Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 110(9): 3507-12, 2013 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-23401516

RESUMEN

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.


Asunto(s)
Genómica , Inflamación/genética , Enfermedad Aguda , Adolescente , Adulto , Animales , Quemaduras/genética , Quemaduras/patología , Modelos Animales de Enfermedad , Endotoxemia/genética , Endotoxemia/patología , Femenino , Regulación de la Expresión Génica , Humanos , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/genética , Factores de Tiempo , Heridas y Lesiones/genética , Heridas y Lesiones/patología , Adulto Joven
2.
Crit Care ; 19: 71, 2015 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-25887472

RESUMEN

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.


Asunto(s)
Endotoxemia/sangre , Inflamación/sangre , Metaboloma/fisiología , Sepsis/sangre , Aminoácidos/sangre , Carbohidratos/sangre , Electrólitos/sangre , Humanos , Metabolismo de los Lípidos , Lípidos/sangre , Lipopolisacáridos/farmacología , Estudios Retrospectivos , Síndrome de Respuesta Inflamatoria Sistémica/sangre
3.
Ann Surg ; 259(5): 999-1006, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-23817504

RESUMEN

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.


Asunto(s)
Regulación de la Expresión Génica , Inmunidad Celular/genética , Inflamación/genética , Leucocitos/metabolismo , ARN/genética , Receptor Toll-Like 4/genética , Adenosina Trifosfato/metabolismo , Western Blotting , Citocinas/sangre , Endotoxinas/efectos adversos , Humanos , Inflamación/sangre , Inflamación/inmunología , Recuento de Leucocitos , Leucocitos/efectos de los fármacos , Leucocitos/inmunología , Receptor Toll-Like 4/biosíntesis
4.
J Theor Biol ; 338: 9-15, 2013 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-23973206

RESUMEN

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.


Asunto(s)
Inflamación/fisiopatología , Modelos Biológicos , Carga Bacteriana , Endotoxemia/microbiología , Endotoxemia/fisiopatología , Humanos , Inflamación/microbiología , Biología de Sistemas/métodos
5.
Crit Rev Biomed Eng ; 41(3): 205-21, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24579644

RESUMEN

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.


Asunto(s)
Metabolómica/métodos , Algoritmos , Animales , Biomarcadores/metabolismo , Biología Computacional/métodos , Enfermedad Coronaria/metabolismo , Enfermedad Crítica , Citocinas/metabolismo , Humanos , Espectroscopía de Resonancia Magnética/métodos , Espectrometría de Masas/métodos , Metabolómica/tendencias , Neoplasias/metabolismo , Obesidad/metabolismo , Fenotipo , Análisis de Componente Principal , Programas Informáticos , Biología de Sistemas , Pruebas de Toxicidad
6.
J Clin Monit Comput ; 27(4): 405-15, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23203205

RESUMEN

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.


Asunto(s)
Endotoxemia/fisiopatología , Animales , Sistema Nervioso Autónomo/fisiopatología , Ritmo Circadiano , Frecuencia Cardíaca/fisiología , Homeostasis , Hormonas/metabolismo , Humanos , Hidrocortisona/metabolismo , Inflamación , Lipopolisacáridos/química , Modelos Teóricos , Sepsis/fisiopatología , Factores de Tiempo , Transcripción Genética , Investigación Biomédica Traslacional/métodos
7.
Physiol Genomics ; 44(2): 121-9, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22128089

RESUMEN

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.


Asunto(s)
Glucocorticoides/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , Transcripción Genética , Animales , Ritmo Circadiano , Retroalimentación Fisiológica , Homeostasis , Humanos , Estrés Fisiológico/genética
8.
Physiol Genomics ; 44(11): 607-21, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22510707

RESUMEN

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.


Asunto(s)
Relojes Biológicos/genética , Hidrocortisona/farmacología , Animales , Relojes Biológicos/fisiología , Ritmo Circadiano/fisiología , Humanos , Mamíferos , Modelos Biológicos , Núcleo Supraquiasmático/fisiología
9.
Crit Rev Biomed Eng ; 40(4): 313-22, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23140122

RESUMEN

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.


Asunto(s)
Citocinas/inmunología , Endotoxemia/inmunología , Modelos Inmunológicos , Modelos Estadísticos , Simulación por Computador , Humanos
10.
Nature ; 437(7061): 1032-7, 2005 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-16136080

RESUMEN

Oligonucleotide and complementary DNA microarrays are being used to subclassify histologically similar tumours, monitor disease progress, and individualize treatment regimens. However, extracting new biological insight from high-throughput genomic studies of human diseases is a challenge, limited by difficulties in recognizing and evaluating relevant biological processes from huge quantities of experimental data. Here we present a structured network knowledge-base approach to analyse genome-wide transcriptional responses in the context of known functional interrelationships among proteins, small molecules and phenotypes. This approach was used to analyse changes in blood leukocyte gene expression patterns in human subjects receiving an inflammatory stimulus (bacterial endotoxin). We explore the known genome-wide interaction network to identify significant functional modules perturbed in response to this stimulus. Our analysis reveals that the human blood leukocyte response to acute systemic inflammation includes the transient dysregulation of leukocyte bioenergetics and modulation of translational machinery. These findings provide insight into the regulation of global leukocyte activities as they relate to innate immune system tolerance and increased susceptibility to infection in humans.


Asunto(s)
Biología Computacional , Perfilación de la Expresión Génica , Genómica , Inflamación/genética , Leucocitos/metabolismo , Enfermedad Aguda , Adolescente , Adulto , Endotoxinas/sangre , Endotoxinas/farmacología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inmunidad Innata/efectos de los fármacos , Inmunidad Innata/genética , Inmunidad Innata/inmunología , Inflamación/sangre , Inflamación/inducido químicamente , Inflamación/inmunología , Leucocitos/efectos de los fármacos , Leucocitos/inmunología , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/análisis , ARN Mensajero/genética , Transcripción Genética/efectos de los fármacos , Transcripción Genética/genética
11.
Physiol Genomics ; 42(1): 5-19, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20233835

RESUMEN

Severe injury and infection are associated with autonomic dysfunction. The realization that a dysregulation in autonomic function may predispose a host to excessive inflammatory processes has renewed interest in understanding the role of central nervous system (CNS) in modulating systemic inflammatory processes. Assessment of heart rate variability (HRV) has been used to evaluate systemic abnormalities and as a predictor of the severity of illness. Dissecting the relevance of neuroimmunomodulation in controlling inflammatory processes requires an understanding of the multiscale interplay between CNS and the immune response. A vital enabler in that respect is the development of a systems-based approach that integrates data across multiple scales, and models the emerging host response as the outcome of interactions of critical modules. Thus, a multiscale model of human endotoxemia, as a prototype model of systemic inflammation in humans, is proposed that integrates processes across the host from the cellular to the systemic host response level. At the cellular level interacting components are associated with elementary signaling pathways that propagate extracellular signals to the transcriptional response level. Further, essential modules associated with the neuroendocrine immune crosstalk are considered. Finally, at the systemic level, phenotypic expressions such as HRV are incorporated to assess systemic decomplexification indicative of the severity of the host response. Thus, the proposed work intends to associate acquired endocrine dysfunction with diminished HRV as a critical enabler for clarifying how cellular inflammatory processes and neural-based pathways mediate the links between patterns of autonomic control (HRV) and clinical outcomes.


Asunto(s)
Algoritmos , Sistema Nervioso Autónomo/fisiopatología , Endotoxemia/fisiopatología , Modelos Biológicos , Corticoesteroides/administración & dosificación , Sistema Nervioso Autónomo/efectos de los fármacos , Endotoxemia/sangre , Endotoxemia/etiología , Endotoxinas/administración & dosificación , Epinefrina/sangre , Expresión Génica/efectos de los fármacos , Perfilación de la Expresión Génica , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Hidrocortisona/sangre , Inflamación/inducido químicamente , Inflamación/fisiopatología , Inflamación/prevención & control , Lipopolisacáridos/administración & dosificación
12.
Crit Care Med ; 38(3): 751-8, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20081528

RESUMEN

OBJECTIVES: The intravenous administration of a bolus dose of endotoxin to healthy human subjects triggers acute systemic inflammatory responses that include cytokine production and dynamic changes in gene expression in peripheral blood leukocytes. This study sought to determine the state of clock gene expression in human peripheral blood leukocytes, and leukocyte subpopulations, challenged with in vivo endotoxin at two circadian/diurnal phases of the clock. DESIGN: Clinical and laboratory investigation. SETTING: University-based research laboratory and clinical research center. SUBJECTS: Human volunteers. INTERVENTIONS: Human subjects were administered a standard dose of endotoxin (2 ng/kg) or saline at either 0900 or 2100 hrs. Blood samples were collected at selected time points pre- and postinfusion. MEASUREMENTS AND MAIN RESULTS: Clock gene expression was determined in human peripheral blood leukocytes, neutrophils, and monocytes by quantitative real-time polymerase chain reaction. The fold change for each gene was determined by use of the 2 method. We show that endotoxin causes profound suppression of circadian clock gene expression, clearly manifested in human peripheral blood leukocytes, neutrophils, and monocytes. Clock, Cry1-2, Per3, CSNK1epsilon, Rora, and Rev-erb gene expression were all reduced by 80% to 90% with the nadir between 3 and 6 hrs postinfusion. Per1 and Per2 reached an expression nadir between 13 and 17 hrs postinfusion. The levels of plasma interleukin-6 and tumor necrosis factor peaked and then returned to baseline within 6 hrs. In contrast, clock gene expression remained suppressed for up to 17 hrs irrespective of the phase of the clock at the time of the endotoxin challenge. Endotoxin did not perturb the melatonin secretory rhythm. CONCLUSIONS: Circadian clock gene expression in peripheral blood leukocytes is dramatically altered and possibly uncoupled from the activity of the central clock during periods of acute systemic inflammation. The realignment of the central and peripheral clocks may constitute a previously unappreciated key factor affecting recovery from disease in humans.


Asunto(s)
Proteínas CLOCK/genética , Endotoxinas/sangre , Regulación de la Expresión Génica/genética , Leucocitos/inmunología , Adolescente , Adulto , Femenino , Humanos , Hidrocortisona/sangre , Interleucina-6/sangre , Masculino , Monocitos/inmunología , Neutrófilos/inmunología , Síndrome de Respuesta Inflamatoria Sistémica/genética , Síndrome de Respuesta Inflamatoria Sistémica/inmunología , Factor de Necrosis Tumoral alfa/sangre , Adulto Joven
13.
J Theor Biol ; 264(3): 1068-76, 2010 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-20307551

RESUMEN

A wide variety of modeling techniques have been applied towards understanding inflammation. These models have broad potential applications, from optimizing clinical trials to improving clinical care. Models have been developed to study specific systems and diseases, but the effect of circadian rhythms on the inflammatory response has not been modeled. Circadian rhythms are normal biological variations obeying the 24-h light/dark cycle and have been shown to play a critical role in the treatment and progression of many diseases. Several of the key components of the inflammatory response, including cytokines and hormones, have been observed to undergo significant diurnal variations in plasma concentration. It is hypothesized that these diurnal rhythms are entrained by the cyclic production of the hormones cortisol and melatonin, as stimulated by the central clock in the suprachiasmatic nucleus. Based on this hypothesis, a mathematical model of the interplay between inflammation and circadian rhythms is developed. The model is validated by its ability to reproduce diverse sets of experimental data and clinical observations concerning the temporal sensitivity of the inflammatory response.


Asunto(s)
Algoritmos , Ritmo Circadiano/fisiología , Inflamación/fisiopatología , Modelos Biológicos , Animales , Simulación por Computador , Citocinas/sangre , Endotoxemia/sangre , Endotoxemia/fisiopatología , Humanos , Hidrocortisona/sangre , Inflamación/sangre , Mediadores de Inflamación/sangre , Melatonina/sangre
14.
Crit Care ; 14(5): R177, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20929567

RESUMEN

INTRODUCTION: An endotoxin challenge, sepsis, and injury/trauma, trigger significant changes in human peripheral blood leukocytes (PBL) gene expression. In this study, we have sought to test the hypothesis that the Toll-like receptor 4 (TLR4) induced transcription patterns elicited in humans exposed to in vivo endotoxin would parallel gene expression patterns observed in trauma patients with initial non-infectious injury. In addition, we sought to identify functional modules that are commonly affected by these two insults of differing magnitude and duration. METHODS: PBL were obtained from seven adult human subject experimental groups. The groups included a group of healthy, hospitalized volunteers (n = 15), that comprised four study groups of subjects challenged with intravenous endotoxin, without or with cortisol, and two serial samplings of trauma patients (n = 5). The PBL were analyzed for gene expression using a 8,793 probe microarray platform (Gene Chip® Focus, Affymetrix). The expression of a subset of genes was determined using qPCR. RESULTS: We describe sequential selection criteria of gene expression data that identifies 445 genes that are significantly differentially expressed (both P ≤ 0.05 and > 1.2 fold-change) in PBL derived from human subjects during the peak of systemic inflammatory responses induced by in vivo endotoxin, as well as in PBL obtained from trauma patients at 1 to 12 days after admission. We identified two functional modules that are commonly represented by this analysis. The first module includes more than 50 suppressed genes that encode ribosomal proteins or translation regulators. The second module includes up-regulated genes encoding key enzymes associated with glycolysis. Finally, we show that several circadian clock genes are also suppressed in PBL of surgical ICU patients. CONCLUSIONS: We identified a group of > 400 genes that exhibit similar expression trends in PBL derived from either endotoxin-challenged subjects or trauma patients. The suppressed translational and circadian clock modules, and the upregulated glycolytic module, constitute a robust and long lasting PBL gene expression signature that may provide a tool for monitoring systemic inflammation and injury.


Asunto(s)
Lesiones Encefálicas/genética , Lesiones Encefálicas/metabolismo , Regulación de la Expresión Génica , Redes Reguladoras de Genes/fisiología , Leucocitos/metabolismo , Receptor Toll-Like 4/fisiología , Adolescente , Adulto , Lesiones Encefálicas/patología , Endotoxinas/fisiología , Femenino , Regulación de la Expresión Génica/inmunología , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Leucocitos/inmunología , Masculino , Persona de Mediana Edad , Receptor Toll-Like 4/genética , Transcripción Genética/inmunología , Adulto Joven
15.
Appl Psychophysiol Biofeedback ; 35(4): 303-15, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20635134

RESUMEN

Exposure of healthy people to lipopolysaccharide (LPS; endotoxin) produces a pro-inflammatory response, subjective symptoms, and decreased heart rate variability (HRV). Given the efficacy of HRV biofeedback (BF) for treating asthma, the large autonomic effects of HRV BF, and the link between vagus nerve activity and inflammation, we hypothesized that HRV BF would dampen the acute manifestations of systemic inflammation induced by LPS challenge. Healthy participants age 18-40 were randomly assigned to four-one-hour training sessions of either HRV BF (n = 6) or a control 15/min paced breathing condition (n = 5) prior to acute experimentally induced LPS exposure. Participants were coached to do the procedures for 10 min each at five hourly time points after LPS injection, and then 2 h later. Subjective symptoms, HRV parameters, and plasma cytokine levels were measured at each time point, 2 h afterward, and the following morning. Participants were able to perform the procedures both during four pre-exposure training sessions and while experiencing LPS-induced symptoms. The HRV BF group showed significant attenuation of the LPS-induced decline in HRV for the 6 h following LPS exposure, suggesting that HRV BF decreased autonomic dysfunction produced by LPS-induced inflammation. HRV BF also reduced symptoms of headache and eye sensitivity to light, but did not affect LPS-induced levels of pro-inflammatory cytokines or symptoms of nausea, muscle aches, or feverishness. Further evaluation of HRV BF appears to be warranted among patients with inflammatory conditions.


Asunto(s)
Sistema Nervioso Autónomo/fisiología , Biorretroalimentación Psicológica/métodos , Endotoxemia/terapia , Frecuencia Cardíaca/fisiología , Inflamación/terapia , Lipopolisacáridos/farmacología , Adolescente , Adulto , Citocinas/sangre , Electrocardiografía , Endotoxemia/sangre , Endotoxemia/inducido químicamente , Endotoxinas/farmacología , Femenino , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Inflamación/sangre , Inflamación/inducido químicamente , Masculino , Resultado del Tratamiento
16.
Ann Surg ; 249(5): 750-6, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19387330

RESUMEN

OBJECTIVE: To determine whether the acute anti-inflammatory influence of epinephrine (EPI) extends to changes in heart rate variability (HRV) induced by the prototypical inflammatory stimulus, endotoxin (lipopolysaccharide [LPS]). SUMMARY BACKGROUND DATA: HRV reflects fluctuating cardiac autonomic inputs and is acutely reduced during the systemic inflammation induced by LPS as well as during severe critical illnesses such as sepsis and traumatic injury. While EPI may diminish proinflammatory cytokine release, it is unknown whether this net anti-inflammatory activity extends to HRV. METHODS: Healthy volunteers (n = 17) were randomized to either saline + LPS (2 ng/kg) or LPS + antecedent EPI infusion (30 ng/kg/min) from -3 to 6 hours relative to LPS. HRV and blood samples were obtained before EPI and LPS as well as hourly afterward. Plasma cytokines were measured by ELISA. Statistical analysis was by repeated measures analysis of variance. This study was registered at Clinicaltrials.gov and is listed under the following ID number: NCT00753402. RESULTS: LPS acutely influenced all measured parameters of HRV including standard deviation of the average beat to beat intervals over a 5-minute period, percentage of interval differences of successive interbeat intervals greater than 50 milliseconds and square root of the mean squared differences, high frequency (HF), low frequency, low frequency/HF, and very low frequency (all P < 0.01). EPI infusion reduced the inflammatory cytokine response to LPS as measured by decreased TNFalpha, IL-6, and IL-8 (P < 0.01). Relative to the saline + LPS group, antecedent EPI infusion was associated with further reductions in parameters of HRV measuring vagal/parasympathetic activity including, percentage of interval differences of successive interbeat intervals greater than 50 milliseconds, square root of the mean squared differences, and HF (P < 0.05). CONCLUSION: Prior EPI exposure exerts anti-inflammatory influences but also may reduce vagus nerve activity. Hence, acute EPI administration may be protective against early inflammatory challenges but diminish vagal nerve responsiveness to subsequent stimuli.


Asunto(s)
Fármacos Cardiovasculares/farmacología , Epinefrina/farmacología , Frecuencia Cardíaca/efectos de los fármacos , Lipopolisacáridos/farmacología , Adolescente , Adulto , Método Doble Ciego , Femenino , Frecuencia Cardíaca/fisiología , Humanos , Masculino , Modelos Biológicos , Estrés Fisiológico/inmunología , Adulto Joven
17.
J Leukoc Biol ; 83(3): 553-7, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17984288

RESUMEN

Human injury is associated with inflammatory responses that are modulated by the acute and chronic activity of endogenous factors and exogenous interventions. A characteristic feature of chronic, severe inflammatory states is the diminished signal output variability of many organ systems, including innate immune responsiveness and endogenous neural and endocrine-mediated functions. The attenuation of signal/response variability and integration of feedback capacity may contribute to systemic and tissue-specific deterioration of function. Some well-intentioned therapies directed toward support of systemic and tissue functions may actually promote the loss of system(s) adaptability and contribute to adverse outcomes in severely stressed patients. In vivo and in silico models of stress, injury, and infection have yet to fully define the influences of ongoing stressful stimulae as well as genetic variation and epigenetic factors in the context of an evolving inflammatory state. Experimental and human models incorporating variable, antecedent stress(es) and altered neuroendocrine rhythms might approximate the altered adaptability in immune and organ function responses. Such models may also provide insights into the salient mechanisms of risk and outcome more precisely than do the constrained study conditions of current animal or human models of systemic inflammation.


Asunto(s)
Inflamación/fisiopatología , Heridas y Lesiones/fisiopatología , Animales , Asma/fisiopatología , Enfermedad Crónica , Enfermedad Crítica , Modelos Animales de Enfermedad , Humanos , Modelos Biológicos , Estrés Fisiológico/fisiopatología
18.
J Endotoxin Res ; 13(6): 358-68, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18182463

RESUMEN

Severe injury and infection are associated with autonomic dysfunction. Diminished heart rate variability (HRV) is also observed as a component of autonomic dysfunction and is induced by endotoxin administration to healthy subjects. It is established that low-dose glucocorticoid administration diminishes the systemic inflammatory manifestations of endotoxinemia but the influence of this anti-inflammatory intervention on overall autonomic dysfunction and HRV responses to endotoxin is unknown. This study was designed to assess the influence of a low-dose hydrocortisone infusion upon endotoxin-elicited systemic inflammatory responses including phenotypic features, cytokine production, and parameters of HRV. Of 19 subjects studied, nine received a continuous infusion of hydrocortisone (3 microg/kg/min continuously over 6 h) prior to intravenous administration of Escherichia coli endotoxin (2 ng/kg, CC-RE, Lot #2) while 10 healthy subjects received only the endotoxin after a 6-h period of saline control infusion. Serial determinations of vital signs, heart rate variability assessments, and cytokine levels were obtained over the subsequent 24 h. Prior cortisol infusion diminished the peak TNF-alpha (P < 0.01) and IL-6 (P < 0.0001) responses after endotoxin challenge, as compared to saline infusion controls and diminished the peak core temperature response to endotoxin (P < 0.01). In contrast to the influence of cortisol on the above parameters of systemic inflammation, the significant endotoxin-induced decreases in HRV time and frequency domains were not influenced by prior hydrocortisone treatment. Hence, alterations in autonomic dysfunction occur despite hydrocortisone attenuation of other traditional systemic manifestations of endotoxinemia. The maintenance or restoration of autonomic balance is not influenced by glucocorticoid administration.


Asunto(s)
Enfermedades del Sistema Nervioso Autónomo/fisiopatología , Endotoxinas/efectos adversos , Hidrocortisona/administración & dosificación , Hidrocortisona/farmacología , Inflamación/inducido químicamente , Inflamación/fisiopatología , Adolescente , Adulto , Enfermedades del Sistema Nervioso Autónomo/sangre , Enfermedades del Sistema Nervioso Autónomo/inducido químicamente , Enfermedades del Sistema Nervioso Autónomo/tratamiento farmacológico , Biomarcadores , Femenino , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Hidrocortisona/sangre , Hidrocortisona/uso terapéutico , Inflamación/sangre , Inflamación/tratamiento farmacológico , Lipopolisacáridos/efectos adversos , Masculino
20.
Shock ; 26(6): 538-43, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17117126

RESUMEN

Gender appears to influence systemic and organ-specific inflammatory sequelae of ischemia-reperfusion and infectious challenge in many animal models. Despite the protection provided by female gender, androgen blockade, and/or estrogen administration in such experimental studies, many questions remain regarding the influence of gender dimorphism upon human responses to injury. We hypothesized that the administration of low-dose lipopolysaccharide (LPS) to otherwise healthy, young adults would provide insights regarding the influence of gender upon physiological and innate immune system responses to a prototypic inflammatory stimulus. To this end, 72 adult subjects (48 men, aged 29 +/- 1.0 years; 24 women, aged 26 +/- 1.0 years) were prospectively evaluated before and after the i.v. administration of LPS (2 ng/kg). All subjects developed symptoms within 1.0 to 1.5 h after LPS, and the men exhibited a greater increase in core temperature (2.1 +/- 0.1 degrees C) compared with the women (1.4 +/- 0.1 degrees C) (P < 0.001). In addition, the men exhibited a greater maximum decrease in mean arterial pressure (-13.0 +/- 1.3 mmHg) compared with the women (-8 +/- 1.3 mmHg) (P < 0.02). The changes in temperature and mean arterial pressure occurred without detectable differences between the male and female cohort responses of circulating white blood cell count and cortisol or cytokine levels. These results suggest that soluble inflammatory mediators generated by in vivo endotoxin activation of the innate immune system are insufficient to explain the resultant gender-specific phenotypic differences observed in young, adult humans.


Asunto(s)
Endotoxinas/farmacología , Adulto , Citocinas/metabolismo , Estradiol/metabolismo , Femenino , Hormonas/metabolismo , Humanos , Inflamación , Lipopolisacáridos/farmacología , Masculino , Factores Sexuales , Esteroides/metabolismo , Temperatura , Testosterona/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA