15 research outputs found

    Role of Calcium in Inflammation: Relevance to Alzheimer\u27s Disease

    Get PDF
    Alzheimer’s disease (AD) is neuropathologically characterized by excessive beta -amyloid (Abeta)plaques and neurofibrillary tangles composed of hyperphosphorylated tau in the brain. Although the etiology of genetic cases of AD has been attributed to mutations in presenilin and amyloid precursor protein (APP) genes, in most sporadic cases of AD, the etiology is still unknown and various predisposing factors could contribute to the pathology of AD. Predominant among these possible predisposing factors that have been implicated in AD are age, hypertension, traumatic brain injury, diabetes, chronic neuroinflammation, alteration in calcium levels and oxidative stress. Since both inflammation and altered calcium levels are implicated in the pathogenesis of AD, we wanted to study the effect of altered levels of calcium on inflammation and the subsequent effect of selective calcium channel blockers on the production of pro-inflammatory cytokines and chemokines. Our hypothesis is that Abeta depending on it conformation, may contribute to altered levels of intracellular calcium in neurons and glial cells. We wanted to determine which conformation of Abeta was most pathogenic in terms of increasing inflammation and calcium influx and further elucidate the possibility of a link between altered calcium levels and inflammation. In addition, we wanted to test whether calcium channel blockers could inhibit the inflammation mediated by the most pathogenic form of Abeta by antagonizing the calcium influx triggered by Abeta. Our results in human glial and neuronal cells demonstrate that the high molecular weight oligomers are the most potent at stimulating the release of pro-inflammatory cytokines IL-6 and IL-8 as well as increasing intracellular levels of calcium compared to other conformations of Abeta. Further, L-type calcium channel blockers and calmodulin kinase inhibitors are able to significantly reduce the levels of IL-6 and IL-8. These results suggest that Abeta induced alteration of intracellular calcium levels contributes to its pro-inflammatory effect

    Elevated interleukin-8 enhances prefrontal synaptic transmission in mice with persistent inflammatory pain

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>Interleukin-8 (IL-8) is known for its roles in inflammation and plays critical roles in the development of pain. Its expression increases in the brain after peripheral inflammation. Prefrontal cortex, including the anterior cingulate cortex (ACC), is a forebrain structure known for its roles in pain transmission and modulation. Painful stimuli potentiate the prefrontal synaptic transmission, however, little is known about the expression of IL-8 and its role in the enhanced ACC synaptic transmission in animals with persistent inflammatory pain.</p> <p>Findings</p> <p>In the present study, we examined IL-8 expression in the ACC, somatosensory cortex (SSC), and the dorsal horn of lumbar spinal cord following hind-paw administration of complete Freund's adjuvant (CFA) in mice and its effects on the ACC synaptic transmission. Quantification of IL-8 at protein level (by ELISA) revealed enhanced expression in the ACC and spinal cord during the chronic phases of CFA-induced peripheral inflammation. In vitro whole-cell patch-clamp recordings revealed that IL-8 significantly enhanced synaptic transmission through increased probability of neurotransmitter release in the ACC slice. ACC local infusion of repertaxin, a non-competitive allosteric blocker of IL-8 receptors, notably prolonged the paw withdrawal latency to thermal radian heat stimuli bilaterally in mice.</p> <p>Conclusions</p> <p>Our findings suggest that up-regulation of IL-8 in the ACC partly attributable to the enhanced prefrontal synaptic transmission in the mice with persistent inflammatory pain.</p

    Early hemoperfusion with an immobilized polymyxin B fiber column eliminates humoral mediators and improves pulmonary oxygenation

    Get PDF
    INTRODUCTION: The objective of this study was to clarify the efficacy and mechanism of action of direct hemoperfusion with an immobilized polymyxin B fiber column (DHP-PMX) in patients with acute lung injury or acute respiratory distress syndrome caused by sepsis. METHOD: Thirty-six patients with sepsis were included. In each patient a thermodilution catheter was inserted, and the oxygen delivery index and oxygen consumption index were measured. DHP-PMX was performed in patients with a normal oxygen delivery index and oxygen consumption index (> 500 ml/minute per m(2 )and >120 ml/minute per m(2), respectively). The Acute Physiology and Chronic Health Evaluation II score was used as an index of the severity of sepsis, and survival was assessed after 1 month. The humoral mediators measured were the chemokine IL-8, plasminogen activator inhibitor-1, and neutrophil elastase (NE). These mediators were measured before DHP-PMX treatment, and at 24, 48, and 78 hours after the start of treatment. The arterial oxygen tension (PaO(2))/fractional inspired oxygen (FiO(2)) ratio was measured before DHP-PMX treatment and at 24, 48, 72, 92, and 120 hours after the start of treatment. RESULTS: All patients remained alive after 1 month. Before DHP-PMX treatment, the Acute Physiology and Chronic Health Evaluation II score was 24 ± 2.0, the IL-8 level was 54 ± 15.8 pg/ml, plasminogen activator inhibitor-1 was 133 ± 28.1 ng/ml, and NE was 418 ± 72.1 μg/l. These three humoral mediators began to decrease from 24 hours after DHP-PMX treatment, and the decline became significant from 48 hours onward. The PaO(2)/FiO(2 )ratio was 244 ± 26.3 before DHP-PMX treatment but improved significantly from 96 hours onward. There were significant negative correlations between the PaO(2)/FiO(2 )ratio and blood levels of NE and IL-8. CONCLUSION: The mechanism of action of DHP-PMX is still not fully understood, but we report the following findings. The mean blood levels of plasminogen activator inhibitor-1, NE, and IL-8 were significantly decreased from 48 hours after DHP-PMX treatment. The mean PaO(2)/FiO(2 )ratio was significantly improved from 96 hours after DHP-PMX treatment. Improvement in the PaO(2)/FiO(2 )ratio appeared to be related to the decreases in blood NE and IL-8 levels

    Cytotoxic effect of interleukin-8 in retinal ganglion cells and its possible mechanisms

    Get PDF
    AIM: To investigate the effect of interleukin-8 (IL-8) on neural retinal ganglion cells (RGCs) and whether it can be alleviated by G31P. METHODS: RGC-5 cells were exposed to IL-8 with or without its specific receptor antagonist G31P for 24h, and the cell viability was assessed by Cell Counting Kit 8 (CCK-8). Apoptosis was measured by examining nuclear morphology and quantifying with flow cytometry. Reverse transcription quantitative real-time polymerase chain reaction (RT-qPCR) and Western blot were used to investigate the expression of apoptosis-related genes. RESULTS: CCK-8 assay showed that IL-8 significantly inhibits the viability of RGC-5 cells in a dose-dependent manner. Cell apoptosis assays exhibited higher apoptotic rate in IL-8 treatment group compared to control group. We further found that IL-8 could promote Bax and caspase-3 expressions, but decrease the level of Bcl-2 in the aspect of mRNA and protein. However, pre-treatment with G31P partly attenuated these effects in RGC-5 cells (P<0.05). CONCLUSION: These results indicate that anti-proliferation effects of IL-8 through induction of cell apoptosis regulated by Bcl-2, Bax and caspase-3 expressions, can be ameliorated by G31P

    МЕХАНИЗМЫ ФОРМИРОВАНИЯ ВТОРИЧНОЙ ИММУНОЛОГИЧЕСКОЙ НЕДОСТАТОЧНОСТИ ПРИ ТЯЖЕЛОЙ ЧЕРЕПНО-МОЗГОВОЙ ТРАВМЕ

    Get PDF
    The article presents the review of pathophysiological mechanisms for the formation of the secondary immunological failure in severe craniocerebral injury: a stress-induced change in the functioning the neurohumoral axis, increase in the levels of catecholamines, cytokines, and activation of apoptosis of lymphoid cells.В статье представлен обзор патофизиологических механизмов формирования вторичной иммунологической недостаточности при тяжелой черепно-мозговой травме: стресс-индуцированное изменение функционирования нейрогуморальной оси, увеличение уровней катехоламинов, цитокинов, активация апоптоза лимфоидных клеток

    MECHANISMS FOR THE FORMATION OF THE SECONDARY IMMUNOLOGICAL FAILURE IN SEVERE CRANIOCEREBRAL INJURY

    Get PDF
    The article presents the review of pathophysiological mechanisms for the formation of the secondary immunological failure in severe craniocerebral injury: a stress-induced change in the functioning the neurohumoral axis, increase in the levels of catecholamines, cytokines, and activation of apoptosis of lymphoid cells

    Systemic, local, and imaging biomarkers of brain injury: more needed, and better use of those already established?

    Get PDF
    Much progress has been made over the past two decades in the treatment of severe acute brain injury, including traumatic brain injury and subarachnoid hemorrhage, resulting in a higher proportion of patients surviving with better outcomes. This has arisen from a combination of factors. These include improvements in procedures at the scene (pre-hospital) and in the hospital emergency department, advances in neuromonitoring in the intensive care unit, both continuously at the bedside and intermittently in scans, evolution and refinement of protocol-driven therapy for better management of patients, and advances in surgical procedures and rehabilitation. Nevertheless, many patients still experience varying degrees of long-term disabilities post-injury with consequent demands on carers and resources, and there is room for improvement. Biomarkers are a key aspect of neuromonitoring. A broad definition of a biomarker is any observable feature that can be used to inform on the state of the patient, e.g., a molecular species, a feature on a scan, or a monitoring characteristic, e.g., cerebrovascular pressure reactivity index. Biomarkers are usually quantitative measures, which can be utilized in diagnosis and monitoring of response to treatment. They are thus crucial to the development of therapies and may be utilized as surrogate endpoints in Phase II clinical trials. To date, there is no specific drug treatment for acute brain injury, and many seemingly promising agents emerging from pre-clinical animal models have failed in clinical trials. Large Phase III studies of clinical outcomes are costly, consuming time and resources. It is therefore important that adequate Phase II clinical studies with informative surrogate endpoints are performed employing appropriate biomarkers. In this article, we review some of the available systemic, local, and imaging biomarkers and technologies relevant in acute brain injury patients, and highlight gaps in the current state of knowledge.We gratefully acknowledge financial support as follows. Research support: the Medical Research Council (MRC, Grant Nos. G0600986 ID79068 and G1002277 ID98489) and the National Institute for Health Research Biomedical Research Centre (NIHR BRC) Cambridge (Neuroscience Theme; Brain Injury and Repair Theme). Authors’ support: Keri Linda H. Carpenter – NIHR BRC Cambridge (Neuroscience Theme; Brain Injury and Repair Theme); Ibrahim Jalloh – MRC (Grant no. G1002277 ID 98489) and NIHR BRC Cambridge; Adel Helmy – MRC/Royal College of Surgeons of England Clinical Research Training Fellowship (Grant no. G0802251) and Raymond and Beverly Sackler Fellowship; Virginia F. J. Newcombe–Health Foundation/Academy of Medical Sciences Clinician Scientist Fellowship; Richard J. Shannon–NIHR BRC (Neuroscience Theme; Brain Injury and Repair Theme); Angelos G. Kolias–Royal College of Surgeons of England Research Fellowship, NIHR Academic Clinical Fellowship, and a Raymond and Beverly Sackler Studentship; David Krishna Menon–NIHR Senior Investigator Award; Peter J. Hutchinson – NIHR Research Professorship, Academy of Medical Sciences/Health Foundation Senior Surgical Scientist Fellowship.This is the final published version. It first appeared at http://journal.frontiersin.org/article/10.3389/fneur.2015.00026/full#h13

    Cerebrospinal Fluid and Microdialysis Cytokines in Severe Traumatic Brain Injury: A Scoping Systematic Review.

    Get PDF
    OBJECTIVE: To perform two scoping systematic reviews of the literature on cytokine measurement in: 1. cerebral microdialysis (CMD) and 2. cerebrospinal fluid (CSF) in severe traumatic brain injury (TBI) patients. METHODS: Two separate systematic reviews were conducted: one for CMD cytokines and the second for CSF cytokines. Both were conducted in severe TBI (sTBI) patients only. DATA SOURCES: Articles from MEDLINE, BIOSIS, EMBASE, Global Health, Scopus, Cochrane Library (inception to October 2016), reference lists of relevant articles, and gray literature were searched. STUDY SELECTION: Two reviewers independently identified all manuscripts utilizing predefined inclusion/exclusion criteria. A two-tier filter of references was conducted. DATA EXTRACTION: Patient demographic and study data were extracted to tables. RESULTS: There were 19 studies identified describing the analysis of cytokines via CMD in 267 sTBI patients. Similarly, there were 32 studies identified describing the analysis of CSF cytokines in 1,363 sTBI patients. The two systematic reviews demonstrated: 1. limited literature available on CMD cytokine measurement in sTBI, with some preliminary data supporting feasibility of measurement and associations between cytokines and patient outcome. 2. Various CSF measured cytokines may be associated with patient outcome at 6-12 months, including interleukin (IL)-1b, IL-1ra, IL-6, IL-8, IL-10, and tumor necrosis factor 3. There is little to no literature in support of an association between CSF cytokines and neurophysiologic or tissue outcomes. CONCLUSION: The evaluation of CMD and CSF cytokines is an emerging area of the literature in sTBI. Further, large prospective multicenter studies on cytokines in CMD and CSF need to be conducted.This work was made possible through salary support through: the Cambridge Commonwealth Trust Scholarship, the Royal College of Surgeons of Canada—Harry S. Morton Traveling Fellowship in Surgery, the University of Manitoba Clinician Investigator Program, R. Samuel McLaughlin Research and Education Award, the Manitoba Medical Service Foundation, and the University of Manitoba Faculty of Medicine Dean’s Fellowship Fund. These studies were supported by National Institute for Healthcare Research (NIHR, UK) through the Acute Brain Injury and Repair theme of the Cambridge NIHR Biomedical Research Center, an NIHR Senior Investigator Award to DM, and an NIHR Research Professorship to PH. Authors were also supported by a European Union Framework Program 7 grant (CENTER-TBI; Grant Agreement No. 602150). ET has received funding support from Swedish Society of Medicine (Grant no. SLS-587221). AH is supported by an MRC Studentship for Neuro-inflammation following Human Traumatic Brain injury (G0802251)

    Analyse der Zytokinsynthese-Kapazität von Monozyten polytraumatisierter Patienten

    Get PDF
    Die Destabilisierung des humanen Immunsystems stellt für polytraumatisierte Patienten ein relevantes Problem dar und manifestiert sich auf der Ebene von Organfunktionsstörungen mit nach wie vor erheblicher Letalität. Zahlreiche Untersuchungen der jüngsten Vergangenheit haben klare Hinweise dafür gegeben, daß den zellulären Komponenten des Immunsystems eine zentrale Rolle für die Ausbildung und Ausprägung des posttraumatischen Multi-Organ-Dysfunktions-Syndroms (MODS) und des Multi-Organ-Versagens (Multiple Organ Failure, MOF) zukommt. Dabei hat sich die Fähigkeit monozytärer Zellen auf einen pathologischen Stimulus reagieren zu können, als einer der kritischen Funktionsparameter des menschlichen Immunsystems gezeigt. Die umfangreichen Untersuchungen der jüngsten Vergangenheit konnten jedoch die Frage nach der Dynamik dieser Funktionsstörung bislang nur unzureichend beantworten. Darüber blieb die tatsächliche Synthese-Kapazität relevanter Botenstoffe, wie z.B. von Zytokinen auf intrazellulärem Niveau weitgehend uncharakterisiert. Ziel der vorliegenden Untersuchung war es daher: i) Die intrazelluläre Zytokinsynthese-Kapazität von Monozyten polytraumatisierter Patienten in der direkten posttraumatischen Phase mittels Durchflußzytometrie zu quantifizieren ii) Zu analysieren, ob es einen Zusammenhang zwischen der intrazellulären Aktivierung der Zytokinsynthese-Kapazität und der Änderung der systemischen Zytokin-Konzentration gibt iii) Die dabei gewonnenen Ergebnisse in Vergleich zu klinischen Parametern zu setzen Da es bislang keine validen Versuchsprotokolle für die durchflußzytometrische Analyse der intrazellulären Zytokinsynthese-Kapazität von Monozyten gab, wurden in der ersten Stufe der vorliegenden Studie die Kulturbedingungen für die Stimulation, Sekretionsblockade und Stimulationszeit von Monozyten erarbeitet. Es zeigte sich, daß im Hinblick auf die weitere Fragestellung die Stimulation mit Lipopolysaccharid über 4 Stunden unter einer Sekretionsblockade mit Monensin valide Ergebnisse erbringt. In der zweiten Stufe der Studie wurde erstmalig mittels intrazellulärer single cell Analyse die Synthese-Kapazität von Entzündungs-relevanten Mediatoren (TNF-, Il-1, Il-6 und Il-8) bei polytraumatisierten Patienten untersucht. Gemäß einem seriellen Protokoll wurde zu den Zeitpunkten „Aufnahme in den Schockraum“, 6 Stunden, 12 Stunden, 24 Stunden, 48 Stunden und 72 Stunden nach Trauma bei 13 polytraumatisierten Patienten (ISS >16 Punkte, zwölf überlebt, einer verstorben) jeweils die Zytokinsynthese-Kapazität analysiert. Für TNF- beträgt sie bei Aufnahme 78±5%, für Il-1 ergab sich mit 73±6% ebenso wie für Il-6 mit 58±4%bereits bei Aufnahme eine signifikante Reduktion im Vergleich zur Kontrollgruppe. Es konnte gezeigt werden, daß die Synthese-Kapazität für diese essentiellen proinflammatorischen Zytokine zwischen 12 und 48 Stunden nach Trauma mit 49±5% für TNF-, 53±7% für Il-1, 36,7% für Il-6 und 77,3% für Il-8 signifikant im Vergleich zu den Werten bei Aufnahme reduziert ist. Die vorliegende Untersuchung demonstriert somit eine engmaschige Analyse und Quantifizierung der monozytären Zytokinsynthese-Kapazität für TNF-, Il-6, Il-1 und Il-8 nach Polytrauma. Bezüglich der Analyse, ob ein Zusammenhang zwischen intrazellulärer Aktivierung der Zytokinsynthese-Kapazität und der Änderung der systemischen Zytokin-Konzentration besteht, konnten wir mittels ELISA in der systemischen Zirkulation zwar tendenzielle Veränderungen der einzelnen Faktoren beobachten, jedoch fand sich auf Grund der niedrigen Sensitivität der ELISA-Methode keine signifikante Korrelation zu den hoch-sensitiven intrazellulären Ergebnissen. Bezüglich des Einflusses klinischer Faktoren auf die intrazelluläre Zytokinsynthese-Kapazität ließ sich nachweisen, daß Patienten mit schwerer Verletzung (ISS ≥34) im Zeitraum zwischen 24 Stunden und 72 Stunden nach Trauma eine signifikant niedrigere Zytokinsynthese-Kapazität aufweisen als weniger schwer verletzte Patienten (ISS <34). So beträgt die Kapazität für TNF- in der Schwerer-Verletzten-Gruppe 72 Stunden nach Trauma 62±11% vs. 80±6% in der Leichter-Verletzten-Gruppe. Ähnliche Beobachtungen ließen sich auch für Il-1 und Il-6 machen. Lediglich für die Synthese-Kapazität von Il-8 ließ sich nach 72 Stunden kein Unterschied mehr zwischen den Gruppen feststellen. Die vorliegende Arbeit zeigt somit, daß das, in der Literatur beschriebene Phänomen der posttraumatischen Immundysfunktion bei Monozyten bereits 12 Stunden nach Trauma auftritt, deutlich früher also als bisher in der Literatur anhand von Messungen in Überständen isolierter MØ gezeigt. Des weiteren konnte in Analogie zur Sepsis demonstriert werden, daß die Synthese-Kapazität von Immunzellen offensichtlich mit der Schwere der Erkrankung, i.e. der Schwere der Sepsis bzw. der Schwere der Verletzung korreliert ist. Ziel von Folgestudien muß es nun sein, die den vorgestellten Ergebnissen zugrunde liegenden intrazellulären Steuerungsmechanismen aufzuklären

    Microdiálisis cerebral de alta resolución : Estudio del perfil funcional y estructural de las membranas de 100 kDa

    Get PDF
    La microdiàlisi és una tècnica de neuromonitoratge que permet el mostreig continu del contingut molecular i iònic de l'espai intersticial cerebral. Aquesta tècnica es basa en la implantació d'un catèter en el parènquima cerebral humà de manera mínimament invasiva. Actualment, la microdiàlisi s'ha implantat de manera rutinària en moltes unitats de cures intensives pel neuromonitoratge de pacients amb lesions cerebrals agudes. No obstant, l'estudi in vivo del perfil temporal del proteoma en aquestes lesions i la correcta avaluació de la concentració de les molècules d'interès en el líquid extracel·lular cerebral requereix la determinació prèvia in vitro del percentatge de recuperació relativa de les proteïnes d'estudi.La microdiálisis es una técnica de neuromonitorización que permite el muestreo continuo del contenido molecular e iónico del espacio intersticial cerebral. Esta técnica se basa en la implantación de un catéter en el parénquima cerebral humano de forma mínimamente invasiva. Actualmente, la microdiálisis se ha implantado de forma rutinaria en muchas unidades de cuidados intensivos para la neuromonitorización de pacientes con lesiones cerebrales agudas. Sin embargo, el estudio in vivo el perfil temporal del proteoma en estas lesiones y la evaluación correcta de la concentración de las moléculas de interés en el líquido extracelular cerebral exige la determinación previa in vitro del porcentaje de recuperación relativa de las proteínas de estudio
    corecore