Network


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

Hotspot


Dive into the research topics where Christopher J. Czura is active.

Publication


Featured researches published by Christopher J. Czura.


Nature | 2002

Nicotinic acetylcholine receptor α7 subunit is an essential regulator of inflammation

Hong Wang; Man Yu; Mahendar Ochani; Carol Ann Amella; Mahira Tanovic; Seenu Susarla; Jianhua Li; Haichao Wang; Huan Yang; Luis Ulloa; Yousef Al-Abed; Christopher J. Czura; Kevin J. Tracey

Excessive inflammation and tumour-necrosis factor (TNF) synthesis cause morbidity and mortality in diverse human diseases including endotoxaemia, sepsis, rheumatoid arthritis and inflammatory bowel disease. Highly conserved, endogenous mechanisms normally regulate the magnitude of innate immune responses and prevent excessive inflammation. The nervous system, through the vagus nerve, can inhibit significantly and rapidly the release of macrophage TNF, and attenuate systemic inflammatory responses. This physiological mechanism, termed the ‘cholinergic anti-inflammatory pathway’ has major implications in immunology and in therapeutics; however, the identity of the essential macrophage acetylcholine-mediated (cholinergic) receptor that responds to vagus nerve signals was previously unknown. Here we report that the nicotinic acetylcholine receptor α7 subunit is required for acetylcholine inhibition of macrophage TNF release. Electrical stimulation of the vagus nerve inhibits TNF synthesis in wild-type mice, but fails to inhibit TNF synthesis in α7-deficient mice. Thus, the nicotinic acetylcholine receptor α7 subunit is essential for inhibiting cytokine synthesis by the cholinergic anti-inflammatory pathway.


Proceedings of the National Academy of Sciences of the United States of America | 2004

Reversing established sepsis with antagonists of endogenous high-mobility group box 1

Huan Yang; Mahendar Ochani; Jianhua Li; Xiaoling Qiang; Mahira Tanovic; Helena Erlandsson Harris; Srinivas M. Susarla; Luis Ulloa; Hong Wang; Robert DiRaimo; Christopher J. Czura; Haichao Wang; Jesse Roth; H. Shaw Warren; Mitchell P. Fink; Matthew J. Fenton; Ulf Andersson; Kevin J. Tracey

Despite significant advances in intensive care therapy and antibiotics, severe sepsis accounts for 9% of all deaths in the United States annually. The pathological sequelae of sepsis are characterized by a systemic inflammatory response, but experimental therapeutics that target specific early inflammatory mediators [tumor necrosis factor (TNF) and IL-1β] have not proven efficacious in the clinic. We recently identified high mobility group box 1 (HMGB1) as a late mediator of endotoxin-induced lethality that exhibits significantly delayed kinetics relative to TNF and IL-1β. Here, we report that serum HMGB1 levels are increased significantly in a standardized model of murine sepsis, beginning 18 h after surgical induction of peritonitis. Specific inhibition of HMGB1 activity [with either anti-HMGB1 antibody (600 μg per mouse) or the DNA-binding A box (600 μg per mouse)] beginning as late as 24 h after surgical induction of peritonitis significantly increased survival (nonimmune IgG-treated controls = 28% vs. anti-HMGB1 antibody group = 72%, P < 0.03; GST control protein = 28% vs. A box = 68%, P < 0.03). Animals treated with either HMGB1 antagonist were protected against the development of organ injury, as evidenced by improved levels of serum creatinine and blood urea nitrogen. These observations demonstrate that specific inhibition of endogenous HMGB1 therapeutically reverses lethality of established sepsis indicating that HMGB1 inhibitors can be administered in a clinically relevant time frame.


Shock | 2006

HMGB1 signals through toll-like receptor (TLR) 4 and TLR2.

Man Yu; Haichao Wang; Aihao Ding; Douglas T. Golenbock; Eicke Latz; Christopher J. Czura; Matthew J. Fenton; Kevin J. Tracey; Huan Yang

ABSTRACT In response to bacterial endotoxin (e.g., LPS) or endogenous proinflammatory cytokines (e.g., TNF and IL-1&bgr;), innate immune cells release HMGB1, a late cytokine mediator of lethal endotoxemia and sepsis. The delayed kinetics of HMGB1 release makes it an attractive therapeutic target with a wider window of opportunity for the treatment of lethal systemic inflammation. However, the receptor(s) responsible for HMGB1-mediated production of proinflammatory cytokines has not been well characterized. Here we demonstrate that in human whole blood, neutralizing antibodies against Toll-like receptor 4 (TLR4, but not TLR2 or receptor for advanced glycation end product) dose-dependently attenuate HMGB1-induced IL-8 release. Similarly, in primary human macrophages, HMGB1-induced TNF release is dose-dependently inhibited by anti-TLR4 antibodies. In primary macrophages from knockout mice, HMGB1 activates significantly less TNF release in cells obtained from MyD88 and TLR4 knockout mice as compared with cells from TLR2 knockout and wild-type controls. However, in human embryonic kidney 293 cells transfected with TLR2 or TLR4, HMGB1 effectively induces IL-8 release only from TLR2 overexpressing cells. Consistently, anti-TLR2 antibodies dose-dependently attenuate HMGB1-induced IL-8 release in human embryonic kidney/TLR2-expressing cells and markedly reduce HMGB1 cell surface binding on murine macrophage-like RAW 264.7 cells. Taken together, our data suggest that there is a differential usage of TLR2 and TLR4 in HMGB1 signaling in primary cells and in established cell lines, adding complexity to studies of HMGB1 signaling which was not previously expected.


Journal of Experimental Medicine | 2002

Pharmacological Stimulation of the Cholinergic Antiinflammatory Pathway

Thomas R. Bernik; Steven G. Friedman; Mahendar Ochani; Robert DiRaimo; Luis Ulloa; Huan Yang; Samridhi Sudan; Christopher J. Czura; Svetlana Ivanova; Kevin J. Tracey

Efferent activity in the vagus nerve can prevent endotoxin-induced shock by attenuating tumor necrosis factor (TNF) synthesis. Termed the “cholinergic antiinflammatory pathway,” inhibition of TNF synthesis is dependent on nicotinic α-bungarotoxin-sensitive acetylcholine receptors on macrophages. Vagus nerve firing is also stimulated by CNI-1493, a tetravalent guanylhydrazone molecule that inhibits systemic inflammation. Here, we studied the effects of pharmacological and electrical stimulation of the intact vagus nerve in adult male Lewis rats subjected to endotoxin-induced shock to determine whether intact vagus nerve signaling is required for the antiinflammatory action of CNI-1493. CNI-1493 administered via the intracerebroventricular route was 100,000-fold more effective in suppressing endotoxin-induced TNF release and shock as compared with intravenous dosing. Surgical or chemical vagotomy rendered animals sensitive to TNF release and shock, despite treatment with CNI-1493, indicating that an intact cholinergic antiinflammatory pathway is required for antiinflammatory efficacy in vivo. Electrical stimulation of either the right or left intact vagus nerve conferred significant protection against endotoxin-induced shock, and specifically attenuated serum and myocardial TNF, but not pulmonary TNF synthesis, as compared with sham-operated animals. Together, these results indicate that stimulation of the cholinergic antiinflammatory pathway by either pharmacological or electrical methods can attenuate the systemic inflammatory response to endotoxin-induced shock.


Journal of Immunology | 2003

IFN-γ Induces High Mobility Group Box 1 Protein Release Partly Through a TNF-Dependent Mechanism

Beatriz Rendon-Mitchell; Mahendar Ochani; Jianhua Li; Jialian Han; Hong Wang; Huan Yang; Seenu Susarla; Christopher J. Czura; Robert A. Mitchell; Guoqian Chen; Andrew E. Sama; Kevin J. Tracey; Haichao Wang

We recently discovered that a ubiquitous protein, high mobility group box 1 protein (HMGB1), is released by activated macrophages, and functions as a late mediator of lethal systemic inflammation. To elucidate mechanisms underlying the regulation of HMGB1 release, we examined the roles of other cytokines in induction of HMGB1 release in macrophage cell cultures. Macrophage migration inhibitory factor, macrophage-inflammatory protein 1β, and IL-6 each failed to significantly induce the release of HMGB1 even at supraphysiological levels (up to 200 ng/ml). IFN-γ, an immunoregulatory cytokine known to mediate the innate immune response, dose-dependently induced the release of HMGB1, TNF, and NO, but not other cytokines such as IL-1α, IL-1β, or IL-6. Pharmacological suppression of TNF activity with neutralizing Abs, or genetic disruption of TNF expression (TNF knockout) partially (50–60%) inhibited IFN-γ-mediated HMGB1 release. AG490, a specific inhibitor for Janus kinase 2 of the IFN-γ signaling pathway, dose-dependently attenuated IFN-γ-induced HMGB1 release. These data suggest that IFN-γ plays an important role in the regulation of HMGB1 release through a TNF- and Janus kinase 2-dependent mechanism.


Critical Care Medicine | 2007

Selective α7-nicotinic acetylcholine receptor agonist GTS-21 improves survival in murine endotoxemia and severe sepsis

Valentin A. Pavlov; Mahendar Ochani; Lihong Yang; Margot Gallowitsch-Puerta; Kanta Ochani; Xinchun Lin; Jelena Levi; William R. Parrish; Mauricio Rosas-Ballina; Christopher J. Czura; Gregory J. LaRosa; Edmund J. Miller; Kevin J. Tracey; Yousef Al-Abed

Objective:Tumor necrosis factor and high mobility group box 1 are critical cytokine mediators of inflammation. The efferent vagus nerve inhibits cytokine release through &agr;7-nicotinic acetylcholine receptor-mediated cholinergic signaling. Here we studied whether GTS-21, a selective &agr;7-nicotinic acetylcholine receptor agonist, inhibits proinflammatory cytokines in vitro and in vivo and improves survival in murine endotoxemia and severe sepsis. Design:Randomized and controlled in vitro and in vivo study. Settings:Research laboratory and animal facility rooms. Subjects:RAW 264.7 cells and BALB/c mice treated with endotoxin or subjected to cecal ligation and puncture (CLP). Interventions:RAW 264.7 cells were exposed to endotoxin (4 ng/mL or 10 ng/mL) in the presence or absence of GTS-21 (1–100 &mgr;M), and tumor necrosis factor and high mobility group box 1 release and nuclear factor-&kgr;B activation were analyzed. Mice were treated with GTS-21 (0.4 mg/kg or 4 mg/kg, intraperitoneally) or saline 30 mins before endotoxin (6 mg/kg, intraperitoneally), and serum tumor necrosis factor was analyzed 1.5 hrs after the onset of endotoxemia. In survival experiments, mice were treated with GTS-21 (0.4 or 4.0 mg/kg, intraperitoneally) or saline 30 mins before and 6 hrs after endotoxin and then twice daily for 3 days. Severe sepsis was induced by CLP. Mice were treated with GTS-21 (4 mg/kg) or saline immediately and 6 hrs and 24 hrs after CLP, and serum high mobility group box 1 was analyzed 30 hrs after CLP. In survival experiments, GTS-21 (0.4 or 4 mg/kg) treatment was initiated 24 hrs after CLP and continued twice daily for 3 days. Measurements and Main Results:GTS-21 dose-dependently inhibited tumor necrosis factor and high mobility group box 1 release and nuclear factor-&kgr;B activation in vitro. GTS-21 (4 mg/kg) significantly inhibited serum tumor necrosis factor during endotoxemia and improved survival (p < .0001). GTS-21 (4 mg/kg) significantly inhibited serum high mobility group box 1 levels in CLP mice and improved survival (p < .0006). Conclusion:These findings are of interest for the development of &agr;7-nicotinic acetylcholine receptor agonists as a new class of anti-inflammatory therapeutics.


Critical Care Medicine | 2007

Transcutaneous vagus nerve stimulation reduces serum high mobility group box 1 levels and improves survival in murine sepsis

Jared M. Huston; Margot Gallowitsch-Puerta; Mahendar Ochani; Kanta Ochani; Renqi Yuan; Mauricio Rosas-Ballina; Mala Ashok; Richard S. Goldstein; Sangeeta Chavan; Valentin A. Pavlov; Christine N. Metz; Huan Yang; Christopher J. Czura; Haichao Wang; Kevin J. Tracey

Objective: Electrical vagus nerve stimulation inhibits proinflammatory cytokine production and prevents shock during lethal systemic inflammation through an [alpha]7 nicotinic acetylcholine receptor ([alpha]7nAChR)‐dependent pathway to the spleen, termed the cholinergic anti‐inflammatory pathway. Pharmacologic [alpha]7nAChR agonists inhibit production of the critical proinflammatory mediator high mobility group box 1 (HMGB1) and rescue mice from lethal polymicrobial sepsis. Here we developed a method of transcutaneous mechanical vagus nerve stimulation and then investigated whether this therapy can protect mice against sepsis lethality. Design: Prospective, randomized study. Setting: Institute‐based research laboratory. Subjects: Male BALB/c mice. Interventions: Mice received lipopolysaccharide to induce lethal endotoxemia or underwent cecal ligation and puncture to induce polymicrobial sepsis. Mice were then randomized to receive electrical, transcutaneous, or sham vagus nerve stimulation and were followed for survival or euthanized at predetermined time points for cytokine analysis. Measurements and Main Results: Transcutaneous vagus nerve stimulation dose‐dependently reduced systemic tumor necrosis factor levels during lethal endotoxemia. Treatment with transcutaneous vagus nerve stimulation inhibited HMGB1 levels and improved survival in mice with polymicrobial sepsis, even when administered 24 hrs after the onset of disease. Conclusions: Transcutaneous vagus nerve stimulation is an efficacious treatment for mice with lethal endotoxemia or polymicrobial sepsis.


Molecular Medicine | 2008

Modulation of TNF release by choline requires alpha7 subunit nicotinic acetylcholine receptor-mediated signaling.

William R. Parrish; Mauricio Rosas-Ballina; Margot Gallowitsch-Puerta; Mahendar Ochani; Kanta Ochani; Lihong Yang; LaQueta Hudson; Xinchun Lin; Nirav B Patel; Sarah M. Johnson; Sangeeta Chavan; Richard S. Goldstein; Christopher J. Czura; Edmund J. Miller; Yousef Al-Abed; Kevin J. Tracey; Valentin A. Pavlov

The α7 subunit-containing nicotinic acetylcholine receptor (α7nAChR) is an essential component in the vagus nerve-based cholinergic anti-inflammatory pathway that regulates the levels of TNF, high mobility group box 1 (HMGB1), and other cytokines during inflammation. Choline is an essential nutrient, a cell membrane constituent, a precursor in the biosynthesis of acetylcholine, and a selective natural α7nAChR agonist. Here, we studied the anti-inflammatory potential of choline in murine endotoxemia and sepsis, and the role of the α7nAChR in mediating the suppressive effect of choline on TNF release. Choline (0.1–50 mM) dose-dependently suppressed TNF release from endotoxin-activated RAW macrophage-like cells, and this effect was associated with significant inhibition of NF-κB activation. Choline (50 mg/kg, intraperitoneally (i.p.)) treatment prior to endotoxin administration in mice significantly reduced systemic TNF levels. In contrast to its TNF suppressive effect in wild type mice, choline (50 mg/kg, i.p.) failed to inhibit systemic TNF levels in α7nAChR knockout mice during endotoxemia. Choline also failed to suppress TNF release from endotoxin-activated peritoneal macrophages isolated from α7nAChR knockout mice. Choline treatment prior to endotoxin resulted in a significantly improved survival rate as compared with saline-treated endotoxemic controls. Choline also suppressed HMGB1 release in vitro and in vivo, and choline treatment initiated 24 h after cecal ligation and puncture (CLP)-induced polymicrobial sepsis significantly improved survival in mice. In addition, choline suppressed TNF release from endotoxin-activated human whole blood and macrophages. Collectively, these data characterize the anti-inflammatory efficacy of choline and demonstrate that the modulation of TNF release by choline requires α7nAChR-mediated signaling.


Shock | 2006

Elevated high-mobility group box 1 levels in patients with cerebral and myocardial ischemia

Richard S. Goldstein; Margot Gallowitsch-Puerta; Lihong Yang; Mauricio Rosas-Ballina; Jared M. Huston; Christopher J. Czura; David C. Lee; Mae F. Ward; Annette Bruchfeld; Haichao Wang; Martin Lesser; Adam L. Church; Adam Litroff; Andrew E. Sama; Kevin J. Tracey

ABSTRACT Cerebral and myocardial ischemia, two of the leading causes of morbidity and mortality worldwide, are associated with inflammation that can lead to multiple organ failure and death. High-mobility group box 1(HMGB1), a recently described mediator of lethal systemic inflammation, has been detected in individuals with severe sepsis and hemorrhagic shock, but its role during ischemic injury in humans is unknown. To determine whether systemic HMGB1 levels are elevated after ischemic injury, a prospective observational study was performed in subjects with a diagnosis of either Acute Coronary Syndrome (ACS) or cerebral vascular ischemia (transient ischemic attack or cerebral vascular accident). Subjects (n, 16; age [mean], 67 ± 16.3 years) were enrolled in the North Shore-LIJ emergency department within 24 h of symptom onset. Blood samples were collected, and HMGB1 levels analyzed by Western blot analysis using previously described methods (Wang et al. Science. 1999). Control samples were obtained from healthy age- and sex-matched volunteers (n, 16; age [mean], 68 ± 15.8 years). Here, we report that serum HMGB1 levels were significantly elevated in both myocardial ischemia subjects (myocardial control serum HMGB1, 1.94 ± 2.05 ng/mL, vs. myocardial ischemia serum HMGB1, 159 ± 54.3 ng/mL; P < 0.001); and in cerebral ischemia subjects (cerebral control serum HMGB1, 16.8 ± 10.9 ng/mL, vs. cerebral ischemia serum HMGB1, 218 ± 18.8 ng/mL; P < 0.001). These results suggest that systemic HMGB1 levels are elevated in human ischemic disease.


Journal of Endotoxin Research | 2002

HMGB1 as a cytokine and therapeutic target

Huan Yang; Haichao Wang; Christopher J. Czura; Kevin J. Tracey

HMGB1 is an abundant nuclear and cytoplasmic protein present in mammalian cells. It is traditionally known as a DNA binding protein involved in maintenance of nucleosome structure and regulation of gene transcription. Beyond these intracellular roles, we recently discovered that HMGB1 is released from activated macrophages and functions as a late mediator of lethal endotoxemia. Addition of HMGB1 to macrophage cultures activates cytokine release. When released into the extracellular milieu, HMGB1 causes systemic inflammatory responses including acute lung injury, epithelial barrier dysfunction, and death. Passive immunization with anti-HMGB1 antibodies confers significant protection against lethality induced by LPS administration and sepsis caused by cecal perforation in mice. Truncation of HMGB1 into individual structural domains revealed that the HMGB1 A box, a DNA-binding motif, specifically antagonizes the activity of HMGB1 and rescues mice from lethal sepsis caused by cecal perforation. Thus, strategies that target HMGB1 with specific antibodies or antagonists have potential for treating lethal systemic inflammatory diseases characterized by excessive HMGB1 release.

Collaboration


Dive into the Christopher J. Czura's collaboration.

Top Co-Authors

Avatar

Kevin J. Tracey

The Feinstein Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Mahendar Ochani

The Feinstein Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Huan Yang

The Feinstein Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Jared M. Huston

The Feinstein Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Haichao Wang

North Shore University Hospital

View shared research outputs
Top Co-Authors

Avatar

Kanta Ochani

The Feinstein Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Mauricio Rosas-Ballina

The Feinstein Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Valentin A. Pavlov

The Feinstein Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Luis Ulloa

North Shore University Hospital

View shared research outputs
Top Co-Authors

Avatar

Margot Gallowitsch-Puerta

The Feinstein Institute for Medical Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge