabetes Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Christian Hausding

Baker IDI Heart and Diabetes Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge