Network


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

Hotspot


Dive into the research topics where Alexander Niessner is active.

Publication


Featured researches published by Alexander Niessner.


European Heart Journal | 2018

2017 ESC Guidelines for the management of acute myocardial infarction in patients presenting with ST-segment elevation: The Task Force for the management of acute myocardial infarction in patients presenting with ST-segment elevation of the European Society of Cardiology (ESC).

Borja Ibanez; Stefan James; Stefan Agewall; Manuel J. Antunes; Chiara Bucciarelli-Ducci; Héctor Bueno; Alida L.P. Caforio; Filippo Crea; John A. Goudevenos; Sigrun Halvorsen; Gerhard Hindricks; Adnan Kastrati; Mattie J. Lenzen; Eva Prescott; Marco Roffi; Marco Valgimigli; Christoph Varenhorst; Pascal Vranckx; Petr Widimsky; Jean-Philippe Collet; Steen Dalby Kristensen; Victor Aboyans; Andreas Baumbach; Raffaele Bugiardini; Ioan Mircea Coman; Victoria Delgado; Donna Fitzsimons; Oliver Gaemperli; Anthony H. Gershlick; Stephan Gielen

2017 ESC Guidelines for the management of acute myocardial infarction in patients presenting with ST-segment elevation The Task Force for the management of acute myocardial infarction in patients presenting with ST-segment elevation of the European Society of Cardiology (ESC)


Circulation | 2006

Pathogen-Sensing Plasmacytoid Dendritic Cells Stimulate Cytotoxic T-Cell Function in the Atherosclerotic Plaque Through Interferon-α

Alexander Niessner; Kayoko Sato; Elliot L. Chaikof; Inés Colmegna; Jörg J. Goronzy; Cornelia M. Weyand

Background— Unstable atherosclerotic plaque is characterized by an infiltrate of inflammatory cells. Both macrophages and T cells have been implicated in mediating the tissue injury leading to plaque rupture; however, signals regulating their activation remain unidentified. Infectious episodes have been suspected to render plaques vulnerable to rupture. We therefore explored whether plasmacytoid dendritic cells (pDCs) that specialize in sensing bacterial and viral products can regulate effector functions of plaque-residing T cells and thus connect host infection and plaque instability. Methods and Results— pDCs were identified in 53% of carotid atheromas (n=30) in which they localized to the shoulder region and produced the potent immunoregulatory cytokine interferon (INF)-&agr;. IFN-&agr; transcript concentrations in atheroma tissues correlated strongly with plaque instability (P<0.0001). Plaque-residing pDCs responded to pathogen-derived motifs, CpG-containing oligodeoxynucleotides binding to toll-like receptor 9, with enhanced IFN-&agr; transcription (P=0.03) and secretion (P=0.007). IFN-&agr; emerged as a potent regulator of T-cell function, even in the absence of antigen recognition. Specifically, IFN-&agr; induced a 10-fold increase of tumor necrosis factor–related apoptosis-inducing ligand (TRAIL) on the surface of CD4 T cells (P<0.0001) and enabled them to effectively kill vascular smooth muscle cells (P=0.0003). Conclusions— pDCs in atherosclerotic plaque sense microbial motifs and amplify cytolytic T-cell functions, thus providing a link between host-infectious episodes and acute immune-mediated complications of atherosclerosis.


Journal of Experimental Medicine | 2006

TRAIL-expressing T cells induce apoptosis of vascular smooth muscle cells in the atherosclerotic plaque

Kayoko Sato; Alexander Niessner; Stephen L. Kopecky; Robert L. Frye; Jörg J. Goronzy; Cornelia M. Weyand

Acute coronary syndromes (ACS) are precipitated by a rupture of the atherosclerotic plaque, often at the site of T cell and macrophage infiltration. Here, we show that plaque-infiltrating CD4 T cells effectively kill vascular smooth muscle cells (VSMC). VSMCs sensitive to T cell–mediated killing express the death receptor DR5 (TNF-related apoptosis-inducing ligand [TRAIL] receptor 2), and anti-TRAIL and anti-DR5 antibodies block T cell–mediated apoptosis. CD4 T cells that express TRAIL upon stimulation are expanded in patients with ACS and more effectively induce VSMC apoptosis. Adoptive transfer of plaque-derived CD4 T cells into immunodeficient mice that are engrafted with human atherosclerotic plaque results in apoptosis of VSMCs, which was prevented by coadministration of anti-TRAIL antibody. These data identify that the death pathway is triggered by TRAIL-producing CD4 T cells as a direct mechanism of VSMC apoptosis, a process which may lead to plaque destabilization.


Clinical Immunology | 2010

Dendritic cells in atherosclerotic disease

Alexander Niessner; Cornelia M. Weyand

Atherosclerosis has been considered a syndrome of dysregulated lipid storage until recent evidence has emphasized the critical contribution of the immune system. Dendritic cells (DC) are positioned at the interface of the innate and adaptive immune system. Recognition of danger signals in atheromas leads to DC activation. Activated DC regulate effector T cells which can kill plaque-resident cells and damage the plaque structure. Two types of DC have been identified in atherosclerotic lesions; classical myeloid DC (mDC) which mainly recognize bacterial signatures and plasmacytoid DC (pDC) which specialize in sensing viral fragments and have the unique potential of producing large amounts of type I interferon (IFN). In human atheromas, type I IFN upregulates expression of the cytotoxic molecule TRAIL which leads to apoptosis of plaque-resident cells. This review will elucidate the role of DC in atherogenesis and particularly in plaque rupture, the underlying pathophysiologic cause of myocardial infarction.


Circulation | 2007

Synergistic Proinflammatory Effects of the Antiviral Cytokine Interferon-α and Toll-Like Receptor 4 Ligands in the Atherosclerotic Plaque

Alexander Niessner; Min Sun Shin; Olga Pryshchep; Jörg J. Goronzy; Elliot L. Chaikof; Cornelia M. Weyand

Background— Interferon (IFN)-&agr; is a pluripotent inflammatory cytokine typically induced by viral infections. In rupture-prone atherosclerotic plaques, plasmacytoid dendritic cells produce IFN-&agr;. In the present study we explored the contribution of IFN-&agr; to inflammation and tissue injury in the plaque microenvironment. Methods and Results— In 53% of carotid plaques (n=30), CD123+ plasmacytoid dendritic cells clustered together with CD11c+ myeloid dendritic cells, a distinct dendritic cell subset specialized in sensing danger signals from bacteria and tissue breakdown. Tissue concentrations of IFN-&agr; and tumor necrosis factor (TNF)-&agr; transcripts were tightly correlated (r=0.76, P<0.001), suggesting a regulatory role of IFN-&agr; in TNF-&agr; production. Plaque tissue stimulation with CpG ODN, a Toll-like receptor (TLR) 9 ligand, increased IFN-&agr; production (57.8±23.7 versus 25.9±8.6 pg/mL; P<0.001), whereas the TLR4 ligand lipopolysaccharide induced TNF-&agr; secretion (225.1±3.0 versus 0.7±0.2 pg/mL; P<0.001). Treating plaque tissue with IFN-&agr; markedly enhanced lipopolysaccharide-triggered TNF-&agr; secretion (559.0±25.9 versus 225.1±3.0 pg/mL; P<0.001). IFN-&agr; pretreatment also amplified the effects of lipopolysaccharide on interleukin-12, interleukin-23, and matrix metalloproteinase-9, suggesting that the antiviral cytokine sensitized myeloid dendritic cells and macrophages toward TLR4 ligands. Mechanistic studies demonstrated that IFN-&agr; modulated the myeloid dendritic cell response pattern by upregulating TLR4 expression (P<0.001) involving both the STAT (signal transducer and activator of transcription) and the PI(3)K pathway. Conclusions— In the atherosclerotic plaque, IFN-&agr; functions as an inflammatory amplifier. It sensitizes antigen-presenting cells toward pathogen-derived TLR4 ligands by upregulating TLR4 expression and intensifies TNF-&agr;, interleukin-12, and matrix metalloproteinase-9 production, all implicated in plaque destabilization. Thus, IFN-&agr;–inducing pathogens, even when colonizing distant tissue sites, threaten the stability of inflamed atherosclerotic plaque.


European Heart Journal | 2008

Prognostic value of apoptosis markers in advanced heart failure patients

Alexander Niessner; Philipp J. Hohensinner; Kathrin Rychli; Stephanie Neuhold; Gerlinde Zorn; Bernhard Richter; Martin Hülsmann; Rudolf Berger; Deddo Mörtl; Kurt Huber; Johann Wojta; Richard Pacher

AIMS Apoptosis plays an important role in the progression of heart failure (HF). The purpose of this study was to assess whether the pro-apoptotic molecules apoptosis-stimulating fragment (FAS, CD95/APO-1) and tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) predict event-free survival of HF patients. METHODS AND RESULTS We assayed soluble (s)FAS and sTRAIL levels in 351 patients with advanced HF. During the median follow-up time of 16 months, 175 patients (50%) experienced the composite endpoints: rehospitalization and death. The hazard increased with sFAS concentrations, with a hazard ratio of 2.3 comparing fourth and first quartiles. This association remained significant after adjustment for B-type natriuretic peptide (BNP) and other risk factors in a Cox regression model (P = 0.014). Patients with high sFAS but low BNP had a comparable event-free survival rate with those with elevated BNP only (P = 0.78). Conversely, high sTRAIL concentrations were related to a better prognosis. Particularly, the risk of mortality dropped by 70% in the fourth quartile of sTRAIL (P = 0.001, multivariable Cox regression model). CONCLUSION sFAS is an independent risk predictor in advanced HF patients. It may be of particular value for the identification of high-risk patients in addition to BNP. Conversely, sTRAIL appears to be protective and could be an interesting therapeutic agent.


Circulation Research | 2006

Interleukin 12 Induces T-Cell Recruitment Into the Atherosclerotic Plaque

Xiaoyu Zhang; Alexander Niessner; Takako Nakajima; Wei Ma-Krupa; Stephen L. Kopecky; Robert L. Frye; Jörg J. Goronzy; Cornelia M. Weyand

CD4 T cells, through the release of cytokines as well as direct effector functions, have been implicated in promoting inflammation of the atherosclerotic plaque. Plaque-infiltrating CD4 T cells include a specialized subset of CD4+CD28− T cells that express a unique profile of regulatory receptors and are responsive to novel microenvironmental cues. Here we report that CD4+CD28− T cells, either isolated from the plaque tissue or from the blood of patients with acute coronary syndrome (ACS), spontaneously express interleukin (IL)-12 receptors, even in the absence of antigenic stimulation. CD4+CD28− IL-12R+ cells responded to IL-12 stimulation with the upregulation of the chemokine receptor CCR5 and the C-type lectin receptor CD161, both implicated in regulating tissue homing of effector T cells. IL-12 treatment of CD4+CD28− T cells enhanced their chemotaxis and transendothelial migration toward the chemokine CCL5. In vivo relevance for the role of IL-12 in regulating the recruitment of CD4+CD28− T cells into the atheroma was examined in human atheroma-SCID mouse chimeras. Exposure of nonstimulated CD4+CD28− T cells to IL-12 was sufficient to amplify T-cell accumulation within the inflamed plaque, and coadministration of anti-CCR5 antibodies blocked T-cell recruitment into the plaque. Thus, CD4+CD28− T cells functionally resemble NK cells, which have proinflammatory activity even in the unprimed state and respond to any IL-12–inducing host infection with a shift in tissue trafficking and accrual in inflammatory lesions.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2011

Interleukin-33 Induces Expression of Adhesion Molecules and Inflammatory Activation in Human Endothelial Cells and in Human Atherosclerotic Plaques

Svitlana Demyanets; Viktoria Konya; Stefan P. Kastl; Christoph Kaun; Sabine Rauscher; Alexander Niessner; Richard Pentz; Stefan Pfaffenberger; Kathrin Rychli; Christof E. Lemberger; Rainer de Martin; Akos Heinemann; Ihor Huk; Marion Gröger; Gerald Maurer; Kurt Huber; Johann Wojta

Objective— Interleukin (IL)-33 is the most recently described member of the IL-1 family of cytokines and it is a ligand of the ST2 receptor. While the effects of IL-33 on the immune system have been extensively studied, the properties of this cytokine in the cardiovascular system are much less investigated. Methods/Results— We show here that IL-33 promoted the adhesion of human leukocytes to monolayers of human endothelial cells and robustly increased vascular cell adhesion molecule-1, intercellular adhesion molecule-1, endothelial selectin, and monocyte chemoattractant protein-1 protein production and mRNA expression in human coronary artery and human umbilical vein endothelial cells in vitro as well as in human explanted atherosclerotic plaques ex vivo. ST2-fusion protein, but not IL-1 receptor antagonist, abolished these effects. IL-33 induced translocation of nuclear factor-&kgr;B p50 and p65 subunits to the nucleus in human coronary artery endothelial cells and human umbilical vein endothelial cells and overexpression of dominant negative form of I&kgr;B kinase 2 or I&kgr;B&agr; in human umbilical vein endothelial cells abolished IL-33-induced adhesion molecules and monocyte chemoattractant protein-1 mRNA expression. We detected IL-33 and ST2 on both protein and mRNA level in human carotid atherosclerotic plaques. Conclusion— We hypothesize that IL-33 may contribute to early events in endothelial activation characteristic for the development of atherosclerotic lesions in the vessel wall, by promoting adhesion molecules and proinflammatory cytokine expression in the endothelium.


European Heart Journal | 2016

ESC working group position paper on myocardial infarction with non-obstructive coronary arteries

Stefan Agewall; John F. Beltrame; Harmony R. Reynolds; Alexander Niessner; Giuseppe Rosano; Alida L.P. Caforio; Raffaele De Caterina; Marco Zimarino; Marco Roffi; Keld Kjeldsen; Dan Atar; Juan Carlos Kaski; Udo Sechtem; Per Tornvall

The management of acute myocardial infarction (AMI)1 has evolved over the past century and particularly in the past 50 years. Important milestones include the development of the electrocardiogram, coronary care units, coronary angiography, reperfusion therapies, and troponin assays. These innovations are the foundation of contemporary AMI management strategies that include a diagnosis centred on elevated troponin values associated with corroborative clinical evidence,1 early use of coronary angiography, and reperfusion therapies.2–4 Pivotal in the evolution of these contemporary strategies were the early AMI coronary angiography studies undertaken by DeWood et al. These pioneering studies demonstrated that, in patients presenting with ST elevation myocardial infarction (STEMI), almost 90% had an occluded coronary artery provided that angiography was undertaken within 4 h of chest pain onset.5 In contrast, in AMI patients who did not present with ST elevation (non-ST elevation myocardial infarction or NSTEMI), only 26% had an occluded coronary artery when angiography was performed within 24 h of symptom onset.6 In both of these landmark studies,5,6 >90% of the acute MI patients had angiographic evidence of obstructive coronary artery disease (CAD), underscoring the importance of the atherosclerotic process in the pathogenesis of AMI. Although DeWoods studies underscore the importance of obstructive CAD in AMI, it is fascinating that ∼10% had no significant CAD on coronary angiography. This is confirmed in several large AMI registries7–9 where 1–13% of AMIs occurred in the absence of obstructive CAD thereby eliciting an important set of questions—what is the mechanism of the myocardial damage in these patients? Do these patients differ from those with obstructive CAD? Should they be …


Stroke | 2006

Risk Score for Peri-Interventional Complications of Carotid Artery Stenting

Robert Hofmann; Alexander Niessner; Alexander Kypta; Clemens Steinwender; Jürgen Kammler; Klaus Kerschner; Michael Grund; Franz Leisch; Kurt Huber

Background and Purpose— Routinely available independent risk factors for the peri-interventional outcome of patients undergoing elective carotid artery stenting (CAS) are lacking. The rationale of the study was to create a risk score identifying high-risk patients. Methods— We prospectively enrolled 606 consecutive patients assigned to CAS at a secondary care hospital. Various biochemical, clinical, and lesion-related risk factors were prospectively defined. The primary end point reflecting periprocedural complications encompassed minor and major stroke, nonfatal myocardial infarction and all-cause mortality within 30 days. Results— Three percent of patients (n=18) experienced a nonfatal minor (n=13) or major (n=5) stroke. 1.3% of patients (n=8) died from fatal stroke (n=4) or other causes (n=4). No myocardial infarction was observed within 30 days after stenting. Multivariable analysis revealed diabetes mellitus with inadequate glycemic control (HbA1c >7%), age ≥80 years, ulceration of the carotid artery stenosis, and a contralateral stenosis ≥50% as independent risk factors. A risk score formed with these variables showed a superior predictive value (C-statistic=0.73) compared with single risk factors. The presence of 2 or more of these risk factors identified patients with a risk of 11% for a periprocedural complication compared with 2% in patients with a score of 0 or 1. Conclusions— In patients undergoing elective CAS, a risk score based on routinely accessible variables was able to identify patients at high-risk for atherothrombotic events and all-cause death within 30 days after the intervention.

Collaboration


Dive into the Alexander Niessner's collaboration.

Top Co-Authors

Avatar

Johann Wojta

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Gerald Maurer

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Georg Goliasch

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Kurt Huber

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Lorenz Koller

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Gerlinde Zorn

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Patrick Sulzgruber

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Bernhard Richter

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Martin Hülsmann

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge