Network


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

Hotspot


Dive into the research topics where Kyoung-Jin Chung is active.

Publication


Featured researches published by Kyoung-Jin Chung.


Nature Immunology | 2012

The leukocyte integrin antagonist Del-1 inhibits IL-17-mediated inflammatory bone loss

Mehmet A. Eskan; Ravi Jotwani; Toshiharu Abe; Jindrich Chmelar; Jong-Hyung Lim; Shuang Liang; Paul A Ciero; Jennifer L. Krauss; Fenge Li; Martina Rauner; Lorenz C. Hofbauer; Eun-Young Choi; Kyoung-Jin Chung; Ahmed Hashim; Michael A. Curtis; Triantafyllos Chavakis; George Hajishengallis

Aging is linked to greater susceptibility to chronic inflammatory diseases, several of which, including periodontitis, involve neutrophil-mediated tissue injury. Here we found that aging-associated periodontitis was accompanied by lower expression of Del-1, an endogenous inhibitor of neutrophil adhesion dependent on the integrin LFA-1, and by reciprocal higher expression of interleukin 17 (IL-17). Consistent with that, IL-17 inhibited gingival endothelial cell expression of Del-1, thereby promoting LFA-1-dependent recruitment of neutrophils. Young Del-1-deficient mice developed spontaneous periodontitis that featured excessive neutrophil infiltration and IL-17 expression; disease was prevented in mice doubly deficient in Del-1 and LFA-1 or in Del-1 and the IL-17 receptor. Locally administered Del-1 inhibited IL-17 production, neutrophil accumulation and bone loss. Therefore, Del-1 suppressed LFA-1-dependent recruitment of neutrophils and IL-17-triggered inflammatory pathology and may thus be a promising therapeutic agent for inflammatory diseases.


Blood | 2010

Complement-mediated inhibition of neovascularization reveals a point of convergence between innate immunity and angiogenesis

Harald Langer; Kyoung-Jin Chung; Valeria V. Orlova; Eun Young Choi; Sunil Kaul; Michael J. Kruhlak; Markella Alatsatianos; Robert A. DeAngelis; Paul A. Roche; Paola Magotti; Xuri Li; Matina Economopoulou; Stavros Rafail; John D. Lambris; Triantafyllos Chavakis

Beyond its role in immunity, complement mediates a wide range of functions in the context of morphogenetic or tissue remodeling processes. Angiogenesis is crucial during tissue remodeling in multiple pathologies; however, the knowledge about the regulation of neovascularization by the complement components is scarce. Here we studied the involvement of complement in pathological angiogenesis. Strikingly, we found that mice deficient in the central complement component C3 displayed increased neovascularization in the model of retinopathy of prematurity (ROP) and in the in vivo Matrigel plug assay. In addition, antibody-mediated blockade of C5, treatment with C5aR antagonist, or C5aR deficiency in mice resulted in enhanced pathological retina angiogenesis. While complement did not directly affect angiogenesis-related endothelial cell functions, we found that macrophages mediated the antiangiogenic activity of complement. In particular, C5a-stimulated macrophages were polarized toward an angiogenesis-inhibitory phenotype, including the up-regulated secretion of the antiangiogenic soluble vascular endothelial growth factor receptor-1. Consistently, macrophage depletion in vivo reversed the increased neovascularization associated with C3- or C5aR deficiency. Taken together, complement and in particular the C5a-C5aR axes are potent inhibitors of angiogenesis.


Circulation Research | 2012

Platelets Contribute to the Pathogenesis of Experimental Autoimmune Encephalomyelitis

Harald Langer; Eun Young Choi; Hong Zhou; Rebecca Schleicher; Kyoung-Jin Chung; Zhongshu Tang; Kerstin Göbel; Khalil Bdeir; Antonios Chatzigeorgiou; Connie Hoi Yee Wong; Sumeena Bhatia; Michael J. Kruhlak; John Rose; James Burns; Kenneth E. Hill; Yongqing Zhang; Elin Lehrmann; Kevin G. Becker; Yunmei Wang; Daniel I. Simon; Bernhard Nieswandt; John D. Lambris; Xuri Li; Sven G. Meuth; Paul Kubes; Triantafyllos Chavakis

Rationale: Multiple sclerosis (MS) and its mouse model, experimental autoimmune encephalomyelitis (EAE), are inflammatory disorders of the central nervous system (CNS). The function of platelets in inflammatory and autoimmune pathologies is thus far poorly defined. Objective: We addressed the role of platelets in mediating CNS inflammation in EAE. Methods and Results: We found that platelets were present in human MS lesions as well as in the CNS of mice subjected to EAE but not in the CNS from control nondiseased mice. Platelet depletion at the effector-inflammatory phase of EAE in mice resulted in significantly ameliorated disease development and progression. EAE suppression on platelet depletion was associated with reduced recruitment of leukocytes to the inflamed CNS, as assessed by intravital microscopy, and with a blunted inflammatory response. The platelet-specific receptor glycoprotein Ib&agr; (GPIb&agr;) promotes both platelet adhesion and inflammatory actions of platelets and targeting of GPIb&agr; attenuated EAE in mice. Moreover, targeting another platelet adhesion receptor, glycoprotein IIb/IIIa (GPIIb/IIIa), also reduced EAE severity in mice. Conclusions: Platelets contribute to the pathogenesis of EAE by promoting CNS inflammation. Targeting platelets may therefore represent an important new therapeutic approach for MS treatment.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Blocking CD40-TRAF6 signaling is a therapeutic target in obesity-associated insulin resistance.

Antonios Chatzigeorgiou; Tom Seijkens; Barbara Zarzycka; David Engel; Marjorie Poggi; Susan M. van den Berg; Sjoerd A. A. van den Berg; Oliver Soehnlein; Holger Winkels; Linda Beckers; Dirk Lievens; A. Driessen; Pascal Kusters; Erik A.L. Biessen; Ruben Garcia-Martin; Anne Klotzsche-von Ameln; Marion J. J. Gijbels; Randolph J. Noelle; Louis Boon; Tilman M. Hackeng; Klaus-Martin Schulte; Aimin Xu; Gert Vriend; Sander B. Nabuurs; Kyoung-Jin Chung; Ko Willems van Dijk; Patrick C. N. Rensen; Menno P.J. de Winther; Norman L. Block; Andrew V. Schally

Significance Inflammation is a critical contributor to the pathogenesis of metabolic disorders associated with obesity. A group of molecules crucial in regulating the immune system are costimulatory molecules, including CD40. Our current study shows that CD40 acts as a double-edged sword in the metabolic syndrome through the initiation of differential signaling cascades. The CD40-TNF receptor-associated factor (TRAF) 2/3/5 signaling pathway protects against metabolic dysfunction and inflammation associated with obesity; conversely, the CD40-TRAF6 pathway contributes to the detrimental consequences of obesity. In the present study, we therefore designed, validated, and used a small-molecule inhibitor that blocks CD40-TRAF6 interactions. The improvement of insulin resistance by this specific CD40-TRAF6 inhibitor could represent a therapeutic breakthrough in the field of immunometabolism. The immune system plays an instrumental role in obesity and insulin resistance. Here, we unravel the role of the costimulatory molecule CD40 and its signaling intermediates, TNF receptor-associated factors (TRAFs), in diet-induced obesity (DIO). Although not exhibiting increased weight gain, male CD40−/− mice in DIO displayed worsened insulin resistance, compared with wild-type mice. This worsening was associated with excessive inflammation of adipose tissue (AT), characterized by increased accumulation of CD8+ T cells and M1 macrophages, and enhanced hepatosteatosis. Mice with deficient CD40-TRAF2/3/5 signaling in MHCII+ cells exhibited a similar phenotype in DIO as CD40−/− mice. In contrast, mice with deficient CD40-TRAF6 signaling in MHCII+ cells displayed no insulin resistance and showed a reduction in both AT inflammation and hepatosteatosis in DIO. To prove the therapeutic potential of inhibition of CD40-TRAF6 in obesity, DIO mice were treated with a small-molecule inhibitor that we designed to specifically block CD40-TRAF6 interactions; this compound improved insulin sensitivity, reduced AT inflammation, and decreased hepatosteatosis. Our study reveals that the CD40-TRAF2/3/5 signaling pathway in MHCII+ cells protects against AT inflammation and metabolic complications associated with obesity whereas CD40-TRAF6 interactions in MHCII+ cells aggravate these complications. Inhibition of CD40-TRAF6 signaling by our compound may provide a therapeutic option in obesity-associated insulin resistance.


Thrombosis and Haemostasis | 2013

The role of innate immune cells in obese adipose tissue inflammation and development of insulin resistance

Jindrich Chmelar; Kyoung-Jin Chung; Triantafyllos Chavakis

Obesity is characterised by a chronic state of low-grade inflammation in different tissues including the vasculature. There is a causal link between adipose tissue (AT) inflammation and obesity-related metabolic complications, such as the development of insulin resistance and subsequently of type 2 diabetes. Intense efforts in the recent years have aimed at dissecting the pathophysiology of AT inflammation. The role of both innate and adaptive immune cells, such as macrophages or cytotoxic T cells in AT inflammation has been demonstrated. Besides these cells, more leukocyte subpopulations have been recently implicated in obesity, including neutrophils and eosinophils, mast cells, natural killer cells or dendritic cells. The involvement of multiple leukocyte subpopulations underlines the complexity of obesity-associated AT inflammation. In this review, we discuss the role of innate immune cells in AT inflammation, obesity and related metabolic disorders.


Nature Communications | 2015

Extracellular MRP8/14 is a regulator of β2 integrin-dependent neutrophil slow rolling and adhesion.

Monika Pruenster; Angela R.M. Kurz; Kyoung-Jin Chung; Xiao Cao-Ehlker; Stephanie Bieber; Claudia Nussbaum; Susanne Bierschenk; Tanja K Eggersmann; Ina Rohwedder; Kristina Heinig; Roland Immler; Markus Moser; Uwe Koedel; Sandra Gran; Rodger P. McEver; Dietmar Vestweber; Admar Verschoor; Tomas Leanderson; Triantafyllos Chavakis; J. Roth; Thomas Vogl; Markus Sperandio

Myeloid-related proteins (MRPs) 8 and 14 are cytosolic proteins secreted from myeloid cells as proinflammatory mediators. Currently, the functional role of circulating extracellular MRP8/14 is unclear. Our present study identifies extracellular MRP8/14 as an autocrine player in the leukocyte adhesion cascade. We show that E-selectin–PSGL-1 interaction during neutrophil rolling triggers Mrp8/14 secretion. Released MRP8/14 in turn activates a TLR4-mediated, Rap1-GTPase-dependent pathway of rapid β2 integrin activation in neutrophils. This extracellular activation loop reduces leukocyte rolling velocity and stimulates adhesion. Thus, we identify Mrp8/14 and TLR4 as important modulators of the leukocyte recruitment cascade during inflammation in vivo.


Journal of Immunology | 2013

The Complement Anaphylatoxin C5a Receptor Contributes to Obese Adipose Tissue Inflammation and Insulin Resistance

Julia Phieler; Kyoung-Jin Chung; Antonios Chatzigeorgiou; Anne Klotzsche-von Ameln; Ruben Garcia-Martin; David Sprott; Maria Moisidou; Theodora Tzanavari; Barbara Ludwig; Elena Baraban; Monika Ehrhart-Bornstein; Stefan R. Bornstein; Hassan Mziaut; Michele Solimena; Katia P. Karalis; Matina Economopoulou; John D. Lambris; Triantafyllos Chavakis

Obese adipose tissue (AT) inflammation contributes critically to development of insulin resistance. The complement anaphylatoxin C5a receptor (C5aR) has been implicated in inflammatory processes and as regulator of macrophage activation and polarization. However, the role of C5aR in obesity and AT inflammation has not been addressed. We engaged the model of diet-induced obesity and found that expression of C5aR was significantly upregulated in the obese AT, compared with lean AT. In addition, C5a was present in obese AT in the proximity of macrophage-rich crownlike structures. C5aR-sufficient and -deficient mice were fed a high-fat diet (HFD) or a normal diet (ND). C5aR deficiency was associated with increased AT weight upon ND feeding in males, but not in females, and with increased adipocyte size upon ND and HFD conditions in males. However, obese C5aR−/− mice displayed improved systemic and AT insulin sensitivity. Improved AT insulin sensitivity in C5aR−/− mice was associated with reduced accumulation of total and proinflammatory M1 macrophages in the obese AT, increased expression of IL-10, and decreased AT fibrosis. In contrast, no difference in β cell mass was observed owing to C5aR deficiency under an HFD. These results suggest that C5aR contributes to macrophage accumulation and M1 polarization in the obese AT and thereby to AT dysfunction and development of AT insulin resistance.


Hepatology | 2014

Dual role of B7 costimulation in obesity-related nonalcoholic steatohepatitis and metabolic dysregulation.

Antonios Chatzigeorgiou; Kyoung-Jin Chung; Ruben Garcia-Martin; Vasileia‐Ismini Alexaki; Anne Klotzsche-von Ameln; Julia Phieler; David Sprott; Waldemar Kanczkowski; Theodora Tzanavari; Mohktar Bdeir; Sibylle Bergmann; Marc Cartellieri; Michael H. Bachmann; Polyxeni Nikolakopoulou; Andreas Androutsellis-Theotokis; Gabriele Siegert; Stefan R. Bornstein; Michael H. Muders; Louis Boon; Katia Karalis; Esther Lutgens; Triantafyllos Chavakis

The low‐grade inflammatory state present in obesity contributes to obesity‐related metabolic dysregulation, including nonalcoholic steatohepatitis (NASH) and insulin resistance. Intercellular interactions between immune cells or between immune cells and hepatic parenchymal cells contribute to the exacerbation of liver inflammation and steatosis in obesity. The costimulatory molecules, B7.1 and B7.2, are important regulators of cell‐cell interactions in several immune processes; however, the role of B7 costimulation in obesity‐related liver inflammation is unknown. Here, diet‐induced obesity (DIO) studies in mice with genetic inactivation of both B7.1 and B7.2 (double knockout; DKO) revealed aggravated obesity‐related metabolic dysregulation, reduced insulin signalling in the liver and adipose tissue (AT), glucose intolerance, and enhanced progression to steatohepatitis resulting from B7.1/B7.2 double deficiency. The metabolic phenotype of B7.1/B7.2 double deficiency upon DIO was accompanied by increased hepatic and AT inflammation, associated with largely reduced numbers of regulatory T cells (Tregs) in these organs. In order to assess the role of B7 costimulation in DIO in a non‐Treg‐lacking environment, we performed antibody (Ab)‐mediated inhibition of B7 molecules in wild‐type mice in DIO. Antibody‐blockade of both B7.1 and B7.2 improved the metabolic phenotype of DIO mice, which was linked to amelioration of hepatic steatosis and reduced inflammation in liver and AT. Conclusion: Our study demonstrates a dual role of B7 costimulation in the course of obesity‐related sequelae, particularly NASH. The genetic inactivation of B7.1/B7.2 deteriorates obesity‐related liver steatosis and metabolic dysregulation, likely a result of the intrinsic absence of Tregs in these mice, rendering DKO mice a novel murine model of NASH. In contrast, inhibition of B7 costimulation under conditions where Tregs are present may provide a novel therapeutic approach for obesity‐related metabolic dysregulation and, especially, NASH. (Hepatology 2014;60:1196–1210)


Molecular Psychiatry | 2015

Developmental endothelial locus-1 is a homeostatic factor in the central nervous system limiting neuroinflammation and demyelination.

Eun-Young Choi; Jong-Hyung Lim; Ales Neuwirth; Matina Economopoulou; Antonios Chatzigeorgiou; Kyoung-Jin Chung; Stefan Bittner; Seung Hwan Lee; Harald Langer; Maryna Samus; Hyesoon Kim; Geum-Sil Cho; Tjalf Ziemssen; Khalil Bdeir; Emmanouil Chavakis; Jae-Young Koh; Louis Boon; Kavita B. Hosur; Stefan R. Bornstein; Sven G. Meuth; George Hajishengallis; Triantafyllos Chavakis

Inflammation in the central nervous system (CNS) and disruption of its immune privilege are major contributors to the pathogenesis of multiple sclerosis (MS) and of its rodent counterpart, experimental autoimmune encephalomyelitis (EAE). We have previously identified developmental endothelial locus-1 (Del-1) as an endogenous anti-inflammatory factor, which inhibits integrin-dependent leukocyte adhesion. Here we show that Del-1 contributes to the immune privilege status of the CNS. Intriguingly, Del-1 expression decreased in chronic-active MS lesions and in the inflamed CNS in the course of EAE. Del-1-deficiency was associated with increased EAE severity, accompanied by increased demyelination and axonal loss. As compared with control mice, Del-1−/− mice displayed enhanced disruption of the blood–brain barrier and increased infiltration of neutrophil granulocytes in the spinal cord in the course of EAE, accompanied by elevated levels of inflammatory cytokines, including interleukin-17 (IL-17). The augmented levels of IL-17 in Del-1-deficiency derived predominantly from infiltrated CD8+ T cells. Increased EAE severity and neutrophil infiltration because of Del-1-deficiency was reversed in mice lacking both Del-1 and IL-17 receptor, indicating a crucial role for the IL-17/neutrophil inflammatory axis in EAE pathogenesis in Del-1−/− mice. Strikingly, systemic administration of Del-1-Fc ameliorated clinical relapse in relapsing–remitting EAE. Therefore, Del-1 is an endogenous homeostatic factor in the CNS protecting from neuroinflammation and demyelination. Our findings provide mechanistic underpinnings for the previous implication of Del-1 as a candidate MS susceptibility gene and suggest that Del-1-centered therapeutic approaches may be beneficial in neuroinflammatory and demyelinating disorders.


Molecular and Cellular Biology | 2016

Adipocyte-Specific Hypoxia-Inducible Factor 2α Deficiency Exacerbates Obesity-Induced Brown Adipose Tissue Dysfunction and Metabolic Dysregulation

Ruben Garcia-Martin; Vasileia Ismini Alexaki; Nan Qin; Maria F. Rubin de Celis; Matina Economopoulou; Athanasios Ziogas; Bettina Gercken; Klara Kotlabova; Julia Phieler; Monika Ehrhart-Bornstein; Stefan R. Bornstein; Graeme Eisenhofer; Georg Breier; Matthias Blüher; Jochen Hampe; Ali El-Armouche; Antonios Chatzigeorgiou; Kyoung-Jin Chung; Triantafyllos Chavakis

ABSTRACT Angiogenesis is a central regulator for white (WAT) and brown (BAT) adipose tissue adaptation in the course of obesity. Here we show that deletion of hypoxia-inducible factor 2α (HIF2α) in adipocytes (by using Fabp4-Cre transgenic mice) but not in myeloid or endothelial cells negatively impacted WAT angiogenesis and promoted WAT inflammation, WAT dysfunction, hepatosteatosis, and systemic insulin resistance in obesity. Importantly, adipocyte HIF2α regulated vascular endothelial growth factor (VEGF) expression and angiogenesis of obese BAT as well as its thermogenic function. Consistently, obese adipocyte-specific HIF2α-deficient mice displayed BAT dysregulation, associated with reduced levels of uncoupling protein 1 (UCP1) and a dysfunctional thermogenic response to cold exposure. VEGF administration reversed WAT and BAT inflammation and BAT dysfunction in adipocyte HIF2α-deficient mice. Together, our findings show that adipocyte HIF2α is protective against maladaptation to obesity and metabolic dysregulation by promoting angiogenesis in both WAT and BAT and by counteracting obesity-mediated BAT dysfunction.

Collaboration


Dive into the Kyoung-Jin Chung's collaboration.

Top Co-Authors

Avatar

Triantafyllos Chavakis

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar

Antonios Chatzigeorgiou

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar

Stefan R. Bornstein

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar

Ruben Garcia-Martin

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar

Matina Economopoulou

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar

John D. Lambris

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Anne Klotzsche-von Ameln

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar

Louis Boon

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge