Network


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

Hotspot


Dive into the research topics where Steven E. Calvano is active.

Publication


Featured researches published by Steven E. Calvano.


Journal of Experimental Medicine | 2011

A genomic storm in critically injured humans

Wenzhong Xiao; Michael Mindrinos; Junhee Seok; Joseph Cuschieri; Alex G. Cuenca; Hong Gao; Douglas L. Hayden; Laura Hennessy; Ernest E. Moore; Joseph P. Minei; Paul E. Bankey; Jeffrey L. Johnson; Jason L. Sperry; Avery B. Nathens; Timothy R. Billiar; Michael A. West; Bernard H. Brownstein; Philip H. Mason; Henry V. Baker; Celeste C. Finnerty; Marc G. Jeschke; M. Cecilia Lopez; Matthew B. Klein; Richard L. Gamelli; Nicole S. Gibran; Brett D. Arnoldo; Weihong Xu; Yuping Zhang; Steven E. Calvano; Grace P. McDonald-Smith

Critical injury in humans induces a genomic storm with simultaneous changes in expression of innate and adaptive immunity genes.


Bellman Prize in Mathematical Biosciences | 2009

Modeling endotoxin-induced systemic inflammation using an indirect response approach.

Panagiota T. Foteinou; Steven E. Calvano; Stephen F. Lowry; Ioannis P. Androulakis

A receptor mediated model of endotoxin-induced human inflammation is proposed. The activation of the innate immune system in response to the endotoxin stimulus involves the interaction between the extracellular signal and critical receptors driving downstream signal transduction cascades leading to transcriptional changes. We explore the development of an in silico model that aims at coupling extracellular signals with essential transcriptional responses through a receptor mediated indirect response model. The model consists of eight (8) variables and is evaluated in a series of biologically relevant scenarios indicative of the non-linear behavior of inflammation. Such scenarios involve a self-limited response where the inflammatory stimulus is cleared successfully; a persistent infectious response where the inflammatory instigator is not eliminated, leading to an aberrant inflammatory response, and finally, a persistent non-infectious inflammatory response that can be elicited under an overload of the pathogen-derived product; as such high dose of the inflammatory insult can disturb the dynamics of the host response leading to an unconstrained inflammatory response. Finally, the potential of the model is demonstrated by analyzing scenarios associated with endotoxin tolerance and potentiation effects.


PLOS ONE | 2010

Agent-Based Modeling of Endotoxin-Induced Acute Inflammatory Response in Human Blood Leukocytes

Xu Wei Dong; Panagiota T. Foteinou; Steven E. Calvano; Stephen F. Lowry; Ioannis P. Androulakis

Background Inflammation is a highly complex biological response evoked by many stimuli. A persistent challenge in modeling this dynamic process has been the (nonlinear) nature of the response that precludes the single-variable assumption. Systems-based approaches offer a promising possibility for understanding inflammation in its homeostatic context. In order to study the underlying complexity of the acute inflammatory response, an agent-based framework is developed that models the emerging host response as the outcome of orchestrated interactions associated with intricate signaling cascades and intercellular immune system interactions. Methodology/Principal Findings An agent-based modeling (ABM) framework is proposed to study the nonlinear dynamics of acute human inflammation. The model is implemented using NetLogo software. Interacting agents involve either inflammation-specific molecules or cells essential for the propagation of the inflammatory reaction across the system. Spatial orientation of molecule interactions involved in signaling cascades coupled with the cellular heterogeneity are further taken into account. The proposed in silico model is evaluated through its ability to successfully reproduce a self-limited inflammatory response as well as a series of scenarios indicative of the nonlinear dynamics of the response. Such scenarios involve either a persistent (non)infectious response or innate immune tolerance and potentiation effects followed by perturbations in intracellular signaling molecules and cascades. Conclusions/Significance The ABM framework developed in this study provides insight on the stochastic interactions of the mediators involved in the propagation of endotoxin signaling at the cellular response level. The simulation results are in accordance with our prior research effort associated with the development of deterministic human inflammation models that include transcriptional dynamics, signaling, and physiological components. The hypothetical scenarios explored in this study would potentially improve our understanding of how manipulating the behavior of the molecular species could manifest into emergent behavior of the overall system.


Clinical and Experimental Immunology | 2006

Nicotine exposure alters in vivo human responses to endotoxin

X. Wittebole; Sae J. Hahm; Susette M. Coyle; Ashwini Kumar; Steven E. Calvano; Stephen F. Lowry

The alpha 7 nicotinic receptor is reportedly a key element in the cholinergic anti‐inflammatory pathway. Because a prototypical ligand for this receptor is nicotine, we studied the in vivo human response to bacterial endotoxin or lipopolysaccharide (LPS) in the context of nicotine or placebo pretreatment. Twelve adult male normal subjects were studied prospectively. Six received overnight transcutaneous nicotine administration by application of a standard patch (7 mg). Six hours later, all subjects were given an intravenous dose of endotoxin (2 ng/kg) and were evaluated for an additional 24 h for circulating levels of inflammatory biomarkers, vital signs and symptoms. The nicotine subjects had elevated blood levels of the nicotine metabolite, continine, prior to and throughout the 24‐h post‐endotoxin exposure phase. Subjects receiving nicotine exhibited a significantly lower temperature response as well as attenuated cardiovascular responses for 2·5–6 h after LPS exposure. In addition, increased circulating interkeukin (IL)‐10 and cortisol levels were also noted in nicotine subjects. These data indicate an alteration in LPS‐induced systemic inflammatory responses in normal subjects exposed to transcutaneous nicotine. In this model of abbreviated inflammation, nicotine exposure attenuates the febrile response to LPS and promotes a more prominent anti‐inflammatory phenotype.


Annals of Surgery | 1989

Changes in lymphocyte number and phenotype in seven lymphoid compartments after thermal injury

B. C. Organ; A. C. Antonacci; J. Chiao; Ashwini Kumar; H. F. De Riesthal; Leslie Yuan; D. Black; Steven E. Calvano

Thermal injury is associated with dysfunction of host defense systems. The present study used flow cytometric immunofluorescence analyses to investigate changes in number and phenotype of lymphocytes in seven different lymphoid compartments at 2, 6, 12, 24, 48, and 60 days after 50% total body-surface area thermal injury in the rat. Relative to sham-injured control rats, at postburn day 2, significant lymphopenia was observed in the peripheral blood along with depletion of lymphocytes from the spleen and thymus. By day 6 after injury, lymphocytes in the bone marrow and cervical lymph nodes decreased significantly while numbers in the spleen and thymus remained depressed. Splenic and cervical node lymphocyte numbers normalized by day 12, the bone marrow and thymus numbers still were significantly lower than control, and a 6.5-fold increase in number of lymphocytes was observed in the nodes draining the burn wound, pooled axillary, brachial, inguinal, and lumbar lymph nodes. At day 24 after injury, the thymus and bone marrow virtually were depleted of lymphocytes, the mesenteric lymph nodes manifested a significant decrease, and lymphocytes in the nodes draining the burn wound continued to increase in number. This same pattern was maintained on day 48, but numbers of lymphocytes in the mesenteric nodes normalized. At day 60 after injury, lymphocyte numbers in all tissues were normalized, but the spleen and nodes draining the burn wound where increased numbers compared to control persisted. Cell-surface phenotyping was performed on all lymphoid tissues at all time intervals to determine the percentages of lymphocytes comprising the following subsets: Ia+ cells (B cells and activated T cells), T cells, T-Helper/Inducer cells (T-H/I), and T-Suppressor/Cytotoxic (T-S/C) cells. Although changes in lymphocyte subset percentages were complex, they could be divided grossly into two phases. First, all compartments showed significant phenotypic changes in the first six days after burn. With the exception of the nodes draining the burn wound and the blood, this was followed by a return towards normal on day 12. The second phase then ensued with significant phenotypic changes again occurring in most tissues from days 24 to 60 after injury. These studies demonstrate that burn injury results in dramatic alterations in lymphocyte numbers and subset percentages in different lymphoid compartments. Immune alterations observed following thermal injury may be due, in part, to a redistribution of the cellular elements responsible for generation of the immune response.


Physiological Genomics | 2011

Modeling autonomic regulation of cardiac function and heart rate variability in human endotoxemia.

Jeremy D. Scheff; Panteleimon D. Mavroudis; Steven E. Calvano; Stephen F. Lowry; Ioannis P. Androulakis

Heart rate variability (HRV), the quantification of beat-to-beat variability, has been studied as a potential prognostic marker in inflammatory diseases such as sepsis. HRV normally reflects significant levels of variability in homeostasis, which can be lost under stress. Much effort has been placed in interpreting HRV from the perspective of quantitatively understanding how stressors alter HRV dynamics, but the molecular and cellular mechanisms that give rise to both homeostatic HRV and changes in HRV have received less focus. Here, we develop a mathematical model of human endotoxemia that incorporates the oscillatory signals giving rise to HRV and their signal transduction to the heart. Connections between processes at the cellular, molecular, and neural levels are quantitatively linked to HRV. Rhythmic signals representing autonomic oscillations and circadian rhythms converge to modulate the pattern of heartbeats, and the effects of these oscillators are diminished in the acute endotoxemia response. Based on the semimechanistic model developed herein, homeostatic and acute stress responses of HRV are studied in terms of these oscillatory signals. Understanding the loss of HRV in endotoxemia serves as a step toward understanding changes in HRV observed clinically through translational applications of systems biology based on the relationship between biological processes and clinical outcomes.


Clinical and Translational Science | 2009

Translational Potential of Systems‐Based Models of Inflammation

Panagiota T. Foteinou; Steven E. Calvano; Stephen F. Lowry; Ioannis P. Androulakis

A critical goal of translational research is to convert basic science to clinically relevant actions related to disease prevention, diagnosis, and eventually enable physicians to identify and evaluate treatment strategies. Integrated initiatives are identified as valuable in uncovering the mechanism underpinning the progression of human diseases. Tremendous opportunities have emerged in the context of systems biology that aims at the deconvolution of complex phenomena to their constituent elements and the quantification of the dynamic nteractions between these components through the development of appropriate computational and mathematical models. In this review, we discuss the potential role systems‐based translation research can have in the quest to better understand and modulate the nflammatory response.


Clinical and Experimental Immunology | 2005

Expression of tumour necrosis factor receptor and Toll-like receptor 2 and 4 on peripheral blood leucocytes of human volunteers after endotoxin challenge: a comparison of flow cytometric light scatter and immunofluorescence gating

X. Wittebole; Susette M. Coyle; Ashwini Kumar; M. Goshima; Stephen F. Lowry; Steven E. Calvano

Toll‐like receptors (TLRs) are involved in the recognition of bacterial products and thus participate in the induction of the inflammatory cascade. However, much less is known about the evolution of leucocyte TLR expression during human inflammatory stress. We hypothesized that a decrease in leucocyte TLRs could account for the so‐called tolerance or hyporesponsiveness state to subsequent stimulation with bacteria‐derived products. Because of the profound monocytopenia that ensues after in vivo lipopolysaccharide (LPS) challenge, we also compared monocyte TLR expression using two different techniques of flow cytometric gating. In a first set of experiments, 17 healthy volunteers underwent LPS challenge. Blood was drawn at different time‐points and analysed by flow cytometry using light scatter gating and one‐colour analysis to assess the expression of the tumour necrosis factor receptor (TNFR) and TLR2 and TLR4 on both monocytes and granulocytes. In a second set of experiments, the assessment of those receptors was made using a more specific gating method that utilized light scatter and CD14 immunofluorescence in a two‐colour analysis. This was performed using whole blood drawn from five healthy volunteers and incubated ex vivo for different time periods with or without LPS and in 12 volunteers who underwent LPS challenge in vivo. The pattern of expression for monocyte TNFR was similar for both types of gating. Using only the light scatter gating, an initial drop of TLR 2 and 4 was observed on monocytes. By contrast, when using light scatter × immunofluorescence gating, an up‐regulation of these two receptors following both in vivo and in vitro LPS exposure was observed. LPS up‐regulates the expression of TLRs on monocytes and granulocytes. Depending upon the methodology utilized, contrasting results were obtained with respect to TLR2 and TLR4 expression. The flow cytometric gating technique used is of importance in determining cellular TLR2 and TLR4 expression, especially in blood samples exhibiting significant monocytopenia.


Shock | 2011

A physiological model for autonomic heart rate regulation in human endotoxemia.

Panagiota T. Foteinou; Steven E. Calvano; Stephen F. Lowry; Ioannis P. Androulakis

The systemic inflammatory response syndrome often accompanies critical illnesses and can be an important cause of morbidity and mortality. Marked abnormalities in cardiovascular function accompany acute illnesses manifested as sustained tachyarrhythmias, which are but one component of systemic dysregulation. The realization that cardiac pacemaker activity is under control of the autonomic nervous system has promoted the analysis of heart rate (HR) variation for assessing autonomic activities. In acute illnesses, autonomic imbalance manifesting in part as parasympathetic attenuation is associated with increased morbidity in patients who manifest systemic inflammatory response syndrome phenotype. Driven by the premise that biological phenotypes emerge as the outcome of the coordinated action of network elements across the host, a multiscale model of human endotoxemia, as a prototype model of systemic inflammation in humans, is developed that quantifies critical aspects of the complex relationship between inflammation and autonomic HR regulation. In the present study, changes in HR response to acute injury, phenotypically expressed as tachycardia, are simulated as a result of autonomic imbalance that reflects sympathetic activity excess and parasympathetic attenuation. The proposed model assesses both the anti-inflammatory and cardiovascular effects of antecedent stresses upon the systemic inflammatory manifestations of human endotoxemia as well as a series of nonlinear inflammatory relevant scenarios. Such a modeling approach provides a comprehensive conceptual framework linking inflammation and physiological complexity via a multiscale model that may advance the translational potential of systems modeling in clinical research.ABBREVIATIONS-LPS-lipopolysaccharide, endotoxin; R-endotoxin signaling receptor (TLR4); LPSR-endotoxin-TLR4 complex; mRNA,R-gene transcript of endotoxin receptor (TLR4); IKK-kinase activity; NF&kgr;Bn-nuclear concentration of NF-&kgr;B; mRNAI&kgr;Ba-gene transcript of NF-&kgr;B inhibitor (I&kgr;Ba); I&kgr;Ba-protein inhibitor I&kgr;Ba; P-transcriptional proinflammatory response; A-transcriptional anti-inflammatory response; E-transcriptional energetic response; F-cortisol; Rm-gene transcript of glucocorticoid receptor; RF-free cytosolic glucocorticoid receptor; FR -cytosolic steroid-receptor complex; FR(N)-nuclear steroid-receptor complex; EPI-epinephrine; REPI-&bgr;-adrenergic receptor; EPIR-epinephrine-adrenergic receptor complex; fP-efferent (cardiac) nerve activity; Sf-cardiac active signal; HRV-heart rate variability; A1-catecholamines at the SNS nerve ending; A2-catecholamines at the sinus node (SA); B-chemical substance at the SA of the heart; Tsym-efferent sympathetic activity; Tpar-efferent parasympathetic activity; HR-heart rate


Bellman Prize in Mathematical Biosciences | 2011

A dual negative regulation model of Toll-like receptor 4 signaling for endotoxin preconditioning in human endotoxemia.

Qian Yang; Steven E. Calvano; Stephen F. Lowry; Ioannis P. Androulakis

We discuss a model illustrating how the outcome of repeated endotoxin administration experiments can emerge as a natural consequence of the tightly regulated signaling pathways and also highlight the importance of a dual negative feedback regulation including PI3K/Akt and IRAK-M (IRAK3). We identify the relative time scales of the onset and the magnitude of the stimulus as key determinants of outcome in repeated administration experiments. The results of our simulations involve potentiated response, tolerance, and protective tolerance. Moreover, the knockout of negative regulators shows that IRAK-M is a necessary and sufficient factor for generation of endotoxin tolerance (ET). The effects of the knockout of IRAK-M gene or administration of PI3K inhibitor do yield predictions that have been verified experimentally. Finally, the pretreatment with PI3K inhibitor reveals the interaction between these two negative regulations.

Collaboration


Dive into the Steven E. Calvano's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bernard H. Brownstein

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Celeste C. Finnerty

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

David G. Camp

Pacific Northwest National Laboratory

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge