Network


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

Hotspot


Dive into the research topics where Andrew E. Sama is active.

Publication


Featured researches published by Andrew E. Sama.


Surgery | 1999

Proinflammatory cytokines (tumor necrosis factor and interleukin 1) stimulate release of high mobility group protein-1 by pituicytes

Haichao Wang; Jaideep M. Vishnubhakat; Ona Bloom; Minghuang Zhang; Michael Ombrellino; Andrew E. Sama; Kevin J. Tracey

BACKGROUNDnCytokines mediate the metabolic and physiologic responses to injury and infection. Anterior pituitary cells express receptors for tumor necrosis factor (TNF) and interleukin 1 (IL-1), which can signal these cells to release corticotropin, growth hormone, and cytokines such as IL-1 and macrophage migration inhibitory factor. This interaction provides an important link between the immune system and the neuroendocrine system. We reasoned that pituicytes activated with TNF or IL-1 might release previously unrecognized factors that could participate in this signaling from the neuroendocrine to the immune system.nnnMETHODSnProteins released from rat pituicytes (GH3) after stimulation with proinflammatory cytokines were identified by N-terminal amino acid sequencing. Polyclonal antibodies against a peptide corresponding to the N-terminal amino acid sequence were generated and used to determine the kinetics of protein release.nnnRESULTSnCytokine stimulation induced the release of a 30-kd protein from rat pituicytes. After the protein was isolated and the N-terminal amino acid sequence determined, a protein database analysis revealed that it is high mobility group-1 (HMG-1) protein. TNF and IL-1 induced the release of HMG-1 from pituicytes in a time- and dose-dependent manner. Interferon gamma alone did not induce the release of HMG-1, but it enhanced TNF-induced HMG-1 release.nnnCONCLUSIONnStimulation of pituicytes by TNF or IL-1 induces the release of HMG-1, which may participate in the regulation of neuroendocrine and immune responses to infection or injury.


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.


Journal of Leukocyte Biology | 2004

Bacterial endotoxin stimulates macrophages to release HMGB1 partly through CD14- and TNF-dependent mechanisms.

Guoqian Chen; Jianhua Li; Mahendar Ochani; Beatriz Rendon-Mitchell; Xiaoling Qiang; Seenu Susarla; Luis Ulloa; Huan Yang; Saijun Fan; Sanna M. Goyert; Ping Wang; Kevin J. Tracey; Andrew E. Sama; Haichao Wang

Bacterial endotoxin [lipopolysaccharide (LPS)] stimulates macrophages to sequentially release early [tumor necrosis factor (TNF)] and late [high mobility group box 1 (HMGB1)] proinflammatory cytokines. The requirement of CD14 and mitogen‐activated protein kinases [MAPK; e.g., p38 and extracellular signal‐regulated kinase (ERK)1/2] for endotoxin‐induced TNF production has been demonstrated previously, but little is known about their involvement in endotoxin‐mediated HMGB1 release. Here, we demonstrated that genetic disruption of CD14 expression abrogated LPS‐induced TNF production but only partially attenuated LPS‐induced HMGB1 release in cultures of primary murine peritoneal macrophages. Pharmacological suppression of p38 or ERK1/2 MAPK with specific inhibitors (SB203580, SB202190, U0126, or PD98059) significantly attenuated LPS‐induced TNF production but failed to inhibit LPS‐induced HMGB1 release. Consistently, an endogenous, immunosuppressive molecule, spermine, failed to inhibit LPS‐induced activation of p38 MAPK and yet, still significantly attenuated LPS‐mediated HMGB1 release. Direct suppression of TNF activity with neutralizing antibodies or genetic disruption of TNF expression partially attenuated HMGB1 release from macrophages induced by LPS at lower concentrations (e.g., 10 ng/ml). Taken together, these data suggest that LPS stimulates macrophages to release HMGB1 partly through CD14‐ and TNF‐dependent mechanisms.


Journal of Immunology | 2007

A Cardiovascular Drug Rescues Mice from Lethal Sepsis by Selectively Attenuating a Late-Acting Proinflammatory Mediator, High Mobility Group Box 1

Jianhua Li; Mala Ashok; Rongqian Wu; Dazhi Chen; Lihong Yang; Huan Yang; Kevin J. Tracey; Ping Wang; Andrew E. Sama; Haichao Wang

The pathogenesis of sepsis is mediated in part by bacterial endotoxin, which stimulates macrophages/monocytes to sequentially release early (e.g., TNF, IL-1, and IFN-γ) and late (e.g., high mobility group box 1 (HMGB1) protein) proinflammatory cytokines. The recent discovery of HMGB1 as a late mediator of lethal sepsis has prompted investigation for development of new experimental therapeutics. We found that many steroidal drugs (such as dexamethasone and cortisone) and nonsteroidal anti-inflammatory drugs (such as aspirin, ibuprofen, and indomethacin) failed to influence endotoxin-induced HMGB1 release even at superpharmacological concentrations (up to 10–25 μM). However, several steroid-like pigments (tanshinone I, tanshinone IIA, and cryptotanshinone) of a popular Chinese herb, Danshen (Salvia miltiorrhiza), dose dependently attenuated endotoxin-induced HMGB1 release in macrophage/monocyte cultures. A water-soluble tanshinone IIA sodium sulfonate derivative (TSNIIA-SS), which has been widely used as a Chinese medicine for patients with cardiovascular disorders, selectively abrogated endotoxin-induced HMGB1 cytoplasmic translocation and release in a glucocorticoid receptor-independent manner. Administration of TSNIIA-SS significantly protected mice against lethal endotoxemia and rescued mice from lethal sepsis even when the first dose was given 24 h after the onset of sepsis. The therapeutic effects were partly attributable to attenuation of systemic accumulation of HMGB1 (but not TNF and NO) and improvement of cardiovascular physiologic parameters (e.g., decrease in total peripheral vascular resistance and increase in cardiac stroke volume) in septic animals. Taken together, these data re-enforce the pathogenic role of HMGB1 in lethal sepsis, and support a therapeutic potential for TSNIIA-SS in the treatment of human sepsis.


PLOS ONE | 2007

A Major Ingredient of Green Tea Rescues Mice from Lethal Sepsis Partly by Inhibiting HMGB1

Mala Ashok; Jianhua Li; Huan Yang; Andrew E. Sama; Haichao Wang

Background The pathogenesis of sepsis is mediated in part by bacterial endotoxin, which stimulates macrophages/monocytes to sequentially release early (e.g., TNF, IL-1, and IFN-γ) and late (e.g., HMGB1) pro-inflammatory cytokines. Our recent discovery of HMGB1 as a late mediator of lethal sepsis has prompted investigation for development of new experimental therapeutics. We previously reported that green tea brewed from the leaves of the plant Camellia sinensis is effective in inhibiting endotoxin-induced HMGB1 release. Methods and Findings Here we demonstrate that its major component, (-)-epigallocatechin-3-gallate (EGCG), but not catechin or ethyl gallate, dose-dependently abrogated HMGB1 release in macrophage/monocyte cultures, even when given 2–6 hours post LPS stimulation. Intraperitoneal administration of EGCG protected mice against lethal endotoxemia, and rescued mice from lethal sepsis even when the first dose was given 24 hours after cecal ligation and puncture. The therapeutic effects were partly attributable to: 1) attenuation of systemic accumulation of proinflammatory mediator (e.g., HMGB1) and surrogate marker (e.g., IL-6 and KC) of lethal sepsis; and 2) suppression of HMGB1-mediated inflammatory responses by preventing clustering of exogenous HMGB1 on macrophage cell surface. Conclusions Taken together, these data suggest a novel mechanism by which the major green tea component, EGCG, protects against lethal endotoxemia and sepsis.


Journal of Cerebral Blood Flow and Metabolism | 2010

Peripheral Administration of Fetuin-A Attenuates Early Cerebral Ischemic Injury in Rats

Haichao Wang; Shu Zhu; Jianhua Li; Jason D'Amore; Mary F. Ward; Huan Yang; Rongqian Wu; Willi Jahnen-Dechent; Kevin J. Tracey; Ping Wang; Andrew E. Sama

Cerebral ischemia-elicited inflammatory responses are driven by inflammatory mediators produced both by central (e.g., neurons and microglia) and infiltrating peripheral immune cells (e.g., macrophage/monocyte), and contribute to the evolution of tissue injury. A ubiquitous molecule, spermine, is released from injured cells, and counter-regulates release of various proinflammatory cytokines. However, the spermine-mediated anti-inflammatory activities are dependent on the availability of fetuin-A, a liver-derived negative acute-phase protein. Using an animal model of focal cerebral ischemia (i.e., permanent middle cerebral artery occlusion, MCAo), we found that levels of fetuin-A in the ischemic brain tissue were elevated in a time-dependent manner, starting between 2 and 6 h, peaking around 24 to 48 h, and returning to baseline 72 h after MCAo. When administered peripherally, exogenous fetuin-A gained entry across the BBB into the ischemic brain tissue, and dose dependently reduced brain infarct volume at 24 h after MCAo. Meanwhile, fetuin-A effectively attenuated (i) ischemia-induced HMGB1 depletion from the ischemic core; (ii) activation of centrally (e.g., microglia) and peripherally derived immune cells (e.g., macrophage/monocytes); and (iii) TNF production in ischemic brain tissue. Taken together, these experimental data suggest that fetuin-A protects against early cerebral ischemic injury partly by attenuating the brain inflammatory response.


Medical Hypotheses | 2004

Pathogenic role of HMGB1 in SARS

Guoqian Chen; Dazhi Chen; Jianhua Li; Christopher J. Czura; Kevin J. Tracey; Andrew E. Sama; Haichao Wang

n Abstractn n High mobility group box 1 protein (HMGB1) is released by necrotic cells or activated macrophages/monocytes, and functions as a late mediator of lethal systemic and local pulmonary inflammation. Passive immunization with anti-HMGB1 antibodies confers significant protection against lethal endotoxemia, sepsis, and acute lung injury, even when antibodies are administered after the onset of these diseases. In light of observations that three Chinese herbal formulations recommended for treatment of severe acute respiratory syndrome (SARS) specifically inhibited the release of HMGB1 from innate immune cells, we hypothesize that HMGB1 might occupy a pathogenic role in SARS by mediating an injurious pulmonary inflammatory response.n n


Current Opinion in Pharmacology | 2006

Role of HMGB1 in cardiovascular diseases

Andrew E. Sama; Haichao Wang


Annals of Emergency Medicine | 2001

Comparison of the auditory and visual privacy of emergency department treatment areas with curtains versus those with solid walls

David Barlas; Andrew E. Sama; Mary F. Ward; Martin Lesser


Journal of Nutrition | 2006

The Aqueous Extract of a Popular Herbal Nutrient Supplement, Angelica sinensis, Protects Mice against Lethal Endotoxemia and Sepsis

Haichao Wang; Jianhua Li; Beatriz Rendon-Mitchell; Mahendar Ochani; Mala Ashok; Lihong Yang; Huan Yang; Kevin J. Tracey; Ping Wang; Andrew E. Sama

Collaboration


Dive into the Andrew E. Sama's collaboration.

Top Co-Authors

Avatar

Haichao Wang

North Shore University Hospital

View shared research outputs
Top Co-Authors

Avatar

Kevin J. Tracey

The Feinstein Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Jianhua Li

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
Top Co-Authors

Avatar

Ping Wang

The Feinstein Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mahendar Ochani

The Feinstein Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Christopher J. Czura

The Feinstein Institute for Medical Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge