Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Crit Care ; 19: 71, 2015 Mar 03.
Article in English | MEDLINE | ID: mdl-25887472

ABSTRACT

INTRODUCTION: Two recent, independent, studies conducted novel metabolomics analyses relevant to human sepsis progression; one was a human model of endotoxin (lipopolysaccharide (LPS)) challenge (experimental endotoxemia) and the other was community acquired pneumonia and sepsis outcome diagnostic study (CAPSOD). The purpose of the present study was to assess the concordance of metabolic responses to LPS and community-acquired sepsis. METHODS: We tested the hypothesis that the patterns of metabolic response elicited by endotoxin would agree with those in clinical sepsis. Alterations in the plasma metabolome of the subjects challenged with LPS were compared with those of sepsis patients who had been stratified into two groups: sepsis patients with confirmed infection and non-infected patients who exhibited systemic inflammatory response syndrome (SIRS) criteria. Common metabolites between endotoxemia and both these groups were individually identified, together with their direction of change and functional classifications. RESULTS: Response to endotoxemia at the metabolome level elicited characteristics that agree well with those observed in sepsis patients despite the high degree of variability in the response of these patients. Moreover, some distinct features of SIRS have been identified. Upon stratification of sepsis patients based on 28-day survival, the direction of change in 21 of 23 metabolites was the same in endotoxemia and sepsis survival groups. CONCLUSIONS: The observed concordance in plasma metabolomes of LPS-treated subjects and sepsis survivors strengthens the relevance of endotoxemia to clinical research as a physiological model of community-acquired sepsis, and gives valuable insights into the metabolic changes that constitute a homeostatic response. Furthermore, recapitulation of metabolic differences between sepsis non-survivors and survivors in LPS-treated subjects can enable further research on the development and assessment of rational clinical therapies to prevent sepsis mortality. Compared with earlier studies which focused exclusively on comparing transcriptional dynamics, the distinct metabolomic responses to systemic inflammation with or without confirmed infection, suggest that the metabolome is much better at differentiating these pathophysiologies. Finally, the metabolic changes in the recovering patients shift towards the LPS-induced response pattern strengthening the notion that the metabolic, as well as transcriptional responses, characteristic to the endotoxemia model represent necessary and "healthy" responses to infectious stimuli.


Subject(s)
Endotoxemia/blood , Inflammation/blood , Metabolome/physiology , Sepsis/blood , Amino Acids/blood , Carbohydrates/blood , Electrolytes/blood , Humans , Lipid Metabolism , Lipids/blood , Lipopolysaccharides/pharmacology , Retrospective Studies , Systemic Inflammatory Response Syndrome/blood
2.
Shock ; 43(2): 133-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25526373

ABSTRACT

INTRODUCTION: Human injury or infection induces systemic inflammation with characteristic neuroendocrine responses. Fluctuations in autonomic function during inflammation are reflected by beat-to-beat variation in heart rate, termed heart rate variability (HRV). In the present study, we determine threshold doses of endotoxin needed to induce observable changes in markers of systemic inflammation, investigate whether metrics of HRV exhibit a differing threshold dose from other inflammatory markers, and investigate the size of data sets required for meaningful use of multiscale entropy (MSE) analysis of HRV. METHODS: Healthy human volunteers (n = 25) were randomized to receive placebo (normal saline) or endotoxin/lipopolysaccharide (LPS): 0.1, 0.25, 0.5, 1.0, or 2.0 ng/kg administered intravenously. Vital signs were recorded every 30 min for 6 h and then at 9, 12, and 24 h after LPS. Blood samples were drawn at specific time points for cytokine measurements. Heart rate variability analysis was performed using electrocardiogram epochs of 5 min. Multiscale entropy for HRV was calculated for all dose groups to scale factor 40. RESULTS: The lowest significant threshold dose was noted in core temperature at 0.25 ng/kg. Endogenous tumor necrosis factor α and interleukin 6 were significantly responsive at the next dosage level (0.5 ng/kg) along with elevations in circulating leukocytes and heart rate. Responses were exaggerated at higher doses (1 and 2 ng/kg). Time domain and frequency domain HRV metrics similarly suggested a threshold dose, differing from placebo at 1.0 and 2.0 ng/kg, below which no clear pattern in response was evident. By applying repeated-measures analysis of variance across scale factors, a significant decrease in MSE was seen at 1.0 and 2.0 ng/kg by 2 h after exposure to LPS. Although not statistically significant below 1.0 ng/kg, MSE unexpectedly decreased across all groups in an orderly dose-response pattern not seen in the other outcomes. CONCLUSIONS: By using repeated-measures analysis of variance across scale factors, MSE can detect autonomic change after LPS challenge in a group of 25 subjects using electrocardiogram epochs of only 5 min and entropy analysis to scale factor of only 40, potentially facilitating MSE's wider use as a research tool or bedside monitor. Traditional markers of inflammation generally exhibit threshold dose behavior. In contrast, MSE's apparent continuous dose-response pattern, although not statistically verifiable in this study, suggests a potential subclinical harbinger of infectious or other insult. The possible derangement of autonomic complexity prior to or independent of the cytokine surge cannot be ruled out. Future investigation should focus on confirmation of overt inflammation following observed decreases in MSE in a clinical setting.


Subject(s)
Endotoxins/pharmacology , Heart Rate/drug effects , Inflammation/physiopathology , Adult , Body Temperature/drug effects , Cytokines/biosynthesis , Dose-Response Relationship, Drug , Electrocardiography , Endotoxins/administration & dosage , Entropy , Female , Humans , Inflammation Mediators/blood , Leukocyte Count , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/pharmacology , Male , Random Allocation , Young Adult
3.
Shock ; 42(6): 499-508, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25061728

ABSTRACT

In this meta-study, we aimed to integrate biological insights gained from two levels of -omics analyses on the response to systemic inflammation induced by lipopolysaccharide in humans. We characterized the interplay between plasma metabolite compositions and transcriptional response of leukocytes through integration of transcriptomics with plasma metabonomics. We hypothesized that the drastic changes in the immediate environment of the leukocytes might have an adaptive effect on shaping their transcriptional response in conjunction with the initial inflammatory stimuli. Indeed, we observed that leukocytes, most notably, tune the activity of lipid- and protein-associated processes at the transcriptional level in accordance with the fluctuations in metabolite compositions of surrounding plasma. A closer look into the transcriptional control of only metabolic pathways uncovered alterations in bioenergetics and defenses against oxidative stress closely associated with mitochondrial dysfunction and shifts in energy production observed during inflammatory processes.


Subject(s)
Endotoxemia/metabolism , Endotoxins/chemistry , Gene Expression Regulation , Adolescent , Adult , Endotoxemia/physiopathology , Female , Humans , Lipopolysaccharides/chemistry , Male , Metabolome , Mitochondria/metabolism , Oxidative Stress , Transcription, Genetic , Transcriptome , Young Adult
4.
Math Biosci ; 252: 36-44, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24680646

ABSTRACT

Analysis of heart rate variability (HRV) is a promising diagnostic technique due to the noninvasive nature of the measurements involved and established correlations with disease severity, particularly in inflammation-linked disorders. However, the complexities underlying the interpretation of HRV complicate understanding the mechanisms that cause variability. Despite this, such interpretations are often found in literature. In this paper we explored mathematical modeling of the relationship between the autonomic nervous system and the heart, incorporating basic mechanisms such as perturbing mean values of oscillating autonomic activities and saturating signal transduction pathways to explore their impacts on HRV. We focused our analysis on human endotoxemia, a well-established, controlled experimental model of systemic inflammation that provokes changes in HRV representative of acute stress. By contrasting modeling results with published experimental data and analyses, we found that even a simple model linking the autonomic nervous system and the heart confound the interpretation of HRV changes in human endotoxemia. Multiple plausible alternative hypotheses, encoded in a model-based framework, equally reconciled experimental results. In total, our work illustrates how conventional assumptions about the relationships between autonomic activity and frequency-domain HRV metrics break down, even in a simple model. This underscores the need for further experimental work towards unraveling the underlying mechanisms of autonomic dysfunction and HRV changes in systemic inflammation. Understanding the extent of information encoded in HRV signals is critical in appropriately analyzing prior and future studies.


Subject(s)
Autonomic Nervous System/physiopathology , Endotoxemia/physiopathology , Heart Rate/physiology , Models, Cardiovascular , Homeostasis/physiology , Humans , Neurotransmitter Agents/physiology
5.
Innate Immun ; 20(7): 774-84, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24217219

ABSTRACT

Severe traumas are associated with hypercortisolemia due to both disruption of cortisol secretion rhythm and increase in its total concentration. Understanding the effects of altered cortisol levels and rhythms on immune function is of great clinical interest, to prevent conditions such as sepsis from complicating the recovery. This in vivo study assesses the responses of circulating leukocytes to coupled dose and rhythm manipulation of cortisol, preceding an immune challenge induced by endotoxin administration. Through continuous infusion, plasma cortisol concentration was increased to and kept constant at a level associated with major physiologic stress. In response, transcriptional programming of leukocytes was altered to display a priming response before endotoxin exposure. Enhanced expression of a number of receptors and signaling proteins, as well as lowered protein translation and mitochondrial function indicated a sensitization against potential infectious threats. Despite these changes, response to endotoxin followed very similar patterns in both cortisol and saline pre-treated groups except one cluster including probe sets associated with major players regulating inflammatory response. In sum, altered dose and rhythm of plasma cortisol levels engendered priming of circulating leukocytes when preceded an immune challenge. This transcriptional program change associated with stimulated surveillance function and suppressed energy-intensive processes, emphasized permissive actions of cortisol on immune function.


Subject(s)
Endotoxins/pharmacology , Hydrocortisone/blood , Hydrocortisone/pharmacology , Leukocytes/metabolism , Adolescent , Adult , Cell Differentiation/drug effects , Female , Gene Expression Regulation/drug effects , Humans , Hydrocortisone/administration & dosage , Leukocyte Count , Leukocytes/drug effects , Lipopolysaccharides/pharmacology , Male , Microarray Analysis , Promoter Regions, Genetic/drug effects , Young Adult
6.
Ann Surg ; 259(5): 999-1006, 2014 May.
Article in English | MEDLINE | ID: mdl-23817504

ABSTRACT

OBJECTIVE: The Toll-like receptor 4 (TLR4) ligand endotoxin triggers robust systemic inflammatory responses in humans at doses equal to or greater than 1 ng/kg. In this study, we tested the hypothesis that evidence of TLR4-induced responses would be detectable in leukocytes challenged with endotoxin doses that are below the threshold needed to trigger a characteristic systemic inflammatory phenotype in humans. METHODS: Subjects were challenged with endotoxin at 1, 0.5, or 0.1 ng/kg (n = 5 per dose). Systemic responses were monitored for 24 hours. Blood samples, collected at designated intervals, were used to determine plasma cytokines levels, total and differential leukocyte counts, expression of leukocyte cell surface receptors, and changes in the leukocyte transcriptome. Western blotting was used to determine changes in leukocyte protein expression. RESULTS: We found that in vivo endotoxin at doses below 1.0 ng/kg triggers weak and variable responses in humans. In marked contrast, we show that endotoxin at a concentration as low as 0.1 ng/kg triggers a transient decline in cellular ATP levels in leukocytes. This is associated with the appearance of a unique protein expression signature in leukocytes. The protein expression signature includes 3 prominent features: (i) AMP-activated protein kinase subunit α (AMPKα) degradation, (ii) increased hypoxia inducible factor-1 (HIF-1) α expression, and (iii) autophagy, collectively indicative of a regulated metabolic response. An indistinguishable response phenotype was observed in human leukocytes treated with endotoxin in vitro. CONCLUSIONS: These data demonstrate for the first time in humans that a TLR4 ligand concentration that is below the threshold needed to trigger clinically evident systemic inflammatory manifestations initiates a transient decline in ATP levels, AMPKα degradation, HIF-1α expression, and autophagy in leukocytes. This establishes that low-grade TLR4 activation exerts control over leukocyte metabolism in the absence of systemic inflammatory indicators.


Subject(s)
Gene Expression Regulation , Immunity, Cellular/genetics , Inflammation/genetics , Leukocytes/metabolism , RNA/genetics , Toll-Like Receptor 4/genetics , Adenosine Triphosphate/metabolism , Blotting, Western , Cytokines/blood , Endotoxins/adverse effects , Humans , Inflammation/blood , Inflammation/immunology , Leukocyte Count , Leukocytes/drug effects , Leukocytes/immunology , Toll-Like Receptor 4/biosynthesis
7.
Shock ; 40(6): 519-26, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24089011

ABSTRACT

Endotoxemia induced by the administration of low-dose lipopolysaccharide (LPS) to healthy human volunteers is a well-established experimental protocol and has served as a reproducible platform for investigating the responses to systemic inflammation. Because metabolic composition of a tissue or body fluid is uniquely altered by stimuli and provides information about the dominant regulatory mechanisms at various cellular processes, understanding the global metabolic response to systemic inflammation constitutes a major part in this investigation complementing the studies undertaken so far in both clinical and systems biology fields. This article communicates the first proof-of-principle metabonomic analysis, which comprised global biochemical profiles in human plasma samples from healthy subjects given intravenous endotoxin at 2 ng/kg. Concentrations of a total of 366 plasma biochemicals were determined in archived blood samples collected from 15 endotoxin-treated subjects at five time points within 24 h after treatment and compared with control samples collected from four saline-treated subjects. Principal component analysis within this data set determined the sixth hour as a critical time point separating development and recovery phases of the LPS-induced metabolic changes. Consensus clustering of the differential metabolites identified two distinct subsets of metabolites that displayed common coherent profiles with opposing directionality. The first group of metabolites, which were mostly associated with pathways related to lipid metabolism, was upregulated within the first 6 h and downregulated by the 24th hour following LPS administration. The second group of metabolites, in contrast, was first downregulated until the sixth hour, then upregulated. Metabolites in this group were predominantly amino acids or their derivatives. In summary, nontargeted biochemical profiling and unsupervised multivariate analyses highlighted the prominent roles of lipid and protein metabolism in regulating the response to systemic inflammation while also revealing their dynamics in opposite directions.


Subject(s)
Endotoxemia/blood , Metabolome/physiology , Adolescent , Adult , Amino Acids/blood , Blood Proteins/metabolism , Down-Regulation/drug effects , Female , Humans , Lipid Metabolism/drug effects , Lipid Metabolism/physiology , Lipids/blood , Lipopolysaccharides/pharmacology , Male , Metabolome/drug effects , Metabolomics/methods , Time Factors , Up-Regulation/drug effects , Young Adult
8.
J Theor Biol ; 338: 9-15, 2013 Dec 07.
Article in English | MEDLINE | ID: mdl-23973206

ABSTRACT

The human body can be viewed as a dynamical system, with physiological states such as health and disease broadly representing steady states. From this perspective, and given inter- and intra-individual heterogeneity, an important task is identifying the propensity to transition from one steady state to another, which in practice can occur abruptly. Detecting impending transitions between steady states is of significant importance in many fields, and thus a variety of methods have been developed for this purpose, but lack of data has limited applications in physiology. Here, we propose a model-based approach towards identifying critical transitions in systemic inflammation based on a minimal amount of assumptions about the availability of data and the structure of the system. We derived a warning signal metric to identify forthcoming abrupt transitions occurring in a mathematical model of systemic inflammation with a gradually increasing bacterial load. Intervention to remove the inflammatory stimulus was successful in restoring homeostasis if undertaken when the warning signal was elevated rather than waiting for the state variables of the system themselves to begin moving to a new steady state. The proposed combination of data and model-based analysis for predicting physiological transitions represents a step forward towards the quantitative study of complex biological systems.


Subject(s)
Inflammation/physiopathology , Models, Biological , Bacterial Load , Endotoxemia/microbiology , Endotoxemia/physiopathology , Humans , Inflammation/microbiology , Systems Biology/methods
9.
Proc Natl Acad Sci U S A ; 110(9): 3507-12, 2013 Feb 26.
Article in English | MEDLINE | ID: mdl-23401516

ABSTRACT

A cornerstone of modern biomedical research is the use of mouse models to explore basic pathophysiological mechanisms, evaluate new therapeutic approaches, and make go or no-go decisions to carry new drug candidates forward into clinical trials. Systematic studies evaluating how well murine models mimic human inflammatory diseases are nonexistent. Here, we show that, although acute inflammatory stresses from different etiologies result in highly similar genomic responses in humans, the responses in corresponding mouse models correlate poorly with the human conditions and also, one another. Among genes changed significantly in humans, the murine orthologs are close to random in matching their human counterparts (e.g., R(2) between 0.0 and 0.1). In addition to improvements in the current animal model systems, our study supports higher priority for translational medical research to focus on the more complex human conditions rather than relying on mouse models to study human inflammatory diseases.


Subject(s)
Genomics , Inflammation/genetics , Acute Disease , Adolescent , Adult , Animals , Burns/genetics , Burns/pathology , Disease Models, Animal , Endotoxemia/genetics , Endotoxemia/pathology , Female , Gene Expression Regulation , Humans , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Signal Transduction/genetics , Time Factors , Wounds and Injuries/genetics , Wounds and Injuries/pathology , Young Adult
10.
PLoS One ; 8(1): e55550, 2013.
Article in English | MEDLINE | ID: mdl-23383223

ABSTRACT

As cellular variability and circadian rhythmicity play critical roles in immune and inflammatory responses, we present in this study an agent-based model of human endotoxemia to examine the interplay between circadian controls, cellular variability and stochastic dynamics of inflammatory cytokines. The model is qualitatively validated by its ability to reproduce circadian dynamics of inflammatory mediators and critical inflammatory responses after endotoxin administration in vivo. Novel computational concepts are proposed to characterize the cellular variability and synchronization of inflammatory cytokines in a population of heterogeneous leukocytes. Our results suggest that there is a decrease in cell-to-cell variability of inflammatory cytokines while their synchronization is increased after endotoxin challenge. Model parameters that are responsible for IκB production stimulated by NFκB activation and for the production of anti-inflammatory cytokines have large impacts on system behaviors. Additionally, examining time-dependent systemic responses revealed that the system is least vulnerable to endotoxin in the early morning and most vulnerable around midnight. Although much remains to be explored, proposed computational concepts and the model we have pioneered will provide important insights for future investigations and extensions, especially for single-cell studies to discover how cellular variability contributes to clinical implications.


Subject(s)
Circadian Rhythm/immunology , Endotoxemia/immunology , Inflammation/immunology , Models, Biological , Algorithms , Cell Survival/immunology , Computer Simulation , Cytokines , Endotoxemia/metabolism , Endotoxins/adverse effects , Humans , Inflammation/metabolism , Inflammation Mediators , Time Factors
11.
J Clin Monit Comput ; 27(4): 405-15, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23203205

ABSTRACT

Dysregulation of the inflammatory response is a critical component of many clinically challenging disorders such as sepsis. Inflammation is a biological process designed to lead to healing and recovery, ultimately restoring homeostasis; however, the failure to fully achieve those beneficial results can leave a patient in a dangerous persistent inflammatory state. One of the primary challenges in developing novel therapies in this area is that inflammation is comprised of a complex network of interacting pathways. Here, we discuss our approaches towards addressing this problem through computational systems biology, with a particular focus on how the presence of biological rhythms and the disruption of these rhythms in inflammation may be applied in a translational context. By leveraging the information content embedded in physiologic variability, ranging in scale from oscillations in autonomic activity driving short-term heart rate variability to circadian rhythms in immunomodulatory hormones, there is significant potential to gain insight into the underlying physiology.


Subject(s)
Endotoxemia/physiopathology , Animals , Autonomic Nervous System/physiopathology , Circadian Rhythm , Heart Rate/physiology , Homeostasis , Hormones/metabolism , Humans , Hydrocortisone/metabolism , Inflammation , Lipopolysaccharides/chemistry , Models, Theoretical , Sepsis/physiopathology , Time Factors , Transcription, Genetic , Translational Research, Biomedical/methods
12.
Crit Rev Biomed Eng ; 41(3): 205-21, 2013.
Article in English | MEDLINE | ID: mdl-24579644

ABSTRACT

Systems biology has primarily focused on studying genomics, transcriptomics, and proteomics and their dynamic interactions. These, however, represent only the potential for a biological outcome since the ultimate phenotype at the level of the eventually produced metabolites is not taken into consideration. The emerging field of metabolomics provides complementary guidance toward an integrated approach to this problem: It allows global profiling of the metabolites of a cell, tissue, or host and presents information on the actual end points of a response. A wide range of data collection methods are currently used and allow the extraction of global or tissue-specific metabolic profiles. The great amount and complexity of data that are collected require multivariate analysis techniques, but the increasing amount of work in this field has made easy-to-use analysis programs readily available. Metabolomics has already shown great potential in drug toxicity studies, disease modeling, and diagnostics and may be integrated with genomic and proteomic data in the future to provide in-depth understanding of systems, pathways, and their functionally dynamic interactions. In this review we discuss the current state of the art of metabolomics, its applications, and future potential.


Subject(s)
Metabolomics/methods , Algorithms , Animals , Biomarkers/metabolism , Computational Biology/methods , Coronary Disease/metabolism , Critical Illness , Cytokines/metabolism , Humans , Magnetic Resonance Spectroscopy/methods , Mass Spectrometry/methods , Metabolomics/trends , Neoplasms/metabolism , Obesity/metabolism , Phenotype , Principal Component Analysis , Software , Systems Biology , Toxicity Tests
14.
Crit Rev Biomed Eng ; 40(4): 313-22, 2012.
Article in English | MEDLINE | ID: mdl-23140122

ABSTRACT

The control and management of inflammation is a key aspect of clinical care for critical illnesses such as sepsis. In an ideal reaction to injury, the inflammatory response provokes a strong enough response to heal the injury and then restores homeostasis. When inflammation becomes dysregulated, a persistent inflammatory state can lead to significant deleterious effects and clinical challenges. Thus, gaining a better biological understanding of the mechanisms driving the inflammatory response is of the utmost importance. In this review, we discuss our work with the late Stephen F. Lowry to investigate systemic inflammation through systems biology of human endotoxemia. We present our efforts in modeling the human endotoxemia response with a particular focus on physiologic variability. Through modeling, with a focus ultimately on translational applications, we obtain more fundamental understanding of relevant physiological processes. And by taking advantage of the information embedded in biological rhythms, ranging in time scale from high-frequency autonomic oscillations reflected in heart rate variability to circadian rhythms in inflammatory mediators, we gain insight into the underlying physiology.


Subject(s)
Cytokines/immunology , Endotoxemia/immunology , Models, Immunological , Models, Statistical , Computer Simulation , Humans
15.
Surg Infect (Larchmt) ; 13(5): 293-9, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23072275

ABSTRACT

BACKGROUND: The normal human intravenous endotoxin model has been used for more than 50 years. It was once considered a possible model of sepsis, but, because no infection is present, it is better described as a model of systemic inflammation. We demonstrate herein that at least three of four systemic inflammatory response syndrome (SIRS) criteria are achieved with the model. METHODS: Otherwise healthy human volunteers were given Escherichia coli endotoxin 2 ng/kg intravenously. Vital signs were monitored, and blood samples were collected over time for assessment of white blood cells (WBCs), cytokines, counter-regulatory hormones, and monocyte receptors. RESULTS: The means of three variables (core temperature, heart rate, WBC) met the SIRS criteria. Compared with baseline, cytokines were elevated acutely, with tumor necrosis factor-alpha (TNFα) exhibiting temporal primacy over the other cytokines. Counter-regulatory hormones (cortisol, epinephrine) also were elevated acutely. Finally, the monocyte cell-surface receptors cluster of differentiation molecule (CD) 11b and TNF receptor-II were elevated and decreased, respectively. CONCLUSIONS: The experimental human endotoxin model satisfies SIRS criteria and probably is best described as a model of Toll-like receptor 4 agonist-induced systemic inflammation.


Subject(s)
Endotoxemia/physiopathology , Models, Biological , Systemic Inflammatory Response Syndrome/physiopathology , Adolescent , Adult , Body Temperature/drug effects , CD11b Antigen/blood , Cytokines/blood , Endotoxemia/blood , Endotoxemia/chemically induced , Endotoxemia/metabolism , Endotoxins , Epinephrine/blood , Escherichia coli , Female , Heart Rate/drug effects , Humans , Hydrocortisone/blood , Leukocyte Count , Lipopolysaccharides , Male , Receptors, Tumor Necrosis Factor, Type II/blood , Respiratory Rate/drug effects , Toll-Like Receptor 4/agonists
16.
Shock ; 38(3): 255-61, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22777119

ABSTRACT

Enteral (EN) or parenteral (PN) nutrition is used to support critically ill patients until oral feeding resumes. Enteral nutrition is assumed preferable to PN, but the differential influence on immune function is not well defined. Autonomic nervous activity is known to influence innate immune responses, and we hypothesized that EN and PN could influence both autonomic signaling and gene activation in peripheral blood monocytes (PBMs). Ten subjects (aged 18-36 years) received continuous EN or PN for 72 h. Peripheral blood monocytes were isolated from whole blood before and after continuous feeding and were analyzed for gene expression using a microarray platform. Gene expression after feeding was compared from baseline and between groups. To measure autonomic outflow, subjects also underwent heart rate variability (HRV) monitoring during feeding. Time and frequency domain HRV data were compared between groups and five orally fed subjects for changes from baseline and changes over time. During continuous EN and PN, subjects exhibited declines in both time and frequency domain HRV parameters compared with baseline and with PO subjects, indicating a loss of vagal/parasympathetic tone. However, PN feeding had a much greater influence on PBM gene expression compared with baseline than EN, including genes important to innate immunity. Continuous EN and PN are both associated with decreasing vagal tone over time, yet contribute differently to PBM gene expression, in humans. These preliminary findings support assumptions that PN imposes a systemic inflammatory risk but also imply that continuous feeding, independent of route, may impart additional risk through different mechanisms.


Subject(s)
Enteral Nutrition/adverse effects , Gene Expression/physiology , Heart Rate/physiology , Leukocytes, Mononuclear/physiology , Parenteral Nutrition/adverse effects , Adolescent , Adult , Critical Illness/therapy , DNA, Complementary/biosynthesis , Female , Humans , Immunity, Innate/genetics , Immunity, Innate/physiology , Male , Pilot Projects , Protein Array Analysis , RNA/metabolism , RNA, Complementary/biosynthesis , Systemic Inflammatory Response Syndrome/genetics , Systemic Inflammatory Response Syndrome/immunology , Young Adult
17.
Front Physiol ; 3: 222, 2012.
Article in English | MEDLINE | ID: mdl-22783197

ABSTRACT

Acute inflammation leads to organ failure by engaging catastrophic feedback loops in which stressed tissue evokes an inflammatory response and, in turn, inflammation damages tissue. Manifestations of this maladaptive inflammatory response include cardio-respiratory dysfunction that may be reflected in reduced heart rate and ventilatory pattern variabilities. We have developed signal-processing algorithms that quantify non-linear deterministic characteristics of variability in biologic signals. Now, coalescing under the aegis of the NIH Computational Biology Program and the Society for Complexity in Acute Illness, two research teams performed iterative experiments and computational modeling on inflammation and cardio-pulmonary dysfunction in sepsis as well as on neural control of respiration and ventilatory pattern variability. These teams, with additional collaborators, have recently formed a multi-institutional, interdisciplinary consortium, whose goal is to delineate the fundamental interrelationship between the inflammatory response and physiologic variability. Multi-scale mathematical modeling and complementary physiological experiments will provide insight into autonomic neural mechanisms that may modulate the inflammatory response to sepsis and simultaneously reduce heart rate and ventilatory pattern variabilities associated with sepsis. This approach integrates computational models of neural control of breathing and cardio-respiratory coupling with models that combine inflammation, cardiovascular function, and heart rate variability. The resulting integrated model will provide mechanistic explanations for the phenomena of respiratory sinus-arrhythmia and cardio-ventilatory coupling observed under normal conditions, and the loss of these properties during sepsis. This approach holds the potential of modeling cross-scale physiological interactions to improve both basic knowledge and clinical management of acute inflammatory diseases such as sepsis and trauma.

18.
Physiol Genomics ; 44(11): 607-21, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22510707

ABSTRACT

Circadian rhythmicity in mammals is primarily driven by the suprachiasmatic nucleus (SCN), often called the central pacemaker, which converts the photic information of light and dark cycles into neuronal and hormonal signals in the periphery of the body. Cells of peripheral tissues respond to these centrally mediated cues by adjusting their molecular function to optimize organism performance. Numerous systemic cues orchestrate peripheral rhythmicity, such as feeding, body temperature, the autonomic nervous system, and hormones. We propose a semimechanistic model for the entrainment of peripheral clock genes by cortisol as a representative entrainer of peripheral cells. This model demonstrates the importance of entrainer's characteristics in terms of the synchronization and entrainment of peripheral clock genes, and predicts the loss of intercellular synchrony when cortisol moves out of its homeostatic amplitude and frequency range, as has been observed clinically in chronic stress and cancer. The model also predicts a dynamic regime of entrainment, when cortisol has a slightly decreased amplitude rhythm, where individual clock genes remain relatively synchronized among themselves but are phase shifted in relation to the entrainer. The model illustrates how the loss of communication between the SCN and peripheral tissues could result in desynchronization of peripheral clocks.


Subject(s)
Biological Clocks/genetics , Hydrocortisone/pharmacology , Animals , Biological Clocks/physiology , Circadian Rhythm/physiology , Humans , Mammals , Models, Biological , Suprachiasmatic Nucleus/physiology
19.
Physiol Genomics ; 44(2): 121-9, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22128089

ABSTRACT

Endogenous glucocorticoids are secreted by the hypothalamic-pituitary-adrenal (HPA) axis in response to a wide range of stressors. Glucocorticoids exert significant downstream effects, including the regulation of many inflammatory genes. The HPA axis functions such that glucocorticoids are released in a pulsatile manner, producing ultradian rhythms in plasma glucocorticoid levels. It is becoming increasingly evident that this ultradian pulsatility is important in maintaining proper homeostatic regulation and responsiveness to stress. This is particularly interesting from a clinical perspective given that pathological dysfunctions of the HPA axis produce altered ultradian patterns. Modeling this system facilitates the understanding of how glucocorticoid pulsatility arises, how it can be lost, and the transcriptional implications of ultradian rhythms. To approach these questions, we developed a mathematical model that integrates the cyclic production of glucocorticoids by the HPA axis and their downstream effects by integrating existing models of the HPA axis and glucocorticoid pharmacodynamics. This combined model allowed us to evaluate the implications of pulsatility in homeostasis as well as in response to acute stress. The presence of ultradian rhythms allows the system to maintain a lower response to homeostatic levels of glucocorticoids, but diminished feedback within the HPA axis leads to a loss of glucocorticoid rhythmicity. Furthermore, the loss of HPA pulsatility in homeostasis correlates with a decrease in the peak output in response to an acute stressor. These results are important in understanding how cyclic glucocorticoid secretion helps maintain the responsiveness of the HPA axis.


Subject(s)
Glucocorticoids/metabolism , Hypothalamo-Hypophyseal System/metabolism , Pituitary-Adrenal System/metabolism , Transcription, Genetic , Animals , Circadian Rhythm , Feedback, Physiological , Homeostasis , Humans , Stress, Physiological/genetics
20.
Shock ; 35(3): 229-39, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21063241

ABSTRACT

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.


Subject(s)
Endotoxemia/physiopathology , Heart Rate/physiology , Models, Theoretical , Autonomic Nervous System/physiopathology , Humans , Systemic Inflammatory Response Syndrome/physiopathology , Tachycardia/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...