51
|
Sandrof MA, Emerich DF, Thanos CG. Primary Choroid Plexus Tissue for Use in Cellular Therapy. Methods Mol Biol 2017; 1479:237-249. [PMID: 27738941 DOI: 10.1007/978-1-4939-6364-5_19] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The choroid plexus (CP) has been explored as a cellular therapeutic due to its broad-ranging secretome and demonstrated longevity in a variety of encapsulation modalities. While the CP organ is normally involved in disease repair processes in the brain, the range of indications that could potentially be ameliorated with exogenous CP therapy is widespread, including diseases of the central nervous system, hearing loss, chronic wounds, and others. The CP can be isolated from animal sources and digested into a highly purified epithelial culture that can withstand encapsulation and transplantation. Its epithelium can adapt to different microenvironments, and depending on culture conditions, can be manipulated into various three-dimensional configurations with distinct gene expression profiles. The cocktail of proteins secreted by the CP can be harvested in culture, and purified forms of these extracts have been evaluated in topical applications to treat poorly healing wounds. When encapsulated, the epithelial clusters can be maintained for extended durations in vitro with minimal impact on potency. A treatment for Parkinson's disease utilizing encapsulated porcine CP has been developed and is currently being evaluated in a Phase I clinical trial. The current chapter serves to summarize recent experience with CP factor delivery, and provides a description of the relevant materials and methods employed in these studies.
Collapse
Affiliation(s)
- M A Sandrof
- Cytosolv, Inc., 117 Chapman Street, Suite 107, Providence, RI, 02905, USA
| | | | - Chris G Thanos
- Cytosolv, Inc., 117 Chapman Street, Suite 107, Providence, RI, 02905, USA.
| |
Collapse
|
52
|
Calderón-Garcidueñas L, Maronpot RR, Torres-Jardon R, Henríquez-Roldán C, Schoonhoven R, Acuña-Ayala H, Villarreal-Calderón A, Nakamura J, Fernando R, Reed W, Azzarelli B, Swenberg JA. DNA Damage in Nasal and Brain Tissues of Canines Exposed to Air Pollutants Is Associated with Evidence of Chronic Brain Inflammation and Neurodegeneration. Toxicol Pathol 2016; 31:524-38. [PMID: 14692621 DOI: 10.1080/01926230390226645] [Citation(s) in RCA: 244] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Acute, subchronic, or chronic exposures to particulate matter (PM) and pollutant gases affect people in urban areas and those exposed to fires, disasters, and wars. Respiratory tract inflammation, production of mediators of inflammation capable of reaching the brain, systemic circulation of PM, and disruption of the nasal respiratory and olfactory barriers are likely in these populations. DNA damage is crucial in aging and in age-associated diseases such as Alzheimer's disease. We evaluated apurinic/apyrimidinic (AP) sites in nasal and brain genomic DNA, and explored by immunohistochemistry the expression of nuclear factor NF κB p65, inducible nitric oxide synthase (iNOS), cyclo-oxygenase 2 (COX2), metallothionein I and II, apolipoprotein E, amyloid precursor protein (APP), and beta-amyloid1-42 in healthy dogs naturally exposed to urban pollution in Mexico City. Nickel (Ni) and vanadium (V) were measured by inductively coupled plasma mass spectrometry (ICP-MS). Forty mongrel dogs, ages 7 days—10 years were studied (14 controls from Tlaxcala and 26 exposed to urban pollution in South West Metropolitan Mexico City (SWMMC)). Nasal respiratory and olfactory epithelium were found to be early pollutant targets. Olfactory bulb and hippocampal AP sites were significantly higher in exposed than in control age matched animals. Ni and V were present in a gradient from olfactory mucosa > olfactory bulb > frontal cortex. Exposed dogs had (a) nuclear neuronal NF κB p65, (b) endothelial, glial and neuronal iNOS, (c) endothelial and glial COX2, (d) ApoE in neuronal, glial and vascular cells, and (e) APP and β amyloid1-42 in neurons, diffuse plaques (the earliest at age 11 months), and in subarachnoid blood vessels. Increased AP sites and the inflammatory and stress protein brain responses were early and significant in dogs exposed to urban pollution. Oil combustion PM-associated metals Ni and V were detected in the brain. There was an acceleration of Alzheimer's-type pathology in dogs chronically exposed to air pollutants. Respiratory tract inflammation and deteriorating olfactory and respiratory barriers may play a role in the observed neuropathology. These data suggest that Alzheimer's disease may be the sequela of air pollutant exposures and the resulting systemic inflammation.
Collapse
Affiliation(s)
- Lilian Calderón-Garcidueñas
- Environmental Pathology Program, University of North Carolina at Chapel Hill, North Carolina 27599-7310, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
53
|
Selmi C, Barin JG, Rose NR. Current trends in autoimmunity and the nervous system. J Autoimmun 2016; 75:20-29. [DOI: 10.1016/j.jaut.2016.08.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Accepted: 08/06/2016] [Indexed: 01/17/2023]
|
54
|
Le Thuc O, Cansell C, Bourourou M, Denis RG, Stobbe K, Devaux N, Guyon A, Cazareth J, Heurteaux C, Rostène W, Luquet S, Blondeau N, Nahon JL, Rovère C. Central CCL2 signaling onto MCH neurons mediates metabolic and behavioral adaptation to inflammation. EMBO Rep 2016; 17:1738-1752. [PMID: 27733491 DOI: 10.15252/embr.201541499] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Revised: 08/25/2016] [Accepted: 08/30/2016] [Indexed: 12/30/2022] Open
Abstract
Sickness behavior defines the endocrine, autonomic, behavioral, and metabolic responses associated with infection. While inflammatory responses were suggested to be instrumental in the loss of appetite and body weight, the molecular underpinning remains unknown. Here, we show that systemic or central lipopolysaccharide (LPS) injection results in specific hypothalamic changes characterized by a precocious increase in the chemokine ligand 2 (CCL2) followed by an increase in pro-inflammatory cytokines and a decrease in the orexigenic neuropeptide melanin-concentrating hormone (MCH). We therefore hypothesized that CCL2 could be the central relay for the loss in body weight induced by the inflammatory signal LPS. We find that central delivery of CCL2 promotes neuroinflammation and the decrease in MCH and body weight. MCH neurons express CCL2 receptor and respond to CCL2 by decreasing both electrical activity and MCH release. Pharmacological or genetic inhibition of CCL2 signaling opposes the response to LPS at both molecular and physiologic levels. We conclude that CCL2 signaling onto MCH neurons represents a core mechanism that relays peripheral inflammation to sickness behavior.
Collapse
Affiliation(s)
- Ophélia Le Thuc
- Université Côte d'Azur, Nice, France.,CNRS, IPMC, Sophia Antipolis, France
| | - Céline Cansell
- Université Côte d'Azur, Nice, France.,CNRS, IPMC, Sophia Antipolis, France
| | - Miled Bourourou
- Université Côte d'Azur, Nice, France.,CNRS, IPMC, Sophia Antipolis, France
| | - Raphaël Gp Denis
- Univ Paris Diderot Sorbonne Paris Cité Unité de Biologie Fonctionnelle et Adaptative CNRS UMR 8251, Paris, France
| | - Katharina Stobbe
- Université Côte d'Azur, Nice, France.,CNRS, IPMC, Sophia Antipolis, France
| | - Nadège Devaux
- Université Côte d'Azur, Nice, France.,CNRS, IPMC, Sophia Antipolis, France
| | - Alice Guyon
- Université Côte d'Azur, Nice, France.,CNRS, IPMC, Sophia Antipolis, France
| | - Julie Cazareth
- Université Côte d'Azur, Nice, France.,CNRS, IPMC, Sophia Antipolis, France
| | | | - William Rostène
- Institut de la Vision UMRS 968-Université Pierre et Marie Curie, Paris, France
| | - Serge Luquet
- Univ Paris Diderot Sorbonne Paris Cité Unité de Biologie Fonctionnelle et Adaptative CNRS UMR 8251, Paris, France
| | - Nicolas Blondeau
- Université Côte d'Azur, Nice, France.,CNRS, IPMC, Sophia Antipolis, France
| | - Jean-Louis Nahon
- Université Côte d'Azur, Nice, France .,CNRS, IPMC, Sophia Antipolis, France.,Station de Primatologie UPS846 CNRS, Rousset-sur-Arc, France
| | - Carole Rovère
- Université Côte d'Azur, Nice, France .,CNRS, IPMC, Sophia Antipolis, France
| |
Collapse
|
55
|
Hurtado-Alvarado G, Domínguez-Salazar E, Pavon L, Velázquez-Moctezuma J, Gómez-González B. Blood-Brain Barrier Disruption Induced by Chronic Sleep Loss: Low-Grade Inflammation May Be the Link. J Immunol Res 2016; 2016:4576012. [PMID: 27738642 PMCID: PMC5050358 DOI: 10.1155/2016/4576012] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Revised: 08/14/2016] [Accepted: 08/24/2016] [Indexed: 12/16/2022] Open
Abstract
Sleep is a vital phenomenon related to immunomodulation at the central and peripheral level. Sleep deficient in duration and/or quality is a common problem in the modern society and is considered a risk factor to develop neurodegenerative diseases. Sleep loss in rodents induces blood-brain barrier disruption and the underlying mechanism is still unknown. Several reports indicate that sleep loss induces a systemic low-grade inflammation characterized by the release of several molecules, such as cytokines, chemokines, and acute-phase proteins; all of them may promote changes in cellular components of the blood-brain barrier, particularly on brain endothelial cells. In the present review we discuss the role of inflammatory mediators that increase during sleep loss and their association with general disturbances in peripheral endothelium and epithelium and how those inflammatory mediators may alter the blood-brain barrier. Finally, this manuscript proposes a hypothetical mechanism by which sleep loss may induce blood-brain barrier disruption, emphasizing the regulatory effect of inflammatory molecules on tight junction proteins.
Collapse
Affiliation(s)
- G. Hurtado-Alvarado
- Area of Neurosciences, Department of Biology of Reproduction, CBS, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico
| | - E. Domínguez-Salazar
- Area of Neurosciences, Department of Biology of Reproduction, CBS, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico
| | - L. Pavon
- Department of Psychoimmunology, National Institute of Psychiatry “Ramón de la Fuente”, Mexico City, Mexico
| | - J. Velázquez-Moctezuma
- Area of Neurosciences, Department of Biology of Reproduction, CBS, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico
| | - B. Gómez-González
- Area of Neurosciences, Department of Biology of Reproduction, CBS, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico
| |
Collapse
|
56
|
Zhu X, Levasseur PR, Michaelis KA, Burfeind KG, Marks DL. A distinct brain pathway links viral RNA exposure to sickness behavior. Sci Rep 2016; 6:29885. [PMID: 27435819 PMCID: PMC4951726 DOI: 10.1038/srep29885] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 06/23/2016] [Indexed: 11/14/2022] Open
Abstract
Sickness behaviors and metabolic responses to invading pathogens are common to nearly all types of infection. These responses evolved to provide short-term benefit to the host to ward off infection, but impact on quality of life, and when prolonged lead to neurodegeneration, depression, and cachexia. Among the major infectious agents, viruses most frequently enter the brain, resulting in profound neuroinflammation. We sought to define the unique features of the inflammatory response in the brain to these infections. We demonstrate that the molecular pathway defining the central response to dsRNA is distinct from that found in the periphery. The behavioral and physical response to the dsRNA mimetic poly I:C is dependent on signaling via MyD88 when it is delivered centrally, whereas this response is mediated via the TRIF pathway when delivered peripherally. We also define the likely cellular candidates for this MyD88-dependent step. These findings suggest that symptom management is possible without ameliorating protective antiviral immune responses.
Collapse
Affiliation(s)
- Xinxia Zhu
- Papé Family Pediatric Research Institute, Oregon Health &Science University, Portland, OR 97239, USA
| | - Pete R Levasseur
- Papé Family Pediatric Research Institute, Oregon Health &Science University, Portland, OR 97239, USA
| | - Katherine A Michaelis
- Papé Family Pediatric Research Institute, Oregon Health &Science University, Portland, OR 97239, USA.,MD/PhD Program, Oregon Health &Science University, Portland, OR 97239, USA
| | - Kevin G Burfeind
- Papé Family Pediatric Research Institute, Oregon Health &Science University, Portland, OR 97239, USA.,MD/PhD Program, Oregon Health &Science University, Portland, OR 97239, USA
| | - Daniel L Marks
- Papé Family Pediatric Research Institute, Oregon Health &Science University, Portland, OR 97239, USA
| |
Collapse
|
57
|
Calderón-Garcidueñas L, Franco-Lira M, Torres-Jardón R, Henriquez-Roldán C, Barragán-Mejía G, Valencia-Salazar G, González-Maciel A, Reynoso-Robles R, Villarreal-Calderón R, Reed W. Pediatric Respiratory and Systemic Effects of Chronic Air Pollution Exposure: Nose, Lung, Heart, and Brain Pathology. Toxicol Pathol 2016; 35:154-62. [PMID: 17325984 DOI: 10.1080/01926230601059985] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Exposures to particulate matter and gaseous air pollutants have been associated with respiratory tract inflammation, disruption of the nasal respiratory and olfactory barriers, systemic inflammation, production of mediators of inflammation capable of reaching the brain and systemic circulation of particulate matter. Mexico City (MC) residents are exposed to significant amounts of ozone, particulate matter and associated lipopolysaccharides. MC dogs exhibit brain inflammation and an acceleration of Alzheimer’s-like pathology, suggesting that the brain is adversely affected by air pollutants. MC children, adolescents and adults have a significant upregulation of cyclooxygenase-2 (COX2) and interleukin-1β (IL-1β) in olfactory bulb and frontal cortex, as well as neuronal and astrocytic accumulation of the 42 amino acid form of β-amyloid peptide (Aβ42), including diffuse amyloid plaques in frontal cortex. The pathogenesis of Alzheimer’s disease (AD) is characterized by brain inflammation and the accumulation of Aβ42, which precede the appearance of neuritic plaques and neurofibrillary tangles, the pathological hallmarks of AD. Our findings of nasal barrier disruption, systemic inflammation, and the upregulation of COX2 and IL-1β expression and Aβ42 accumulation in brain suggests that sustained exposures to significant concentrations of air pollutants such as particulate matter could be a risk factor for AD and other neurodegenerative diseases.
Collapse
|
58
|
Chaskiel L, Paul F, Gerstberger R, Hübschle T, Konsman JP. Brainstem metabotropic glutamate receptors reduce food intake and activate dorsal pontine and medullar structures after peripheral bacterial lipopolysaccharide administration. Neuropharmacology 2016; 107:146-159. [PMID: 27016016 DOI: 10.1016/j.neuropharm.2016.03.030] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 03/18/2016] [Accepted: 03/21/2016] [Indexed: 12/20/2022]
Abstract
During infection-induced inflammation food intake is reduced. Vagal and brainstem pathways are important both in feeding regulation and immune-to-brain communication. Glutamate is released by vagal afferent terminals in the nucleus of the solitary tract and by its neurons projecting to the parabrachial nuclei. We therefore studied the role of brainstem glutamate receptors in spontaneous food intake of healthy animals and during sickness-associated hypophagia after peripheral administration of bacterial lipopolysaccharides or interleukin-1beta. Brainstem group I and II metabotropic, but not ionotropic, glutamate receptor antagonism increased food intake both in saline- and lipopolysaccharide-treated rats. In these animals, expression of the cellular activation marker c-Fos in the lateral parabrachial nuclei and lipopolysaccharide-induced activation of the nucleus of the solitary tract rostral to the area postrema were suppressed. Group I metabotropic glutamate receptors did not colocalize with c-Fos or neurons regulating gastric function in these structures. Group I metabotropic glutamate receptors were, however, found on raphé magnus neurons that were part of the brainstem circuit innervating the stomach and on trigeminal and hypoglossal motor neurons. In conclusion, our findings show that brainstem metabotropic glutamate receptors reduce food intake and activate the lateral parabrachial nuclei as well as the rostral nucleus of the solitary tract after peripheral bacterial lipopolysaccharide administration. They also provide insight into potential group I metabotropic glutamate receptor-dependent brainstem circuits mediating these effects.
Collapse
Affiliation(s)
- Léa Chaskiel
- CNRS, PsychoNeuroImmunologie, Nutrition et Génétique, UMR 5226, Bordeaux, France; Univ. Bordeaux, PsyNuGen, UMR 5226, Bordeaux, France
| | - Flora Paul
- CNRS, PsychoNeuroImmunologie, Nutrition et Génétique, UMR 5226, Bordeaux, France; Univ. Bordeaux, PsyNuGen, UMR 5226, Bordeaux, France
| | - Rüdiger Gerstberger
- Institut für Veterinär-Physiologie und -Biochemie, Justus-Liebig-Universität Giessen, 35392 Giessen, Germany
| | - Thomas Hübschle
- Institut für Veterinär-Physiologie und -Biochemie, Justus-Liebig-Universität Giessen, 35392 Giessen, Germany
| | - Jan Pieter Konsman
- CNRS, PsychoNeuroImmunologie, Nutrition et Génétique, UMR 5226, Bordeaux, France; Univ. Bordeaux, PsyNuGen, UMR 5226, Bordeaux, France.
| |
Collapse
|
59
|
Burrows F, Haley MJ, Scott E, Coutts G, Lawrence CB, Allan SM, Schiessl I. Systemic inflammation affects reperfusion following transient cerebral ischaemia. Exp Neurol 2016; 277:252-260. [PMID: 26795089 PMCID: PMC4767324 DOI: 10.1016/j.expneurol.2016.01.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 01/13/2016] [Accepted: 01/15/2016] [Indexed: 11/30/2022]
Abstract
Reperfusion after stroke is critical for improved patient survival and recovery and can be achieved clinically through pharmacological (recombinant tissue plasminogen activator) or physical (endovascular intervention) means. Yet these approaches remain confined to a small percentage of stroke patients, often with incomplete reperfusion, and therefore there is an urgent need to learn more about the mechanisms underlying the no-reflow phenomenon that prevents restoration of adequate microvascular perfusion. Recent evidence suggests systemic inflammation as an important contributor to no-reflow and to further investigate this here we inject interleukin 1 (IL-1) i.p. 30 min prior to an ischaemic challenge using a remote filament to occlude the middle cerebral artery (MCA) in mice. Before, during and after the injection of IL-1 and occlusion we use two-dimensional optical imaging spectroscopy to record the spatial and temporal dynamics of oxyhaemoglobin concentration in the cortical areas supplied by the MCA. Our results reveal that systemic inflammation significantly reduces oxyhaemoglobin reperfusion as early as 3h after filament removal compared to vehicle injected animals. CD41 immunohistochemistry shows a significant increase of hyper-coagulated platelets within the microvessels in the stroked cortex of the IL-1 group compared to vehicle. We also observed an increase of pathophysiological biomarkers of ischaemic damage including elevated microglial activation co-localized with interleukin 1α (IL-1α), increased blood brain barrier breakdown as shown by IgG infiltration and increased pyknotic morphological changes of cresyl violet stained neurons. These data confirm systemic inflammation as an underlying cause of no-reflow in the post-ischaemic brain and that appropriate anti-inflammatory approaches could be beneficial in treating ischaemic stroke.
Collapse
Affiliation(s)
- F Burrows
- Faculty of Life Sciences, The University of Manchester, Stopford Building, Oxford Road, M13 9PT Manchester, UK
| | - M J Haley
- Faculty of Life Sciences, The University of Manchester, Stopford Building, Oxford Road, M13 9PT Manchester, UK
| | - E Scott
- Faculty of Life Sciences, The University of Manchester, Stopford Building, Oxford Road, M13 9PT Manchester, UK
| | - G Coutts
- Faculty of Life Sciences, The University of Manchester, Stopford Building, Oxford Road, M13 9PT Manchester, UK
| | - C B Lawrence
- Faculty of Life Sciences, The University of Manchester, Stopford Building, Oxford Road, M13 9PT Manchester, UK
| | - S M Allan
- Faculty of Life Sciences, The University of Manchester, Stopford Building, Oxford Road, M13 9PT Manchester, UK
| | - I Schiessl
- Faculty of Life Sciences, The University of Manchester, Stopford Building, Oxford Road, M13 9PT Manchester, UK.
| |
Collapse
|
60
|
Patejdl R, Penner IK, Noack TK, Zettl UK. Multiple sclerosis and fatigue: A review on the contribution of inflammation and immune-mediated neurodegeneration. Autoimmun Rev 2016; 15:210-20. [DOI: 10.1016/j.autrev.2015.11.005] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 11/13/2015] [Indexed: 02/02/2023]
|
61
|
Carpio C, Villasante C, Galera R, Romero D, de Cos A, Hernanz A, García-Río F. Systemic inflammation and higher perception of dyspnea mimicking asthma in obese subjects. J Allergy Clin Immunol 2016; 137:718-26.e4. [PMID: 26768410 DOI: 10.1016/j.jaci.2015.11.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Revised: 09/29/2015] [Accepted: 11/09/2015] [Indexed: 11/27/2022]
Abstract
BACKGROUND There are a variable number of obese subjects with self-reported diagnosis of asthma but without current or previous evidence of airflow limitation, bronchial reversibility, or airway hyperresponsiveness (misdiagnosed asthma). However, the mechanisms of asthma-like symptoms in obesity remain unclear. OBJECTIVES We sought to evaluate the perception of dyspnea during bronchial challenge and exercise testing in obese patients with asthma and misdiagnosed asthma compared with obese control subjects to identify the mechanisms of asthma-like symptoms in obesity. METHODS In a cross-sectional study we included obese subjects with asthma (n = 25), misdiagnosed asthma (n = 23), and no asthma or respiratory symptoms (n = 27). Spirometry, lung volumes, exhaled nitric oxide levels, and systemic biomarker levels were measured. Dyspnea scores during adenosine bronchial challenge and incremental exercise testing were obtained. RESULTS During bronchial challenge, patients with asthma or misdiagnosed asthma reached a higher Borg-FEV1 slope than control subjects. Moreover, maximum dyspnea and the Borg-oxygen uptake (V'O2) slope were significantly greater during exercise in subjects with asthma or misdiagnosed asthma than in control subjects. The maximum dyspnea achieved during bronchial challenge correlated with IL-1β levels, whereas peak respiratory frequency, ventilatory equivalent for CO2, and IL-6 and IL-1β levels were independent predictors of the Borg-V'O2 slope during exercise (r(2) = 0.853, P < .001). CONCLUSIONS A false diagnosis of asthma (misdiagnosed asthma) in obese subjects is attributable to an increased perception of dyspnea, which, during exercise, is mainly associated with systemic inflammation and excessive ventilation for metabolic demands.
Collapse
Affiliation(s)
- Carlos Carpio
- Servicio de Neumología, Hospital Universitario La Paz, IdiPAZ, Madrid, Spain; CIBER de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Carlos Villasante
- Servicio de Neumología, Hospital Universitario La Paz, IdiPAZ, Madrid, Spain; CIBER de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Raúl Galera
- Servicio de Neumología, Hospital Universitario La Paz, IdiPAZ, Madrid, Spain; CIBER de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - David Romero
- Servicio de Neumología, Hospital Universitario La Paz, IdiPAZ, Madrid, Spain; CIBER de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Ana de Cos
- Unidad de Obesidad, Servicio de Endocrinología y Nutrición, IdiPAZ, Madrid, Spain
| | - Angel Hernanz
- Servicio de Bioquímica, Hospital Universitario La Paz, IdiPAZ, Madrid, Spain
| | - Francisco García-Río
- Servicio de Neumología, Hospital Universitario La Paz, IdiPAZ, Madrid, Spain; CIBER de Enfermedades Respiratorias (CIBERES), Madrid, Spain; Universidad Autónoma de Madrid, Madrid, Spain.
| |
Collapse
|
62
|
Marshall SA, Casachahua JD, Rinker JA, Blose AK, Lysle DT, Thiele TE. IL-1 receptor signaling in the basolateral amygdala modulates binge-like ethanol consumption in male C57BL/6J mice. Brain Behav Immun 2016; 51:258-267. [PMID: 26365025 PMCID: PMC4679505 DOI: 10.1016/j.bbi.2015.09.006] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 08/26/2015] [Accepted: 09/09/2015] [Indexed: 12/26/2022] Open
Abstract
Proinflammatory cytokines have been implicated in alcohol-induced neurodegeneration, but the role of the neuroimmune system in alcohol related behaviors has only recently come to the forefront. Herein, the effects of binge-like drinking on IL-1β mRNA and immunoreactivity within the amygdala were measured following the "drinking in the dark" (DID) paradigm, a model of binge-like ethanol drinking in C57BL/6J mice. Moreover, the role of IL-1 receptor signaling in the amygdala on ethanol consumption was assessed. Results indicated that a history of binge-like ethanol drinking promoted a significant increase of IL-1β mRNA expression within the amygdala, and immunohistochemistry analyses revealed that the basolateral amygdala (BLA), but not central amygdala (CeA), exhibited significantly increased IL-1β immunoreactivity. However, Fluoro-Jade® C labeling indicated that multiple cycles of the DID paradigm were not sufficient to elicit neuronal death. Bilateral infusions of IL-1 receptor antagonist (IL-1Ra) reduced ethanol consumption when infused into the BLA but not the CeA. These observations were specific to ethanol drinking as the IL-1Ra did not alter either sucrose drinking or open-field locomotor activity. The current findings highlight a specific role for IL-1 receptor signaling in modulating binge-like ethanol consumption and indicate that proinflammatory cytokines can be induced prior to dependence or any evidence of neuronal cell death. These findings provide a framework in which to understand how neuroimmune adaptations may alter ethanol consumption and therein contribute to alcohol abuse.
Collapse
Affiliation(s)
- S Alex Marshall
- Department of Psychology & Neuroscience, The University of North Carolina, Chapel Hill, NC 27599, USA; Bowles Center for Alcohol Studies, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - John D Casachahua
- Department of Psychology & Neuroscience, The University of North Carolina, Chapel Hill, NC 27599, USA; Bowles Center for Alcohol Studies, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jennifer A Rinker
- Department of Psychology & Neuroscience, The University of North Carolina, Chapel Hill, NC 27599, USA; Bowles Center for Alcohol Studies, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Allyson K Blose
- Department of Psychology & Neuroscience, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Donald T Lysle
- Department of Psychology & Neuroscience, The University of North Carolina, Chapel Hill, NC 27599, USA; Bowles Center for Alcohol Studies, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Todd E Thiele
- Department of Psychology & Neuroscience, The University of North Carolina, Chapel Hill, NC 27599, USA; Bowles Center for Alcohol Studies, The University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
63
|
Burfeind KG, Michaelis KA, Marks DL. The central role of hypothalamic inflammation in the acute illness response and cachexia. Semin Cell Dev Biol 2015; 54:42-52. [PMID: 26541482 DOI: 10.1016/j.semcdb.2015.10.038] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 10/26/2015] [Indexed: 12/19/2022]
Abstract
When challenged with a variety of inflammatory threats, multiple systems across the body undergo physiological responses to promote defense and survival. The constellation of fever, anorexia, and fatigue is known as the acute illness response, and represents an adaptive behavioral and physiological reaction to stimuli such as infection. On the other end of the spectrum, cachexia is a deadly and clinically challenging syndrome involving anorexia, fatigue, and muscle wasting. Both of these processes are governed by inflammatory mediators including cytokines, chemokines, and immune cells. Though the effects of cachexia can be partially explained by direct effects of disease processes on wasting tissues, a growing body of evidence shows the central nervous system (CNS) also plays an essential mechanistic role in cachexia. In the context of inflammatory stress, the hypothalamus integrates signals from peripheral systems, which it translates into neuroendocrine perturbations, altered neuronal signaling, and global metabolic derangements. Therefore, we will discuss how hypothalamic inflammation is an essential driver of both the acute illness response and cachexia, and why this organ is uniquely equipped to generate and maintain chronic inflammation. First, we will focus on the role of the hypothalamus in acute responses to dietary and infectious stimuli. Next, we will discuss the role of cytokines in driving homeostatic disequilibrium, resulting in muscle wasting, anorexia, and weight loss. Finally, we will address mechanisms and mediators of chronic hypothalamic inflammation, including endothelial cells, chemokines, and peripheral leukocytes.
Collapse
Affiliation(s)
- Kevin G Burfeind
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, OR, USA
- MD/PhD Program, Oregon Health & Science University, Portland, OR, USA
| | - Katherine A Michaelis
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, OR, USA
- MD/PhD Program, Oregon Health & Science University, Portland, OR, USA
| | - Daniel L Marks
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
64
|
Brundin L, Erhardt S, Bryleva EY, Achtyes ED, Postolache TT. The role of inflammation in suicidal behaviour. Acta Psychiatr Scand 2015; 132:192-203. [PMID: 26256862 PMCID: PMC4531386 DOI: 10.1111/acps.12458] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/01/2015] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Over the past decade, clinical data have accumulated showing that inflammation might contribute to the pathophysiology of suicide. To evaluate the associations and to identify the support for pathways linking inflammatory processes with suicidal behaviour, a comprehensive review of the literature was undertaken. METHOD The search terms 'cytokine', 'risk factors', 'kynurenine', 'asthma', 'allergy', 'autoimmunity', 'traumatic brain injury', 'infection' along with the terms 'inflammation' and 'suicide' were entered into PubMed, and a thorough analysis of the publications and their reference lists was performed. RESULTS The effects of inflammation on mood and behaviour could partially be mediated by kynurenine pathway metabolites, modulating neuroinflammation and glutamate neurotransmission. At the same time, the triggers of the inflammatory changes documented in suicidal patients may be attributed to diverse mechanisms such as autoimmunity, neurotropic pathogens, stress or traumatic brain injury. CONCLUSION Targeting the inflammatory system might provide novel therapeutic approaches as well as potential biomarkers to identify patients at increased risk. For the goal of improved detection and treatment of suicidal individuals to be achieved, we need to develop a detailed understanding of the origin, mechanisms and outcomes of inflammation in suicidal behaviour.
Collapse
Affiliation(s)
- L. Brundin
- Division of Psychiatry and Behavioral Medicine College of Human Medicine Michigan State University Grand Rapids MI USA
- Laboratory of Behavioral Medicine Center for Neurodegenerative Science Van Andel Research Institute Grand Rapids MI USA
| | - S. Erhardt
- Department of Physiology & Pharmacology Karolinska Institute Stockholm Sweden
| | - E. Y. Bryleva
- Laboratory of Behavioral Medicine Center for Neurodegenerative Science Van Andel Research Institute Grand Rapids MI USA
| | - E. D. Achtyes
- Division of Psychiatry and Behavioral Medicine College of Human Medicine Michigan State University Grand Rapids MI USA
| | - T. T. Postolache
- Department of Psychiatry University of Maryland School of Medicine Baltimore MD USA
- Veterans Integrated Service Network 19 Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC) Denver CO USA
- Veterans Integrated Service Network 5 MIRECC Baltimore MD USA
| |
Collapse
|
65
|
Interleukin 1 type 1 receptor restore: a genetic mouse model for studying interleukin 1 receptor-mediated effects in specific cell types. J Neurosci 2015; 35:2860-70. [PMID: 25698726 DOI: 10.1523/jneurosci.3199-14.2015] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Interleukin-1 (IL-1) mediates diverse neurophysiological and neuropathological effects in the CNS through type I IL-1 receptor (IL-1R1). However, identification of IL-1R1-expressing cell types and cell-type-specific functions of IL-1R1 remains challenging. In this study, we created a novel genetic mouse model in which IL-1R1 gene expression is disrupted by an intronic insertion of a loxP flanked disruptive sequence that can be deleted by Cre recombinase, resulting in restored IL-1R1 gene expression under its endogenous promoters. A second mutation was introduced at stop codon of the IL-1R1 gene to allow tracking of the restored IL-1R1 protein by a 3HA tag and IL-1R1 mRNA by tdTomato fluorescence. These animals were designated as IL-1R1(r/r) and exhibited an IL-1R1 knock-out phenotype. We used IL-1R1 globally restored mice (IL-1R1(GR/GR)) as an IL-1R1 reporter and observed concordant labeling of IL-1R1 mRNA and protein in brain endothelial cells. Two cell-type-specific IL-1R1 restore lines were generated: Tie2Cre-IL-1R1(r/r) and LysMCre-IL-1R1(r/r). Brain endothelial COX-2 expression, CNS leukocyte infiltration, and global microglia activation induced by intracerebroventricular injection of IL-1β were not observed in IL-1R1(r/r) or LysMCre-IL-1R1(r/r) mice, but were restored in Tie2Cre-IL-1R1(r/r) mice. These results reveal IL-1R1 expression in endothelial cells alone is sufficient to mediate these central IL-1-induced responses. In addition, ex vivo IL-1β stimulation increased IL-1β expression in bone marrow cells in wild-type, Tie2Cre-IL-1R1(r/r), and LysMCre-IL-1R1(r/r), but not IL-1R1(r/r) mice. These results demonstrate this IL-1R1 restore model is a valuable tool for studying cell-type-specific functions of IL-1R1.
Collapse
|
66
|
Bajo M, Varodayan FP, Madamba SG, Robert AJ, Casal LM, Oleata CS, Siggins GR, Roberto M. IL-1 interacts with ethanol effects on GABAergic transmission in the mouse central amygdala. Front Pharmacol 2015; 6:49. [PMID: 25852553 PMCID: PMC4365713 DOI: 10.3389/fphar.2015.00049] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2014] [Accepted: 02/28/2015] [Indexed: 11/13/2022] Open
Abstract
Neuroinflammation is hypothesized to enhance alcohol consumption and contribute to the development of alcoholism. GABAergic transmission in the central amygdala (CeA) plays an important role in the transition to alcohol dependence. Therefore, we studied the effects of interleukin-1β (IL-1β), a proinflammatory cytokine mediating ethanol-induced neuroinflammation, and its interaction with ethanol on CeA GABAegic transmission in B6129SF2/J mice. We also assessed ethanol intake in B6129SF2/J mice. Intake with unlimited (24 h) ethanol access was 9.2–12.7 g/kg (3–15% ethanol), while limited (2 h) access produced an intake of 4.1 ± 0.5 g/kg (15% ethanol). In our electrophysiology experiments, we found that recombinant IL-1β (50 and 100 ng/ml) significantly decreased the amplitude of evoked inhibitory postsynaptic potentials (eIPSPs), with no significant effects on paired-pulse facilitation (PPF). IL-1β (50 ng/ml) had dual effects on spontaneous miniature inhibitory postsynaptic currents (mIPSCs): increasing mIPSC frequencies in most CeA neurons, but decreasing both mIPSC frequencies and amplitudes in a few cells. The IL-1β receptor antagonist (IL-1ra; 100 ng/ml) also had dual effects on mIPSCs and prevented the actions of IL-1β on mIPSC frequencies. These results suggest that IL-1β can alter CeA GABAergic transmission at pre- and postsynaptic sites. Ethanol (44 mM) significantly increased eIPSP amplitudes, decreased PPFs, and increased mIPSC frequencies. IL-1β did not alter ethanol’s enhancement of the eIPSP amplitude, but, in IL-1β-responsive neurons, the ethanol effects on mIPSC frequencies were lost. Overall, our data suggest that the IL-1 system is involved in basal GABAergic transmission and that IL-1β interacts with the ethanol-induced facilitation of CeA GABAergic transmission.
Collapse
Affiliation(s)
- Michal Bajo
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute La Jolla, CA, USA
| | - Florence P Varodayan
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute La Jolla, CA, USA
| | - Samuel G Madamba
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute La Jolla, CA, USA
| | - Amanda J Robert
- Molecular and Cellular Neuroscience Department, The Scripps Research Institute La Jolla, CA, USA
| | - Lindsey M Casal
- Molecular and Cellular Neuroscience Department, The Scripps Research Institute La Jolla, CA, USA
| | - Christopher S Oleata
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute La Jolla, CA, USA
| | - George R Siggins
- Molecular and Cellular Neuroscience Department, The Scripps Research Institute La Jolla, CA, USA
| | - Marisa Roberto
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute La Jolla, CA, USA
| |
Collapse
|
67
|
Abstract
The present review assesses the current state of literature defining integrative autonomic-immune physiological processing, focusing on studies that have employed electrophysiological, pharmacological, molecular biological, and central nervous system experimental approaches. Central autonomic neural networks are informed of peripheral immune status via numerous communicating pathways, including neural and non-neural. Cytokines and other immune factors affect the level of activity and responsivity of discharges in sympathetic and parasympathetic nerves innervating diverse targets. Multiple levels of the neuraxis contribute to cytokine-induced changes in efferent parasympathetic and sympathetic nerve outflows, leading to modulation of peripheral immune responses. The functionality of local sympathoimmune interactions depends on the microenvironment created by diverse signaling mechanisms involving integration between sympathetic nervous system neurotransmitters and neuromodulators; specific adrenergic receptors; and the presence or absence of immune cells, cytokines, and bacteria. Functional mechanisms contributing to the cholinergic anti-inflammatory pathway likely involve novel cholinergic-adrenergic interactions at peripheral sites, including autonomic ganglion and lymphoid targets. Immune cells express adrenergic and nicotinic receptors. Neurotransmitters released by sympathetic and parasympathetic nerve endings bind to their respective receptors located on the surface of immune cells and initiate immune-modulatory responses. Both sympathetic and parasympathetic arms of the autonomic nervous system are instrumental in orchestrating neuroimmune processes, although additional studies are required to understand dynamic and complex adrenergic-cholinergic interactions. Further understanding of regulatory mechanisms linking the sympathetic nervous, parasympathetic nervous, and immune systems is critical for understanding relationships between chronic disease development and immune-associated changes in autonomic nervous system function.
Collapse
Affiliation(s)
- M J Kenney
- Department of Anatomy and Physiology, Kansas State University, Manhattan, Kansas
| | | |
Collapse
|
68
|
Braun TP, Marks DL. The regulation of muscle mass by endogenous glucocorticoids. Front Physiol 2015; 6:12. [PMID: 25691871 PMCID: PMC4315033 DOI: 10.3389/fphys.2015.00012] [Citation(s) in RCA: 172] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 01/08/2015] [Indexed: 01/16/2023] Open
Abstract
Glucocorticoids are highly conserved fundamental regulators of energy homeostasis. In response to stress in the form of perceived danger or acute inflammation, glucocorticoids are released from the adrenal gland, rapidly mobilizing energy from carbohydrate, fat and protein stores. In the case of inflammation, mobilized protein is critical for the rapid synthesis of acute phase reactants and an efficient immune response to infection. While adaptive in response to infection, chronic mobilization can lead to a profound depletion of energy stores. Skeletal muscle represents the major body store of protein, and can become substantially atrophied under conditions of chronic inflammation. Glucocorticoids elicit the atrophy of muscle by increasing the rate of protein degradation by the ubiquitin-proteasome system and autophagy lysosome system. Protein synthesis is also suppressed at the level of translational initiation, preventing the production of new myofibrillar protein. Glucocorticoids also antagonize the action of anabolic regulators such as insulin further exacerbating the loss of protein and muscle mass. The loss of muscle mass in the context of chronic disease is a key feature of cachexia and contributes substantially to morbidity and mortality. A growing body of evidence demonstrates that glucocorticoid signaling is a common mediator of wasting, irrespective of the underlying initiator or disease state. This review will highlight fundamental mechanisms of glucocorticoid signaling and detail the mechanisms of glucocorticoid-induced muscle atrophy. Additionally, the evidence for glucocorticoids as a driver of muscle wasting in numerous disease states will be discussed. Given the burden of wasting diseases and the nodal nature of glucocorticoid signaling, effective anti-glucocorticoid therapy would be a valuable clinical tool. Therefore, the progress and potential pitfalls in the development of glucocorticoid antagonists for muscle wasting will be discussed.
Collapse
Affiliation(s)
- Theodore P Braun
- Department of Internal Medicine, University of Washington Medical Center Seattle, WA, USA ; Papé Family Pediatric Research Institute, Oregon Health and Science University Portland, OR, USA
| | - Daniel L Marks
- Department of Internal Medicine, University of Washington Medical Center Seattle, WA, USA
| |
Collapse
|
69
|
Russi AE, Brown MA. The meninges: new therapeutic targets for multiple sclerosis. Transl Res 2015; 165:255-69. [PMID: 25241937 PMCID: PMC4424790 DOI: 10.1016/j.trsl.2014.08.005] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 08/25/2014] [Accepted: 08/26/2014] [Indexed: 12/19/2022]
Abstract
The central nervous system (CNS) largely comprises nonregenerating cells, including neurons and myelin-producing oligodendrocytes, which are particularly vulnerable to immune cell-mediated damage. To protect the CNS, mechanisms exist that normally restrict the transit of peripheral immune cells into the brain and spinal cord, conferring an "immune-specialized" status. Thus, there has been a long-standing debate as to how these restrictions are overcome in several inflammatory diseases of the CNS, including multiple sclerosis (MS). In this review, we highlight the role of the meninges, tissues that surround and protect the CNS and enclose the cerebral spinal fluid, in promoting chronic inflammation that leads to neuronal damage. Although the meninges have traditionally been considered structures that provide physical protection for the brain and spinal cord, new data have established these tissues as sites of active immunity. It has been hypothesized that the meninges are important players in normal immunosurveillance of the CNS but also serve as initial sites of anti-myelin immune responses. The resulting robust meningeal inflammation elicits loss of localized blood-brain barrier (BBB) integrity and facilitates a large-scale influx of immune cells into the CNS parenchyma. We propose that targeting the cells and molecules mediating these inflammatory responses within the meninges offers promising therapies for MS that are free from the constraints imposed by the BBB. Importantly, such therapies may avoid the systemic immunosuppression often associated with the existing treatments.
Collapse
Affiliation(s)
- Abigail E Russi
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Melissa A Brown
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, Ill.
| |
Collapse
|
70
|
Yang L, Zhao Y, Wang Y, Liu L, Zhang X, Li B, Cui R. The Effects of Psychological Stress on Depression. Curr Neuropharmacol 2015; 13:494-504. [PMID: 26412069 PMCID: PMC4790405 DOI: 10.2174/1570159x1304150831150507] [Citation(s) in RCA: 357] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 01/13/2015] [Accepted: 01/25/2015] [Indexed: 01/01/2023] Open
Abstract
Major depressive disorder is a serious mental disorder that profoundly affects an individual's quality of life. Although the aetiologies underlying this disorder remain unclear, an increasing attention has been focused on the influence imposed by psychological stress over depression. Despite limited animal models of psychological stress, significant progress has been made as to be explicated in this review to elucidate the physiopathology underlying depression and to treat depressive symptoms. Therefore, we will review classical models along with new methods that will enrich our knowledge of this disorder.
Collapse
Affiliation(s)
- Longfei Yang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic
| | - Yinghao Zhao
- Department of Thoracic Surgery, Second Hospital of Jilin University, Changchun 130024, China
| | - Yicun Wang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic
| | - Lei Liu
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic
| | - Xingyi Zhang
- Department of Thoracic Surgery, Second Hospital of Jilin University, Changchun 130024, China
| | - Bingjin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic
| | - Ranji Cui
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic
| |
Collapse
|
71
|
Devorak J, Torres-Platas SG, Davoli MA, Prud'homme J, Turecki G, Mechawar N. Cellular and Molecular Inflammatory Profile of the Choroid Plexus in Depression and Suicide. Front Psychiatry 2015; 6:138. [PMID: 26539126 PMCID: PMC4610134 DOI: 10.3389/fpsyt.2015.00138] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 09/17/2015] [Indexed: 01/15/2023] Open
Abstract
The inflammatory hypothesis of depression is one of the main theories that endeavors to explain and describe the underlying biological mechanisms of depression and suicide. While mounting evidence indicates altered peripheral and central inflammatory profiles in depressed patients and suicide completers, little is known about how peripheral and central inflammation might be linked in these contexts. The choroid plexus (ChP), a highly vascularized tissue that produces cerebrospinal fluid (CSF) and lacks a blood-brain-barrier, is an interface between peripheral and central immune responses. In the present study, we investigated the cellular and molecular inflammatory profile of the ChP of the lateral ventricle in depressed suicides and psychiatrically healthy controls. Gene expression of macrophages, pro- and anti-inflammatory cytokines, and various factors implicated in immune cell trafficking were measured; and density of ionized calcium-binding adaptor molecule 1-positive (Iba1+) macrophages associated with the ChP epithelial cell layer (ECL) was examined. Significant downregulations of the genes encoding interleukin 1ß (IL1ß), a pro-inflammatory acute-phase protein; intercellular cell adhesion molecule 1 (ICAM1), a protein implicated in immune cell trafficking in the ChP; and IBA1, a monocyte/macrophage marker; were detected in depressed suicides as compared to controls. No difference in the density of Iba1+ macrophages associated with the ChP ECL was observed. While interpretation of these findings is challenging in the absence of corroborating data from the CSF, peripheral blood, or brain parenchyma of the present cohort, we hypothesize that the present findings reflect a ChP compensatory mechanism that attenuates the detrimental effects of chronically altered pro-inflammatory signaling caused by elevated levels of pro-inflammatory cytokines, such as IL-1ß, peripherally and/or centrally. Together, these findings further implicate neuroimmune processes in the etiology of depression and suicide.
Collapse
Affiliation(s)
- Julia Devorak
- McGill Group for Suicide Studies, Douglas Mental Health University Institute , Verdun, QC , Canada ; Integrated Program in Neuroscience, McGill University , Montréal, QC , Canada
| | - Susana Gabriela Torres-Platas
- McGill Group for Suicide Studies, Douglas Mental Health University Institute , Verdun, QC , Canada ; Integrated Program in Neuroscience, McGill University , Montréal, QC , Canada
| | - Maria Antonietta Davoli
- McGill Group for Suicide Studies, Douglas Mental Health University Institute , Verdun, QC , Canada
| | - Josée Prud'homme
- McGill Group for Suicide Studies, Douglas Mental Health University Institute , Verdun, QC , Canada
| | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Mental Health University Institute , Verdun, QC , Canada ; Integrated Program in Neuroscience, McGill University , Montréal, QC , Canada ; Department of Psychiatry, McGill University , Montréal, QC , Canada
| | - Naguib Mechawar
- McGill Group for Suicide Studies, Douglas Mental Health University Institute , Verdun, QC , Canada ; Integrated Program in Neuroscience, McGill University , Montréal, QC , Canada ; Department of Psychiatry, McGill University , Montréal, QC , Canada
| |
Collapse
|
72
|
Murta V, Farías MI, Pitossi FJ, Ferrari CC. Chronic systemic IL-1β exacerbates central neuroinflammation independently of the blood-brain barrier integrity. J Neuroimmunol 2014; 278:30-43. [PMID: 25595250 DOI: 10.1016/j.jneuroim.2014.11.023] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2014] [Revised: 11/24/2014] [Accepted: 11/27/2014] [Indexed: 12/13/2022]
Abstract
Peripheral circulating cytokines are involved in immune to brain communication and systemic inflammation is considered a risk factor for flaring up the symptoms in most neurodegenerative diseases. We induced both central inflammatory demyelinating lesion, and systemic inflammation with an interleukin-1β expressing adenovector. The peripheral pro-inflammatory stimulus aggravated the ongoing central lesion independently of the blood-brain barrier (BBB) integrity. This model allows studying the role of specific molecules and cells (neutrophils) from the innate immune system, in the relationship between central and peripheral communication, and on relapsing episodes of demyelinating lesions, along with the role of BBB integrity.
Collapse
Affiliation(s)
- Verónica Murta
- Laboratorio de Terapias Regenerativas y Protectoras del Sistema Nervioso, Leloir Institute Foundation, Institute for Biochemical Investigations, CONICET, Buenos Aires, Argentina.
| | - María Isabel Farías
- Laboratorio de Terapias Regenerativas y Protectoras del Sistema Nervioso, Leloir Institute Foundation, Institute for Biochemical Investigations, CONICET, Buenos Aires, Argentina.
| | - Fernando Juan Pitossi
- Laboratorio de Terapias Regenerativas y Protectoras del Sistema Nervioso, Leloir Institute Foundation, Institute for Biochemical Investigations, CONICET, Buenos Aires, Argentina.
| | - Carina Cintia Ferrari
- Laboratorio de Terapias Regenerativas y Protectoras del Sistema Nervioso, Leloir Institute Foundation, Institute for Biochemical Investigations, CONICET, Buenos Aires, Argentina.
| |
Collapse
|
73
|
Harrison NA, Doeller CF, Voon V, Burgess N, Critchley HD. Peripheral inflammation acutely impairs human spatial memory via actions on medial temporal lobe glucose metabolism. Biol Psychiatry 2014; 76:585-93. [PMID: 24534013 PMCID: PMC4166523 DOI: 10.1016/j.biopsych.2014.01.005] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 01/07/2014] [Accepted: 01/07/2014] [Indexed: 11/05/2022]
Abstract
BACKGROUND Inflammation impairs cognitive performance and is implicated in the progression of neurodegenerative disorders. Rodent studies demonstrated key roles for inflammatory mediators in many processes critical to memory, including long-term potentiation, synaptic plasticity, and neurogenesis. They also demonstrated functional impairment of medial temporal lobe (MTL) structures by systemic inflammation. However, human data to support this position are limited. METHODS Sequential fluorodeoxyglucose positron emission tomography together with experimentally induced inflammation was used to investigate effects of a systemic inflammatory challenge on human MTL function. Fluorodeoxyglucose positron emission tomography scanning was performed in 20 healthy participants before and after typhoid vaccination and saline control injection. After each scanning session, participants performed a virtual reality spatial memory task analogous to the Morris water maze and a mirror-tracing procedural memory control task. RESULTS Fluorodeoxyglucose positron emission tomography data demonstrated an acute reduction in human MTL glucose metabolism after inflammation. The inflammatory challenge also selectively compromised human spatial, but not procedural, memory; this effect that was independent of actions on motivation or psychomotor response. Effects of inflammation on parahippocampal and rhinal glucose metabolism directly mediated actions of inflammation on spatial memory. CONCLUSIONS These data demonstrate acute sensitivity of human MTL to mild peripheral inflammation, giving rise to associated functional impairment in the form of reduced spatial memory performance. Our findings suggest a mechanism for the observed epidemiologic link between inflammation and risk of age-related cognitive decline and progression of neurodegenerative disorders including Alzheimer's disease.
Collapse
Affiliation(s)
- Neil A. Harrison
- Department of Psychiatry, Brighton and Sussex Medical School, Falmer, United Kingdom,Sackler Centre for Consciousness Science, University of Sussex, Falmer, United Kingdom,Address correspondence to Neil A. Harrison, M.B.B.S., Ph.D., Clinical Imaging Sciences Centre, Brighton and Sussex Medical School, University of Sussex, Falmer, BN1 9RR, United Kingdom
| | - Christian F. Doeller
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, The Netherlands
| | - Valerie Voon
- Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Neil Burgess
- Institutes of Cognitive Neuroscience and Neurology, University College London, London, United Kingdom
| | - Hugo D. Critchley
- Department of Psychiatry, Brighton and Sussex Medical School, Falmer, United Kingdom,Sackler Centre for Consciousness Science, University of Sussex, Falmer, United Kingdom
| |
Collapse
|
74
|
Durrant DM, Daniels BP, Klein RS. IL-1R1 signaling regulates CXCL12-mediated T cell localization and fate within the central nervous system during West Nile Virus encephalitis. THE JOURNAL OF IMMUNOLOGY 2014; 193:4095-106. [PMID: 25200953 DOI: 10.4049/jimmunol.1401192] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Immune cell entry into the virally infected CNS is vital for promoting viral clearance yet may contribute to neuropathology if not rigorously regulated. We previously showed that signaling through IL-1R1 is critical for effector T cell reactivation and virologic control within the CNS during murine West Nile virus (WNV) encephalitis. WNV-infected IL-1R1(-/-) mice also display increased parenchymal penetration of CD8(+) T cells despite lack of CD4-mediated full activation, suggesting dysregulation of molecular components of CNS immune privilege. In this study, we show that IL-1 signaling regulates the CNS entry of virus-specific lymphocytes, promoting protective immune responses to CNS viral infections that limit immunopathology. Analysis of blood-brain barrier function in the WNV-infected IL-1R1(-/-) mice revealed no alterations in permeability. However, parenchymal proinflammatory chemokine expression, including CCL2, CCL5, and CXCL10, was significantly upregulated, whereas microvasculature CXCL12 expression was significantly decreased in the absence of IL-1 signaling. We show that during WNV infection, CD11b(+)CD45(hi) infiltrating cells (macrophages) are the primary producers of IL-1β within the CNS and, through the use of an in vitro blood-brain barrier model, that IL-1β promotes CXCR4-mediated T cell adhesion to brain microvasculature endothelial cells. Of interest, IFNγ(+) and CD69(+) WNV-primed T cells were able to overcome CXCL12-mediated adhesion via downregulation of CXCR4. These data indicate that infiltrating IL-1β-producing leukocytes contribute to cellular interactions at endothelial barriers that impart protective CNS inflammation by regulating the parenchymal entry of CXCR4(+) virus-specific T cells during WNV infection.
Collapse
Affiliation(s)
- Douglas M Durrant
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Brian P Daniels
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Robyn S Klein
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110; Department of Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, MO 63110; and Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
75
|
Matsuwaki T, Eskilsson A, Kugelberg U, Jönsson JI, Blomqvist A. Interleukin-1β induced activation of the hypothalamus-pituitary-adrenal axis is dependent on interleukin-1 receptors on non-hematopoietic cells. Brain Behav Immun 2014; 40:166-73. [PMID: 24681250 DOI: 10.1016/j.bbi.2014.03.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 03/18/2014] [Accepted: 03/20/2014] [Indexed: 01/29/2023] Open
Abstract
The proinflammatory cytokine interleukin-1β (IL-1β) plays a major role in the signal transduction of immune stimuli from the periphery to the central nervous system, and has been shown to be an important mediator of the immune-induced stress hormone release. The signaling pathway by which IL-1β exerts this function involves the blood-brain-barrier and induced central prostaglandin synthesis, but the identity of the blood-brain-barrier cells responsible for this signal transduction has been unclear, with both endothelial cells and perivascular macrophages suggested as critical components. Here, using an irradiation and transplantation strategy, we generated mice expressing IL-1 type 1 receptors (IL-1R1) either in hematopoietic or non-hematopoietic cells and subjected these mice to peripheral immune challenge with IL-1β. Following both intraperitoneal and intravenous administration of IL-1β, mice lacking IL-1R1 in hematopoietic cells showed induced expression of the activity marker c-Fos in the paraventricular hypothalamic nucleus, and increased plasma levels of ACTH and corticosterone. In contrast, these responses were not observed in mice with IL-1R1 expression only in hematopoietic cells. Immunoreactivity for IL-1R1 was detected in brain vascular cells that displayed induced expression of the prostaglandin synthesizing enzyme cyclooxygenase-2 and that were immunoreactive for the endothelial cell marker CD31, but was not seen in cells positive for the brain macrophage marker CD206. These results imply that activation of the HPA-axis by IL-1β is dependent on IL-1R1s on non-hematopoietic cells, such as brain endothelial cells, and that IL-1R1 on perivascular macrophages are not involved.
Collapse
Affiliation(s)
- Takashi Matsuwaki
- Division of Cell Biology, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, 581 85 Linköping, Sweden
| | - Anna Eskilsson
- Division of Cell Biology, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, 581 85 Linköping, Sweden
| | - Unn Kugelberg
- Division of Cell Biology, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, 581 85 Linköping, Sweden
| | - Jan-Ingvar Jönsson
- Division of Cell Biology, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, 581 85 Linköping, Sweden
| | - Anders Blomqvist
- Division of Cell Biology, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, 581 85 Linköping, Sweden.
| |
Collapse
|
76
|
Liu Q, Wang T, Yu H, Liu B, Jia R. Interaction between interleukin-1 beta and angiotensin II receptor 1 in hypothalamic paraventricular nucleus contributes to progression of heart failure. J Interferon Cytokine Res 2014; 34:870-5. [PMID: 24955935 DOI: 10.1089/jir.2013.0159] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The central mechanisms by which interleukin-1 beta (IL-1β) and angiotension II receptor 1 (AT1-R) contribute to sympathoexcitation in heart failure (HF) are unclear. In this study, we determined whether an interaction between IL-1β and AT1-R in the paraventricular nucleus (PVN) contributes to progression of HF. Rats were implanted with bilateral PVN cannulae and subjected to coronary artery ligation or sham surgery (Sham). Subsequently, animals were treated for 4 weeks through PVN infusion with either vehicle, losartan (LOS, 200 μg/day), IL-1β (IL, 1 μg/day), or IL-1β along with LOS (LOS+IL). HF rats had higher levels of corticotropin-releasing hormone (CRH), norepinephrine (NE), and glutamate (Glu); lower levels of gamma-aminobutyric acid (GABA); and more positive fra-like activity in PVN when compared with Sham rats. HF rats also had higher levels of NE, epinephrine (EPI), and IL-1β in plasma. PVN infusion of LOS attenuated the decreases in GABA and the increases in CRH, NE, and Glu in the PVN of HF rats. IL-1β could further increase the expression of CRH, NE, Glu, EPI, and IL-1β and decrease GABA expression. Treatment with IL-1β along with LOS could eliminate the effects of IL-1β. These findings suggest that an interaction between AT1-R and IL-1β in the PVN contributes to progression in HF.
Collapse
Affiliation(s)
- Qiang Liu
- Department of Cardiology, Medical School, Tai Shan Medical College, The Fourth People's Hospital of Jinan , Jinan, P.R. China
| | | | | | | | | |
Collapse
|
77
|
Wittmann G, Harney JW, Singru PS, Nouriel SS, Reed Larsen P, Lechan RM. Inflammation-inducible type 2 deiodinase expression in the leptomeninges, choroid plexus, and at brain blood vessels in male rodents. Endocrinology 2014; 155:2009-19. [PMID: 24601886 PMCID: PMC3990842 DOI: 10.1210/en.2013-2154] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 02/23/2014] [Indexed: 11/19/2022]
Abstract
Thyroid hormone regulates immune functions and has antiinflammatory effects. In promoter assays, the thyroid hormone-activating enzyme, type 2 deiodinase (D2), is highly inducible by the inflammatory transcription factor nuclear factor-κ B (NF-κB), but it is unknown whether D2 is induced in a similar fashion in vivo during inflammation. We first reexamined the effect of bacterial lipopolysaccharide (LPS) on D2 expression and NF-κB activation in the rat and mouse brain using in situ hybridization. In rats, LPS induced very robust D2 expression in normally non-D2-expressing cells in the leptomeninges, adjacent brain blood vessels, and the choroid plexus. These cells were vimentin-positive fibroblasts and expressed the NF-κB activation marker, inhibitor κ B-α mRNA, at 2 hours after injection, before the increase in D2 mRNA. In mice, LPS induced intense D2 expression in the choroid plexus but not in leptomeninges, with an early expression peak at 2 hours. Moderate D2 expression along numerous brain blood vessels appeared later. D2 and NF-κB activation was induced in tanycytes in both species but with a different time course. Enzymatic assays from leptomeningeal and choroid plexus samples revealed exceptionally high D2 activity in LPS-treated rats and Syrian hamsters and moderate but significant increases in mice. These data demonstrate the cell type-specific, highly inducible nature of D2 expression by inflammation, and NF-κB as a possible initiating factor, but also warrant attention for species differences. The results suggest that D2-mediated T₃ production by fibroblasts regulate local inflammatory actions in the leptomeninges, choroid plexus and brain blood vessels, and perhaps also in other organs.
Collapse
Affiliation(s)
- Gábor Wittmann
- Department of Medicine (G.W., P.S.S., S.S.N., R.M.L.), Division of Endocrinology, Diabetes and Metabolism, Tupper Research Institute, Tufts Medical Center, and Department of Neuroscience (R.M.L.), Tufts University School of Medicine, Boston, Massachusetts 02111; Thyroid Section (J.W.H., P.R.L.), Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Boston, Massachusetts 02115; and School of Biological Sciences (P.S.S.), National Institute of Science Education and Research, Institute of Physics Campus, PO Sainik School, Bhubaneswar-751005, India
| | | | | | | | | | | |
Collapse
|
78
|
Knockdown of interleukin-1 receptor type-1 on endothelial cells attenuated stress-induced neuroinflammation and prevented anxiety-like behavior. J Neurosci 2014; 34:2583-91. [PMID: 24523548 DOI: 10.1523/jneurosci.3723-13.2014] [Citation(s) in RCA: 160] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Interleukin-1β (IL-1β) is an inflammatory cytokine that plays a prominent role in stress-induced behavioral changes. In a model of repeated social defeat (RSD), elevated IL-1β expression in the brain was associated with recruitment of primed macrophages that were necessary for development of anxiety-like behavior. Moreover, microglia activation and anxiety-like behavior associated with RSD did not occur in IL-1 receptor type-1 knock-out (IL-1R1(KO)) mice. Therefore, the objective of this study was to examine the role of IL-1 signaling in RSD-induced macrophage trafficking to the brain and anxiety-like behavior. Initial studies revealed that RSD did not increase circulating myeloid cells in IL-1R1(KO) mice, resulting in limited macrophage trafficking to the brain. In addition, IL-1R1(KO) bone marrow-chimera mice showed that IL-1R1 expression was essential for macrophage trafficking into the brain. To differentiate cellular mediators of stress-induced IL-1 signaling, endothelial-specific IL-1R1 knock-down (eIL-1R1kd) mice were used. Both wild-type (WT) and eIL-1R1kd mice had increased circulating monocytes, recruitment of macrophages to the brain, and altered microglia activation after RSD. Nonetheless, RSD-induced expression of IL-1β, TNF-α, and IL-6 mRNA in brain CD11b(+) cells was attenuated in eIL-1R1kd mice compared with WT. Moreover, anxiety-like behavior did not develop in eIL-1R1kd mice. Collectively, these findings demonstrated that there was limited RSD-induced priming of myeloid cells in IL-1R1(KO) mice and disrupted propagation of neuroinflammatory signals in the brain of eIL-1R1kd mice. Furthermore, these data showed that transduction of IL-1 signaling by endothelial cells potentiates stress-induced neuroinflammation and promotes anxiety-like behavior.
Collapse
|
79
|
Impaired suppression of feeding by the gut hormone xenin in type I interleukin-1 receptor-deficient mice. Behav Brain Res 2014; 261:60-4. [DOI: 10.1016/j.bbr.2013.12.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 11/10/2013] [Accepted: 12/02/2013] [Indexed: 11/19/2022]
|
80
|
Bellinger DL, Lorton D. Autonomic regulation of cellular immune function. Auton Neurosci 2014; 182:15-41. [PMID: 24685093 DOI: 10.1016/j.autneu.2014.01.006] [Citation(s) in RCA: 156] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 01/17/2014] [Indexed: 12/21/2022]
Abstract
The nervous system and the immune system (IS) are two integrative systems that work together to detect threats and provide host defense, and to maintain/restore homeostasis. Cross-talk between the nervous system and the IS is vital for health and well-being. One of the major neural pathways responsible for regulating host defense against injury and foreign antigens and pathogens is the sympathetic nervous system (SNS). Stimulation of adrenergic receptors (ARs) on immune cells regulates immune cell development, survival, proliferative capacity, circulation, trafficking for immune surveillance and recruitment, and directs the cell surface expression of molecules and cytokine production important for cell-to-cell interactions necessary for a coordinated immune response. Finally, AR stimulation of effector immune cells regulates the activational state of immune cells and modulates their functional capacity. This review focuses on our current understanding of the role of the SNS in regulating host defense and immune homeostasis. SNS regulation of IS functioning is a critical link to the development and exacerbation of chronic immune-mediated diseases. However, there are many mechanisms that need to be further unraveled in order to develop sound treatment strategies that act on neural-immune interaction to resolve or prevent chronic inflammatory diseases, and to improve health and quality of life.
Collapse
Affiliation(s)
- Denise L Bellinger
- Department of Pathology and Human Anatomy, Loma Linda University, School of Medicine, Loma Linda, CA, 92350, USA.
| | - Dianne Lorton
- College of Arts and Sciences, Kent State University and the Kent Summa Initiative for Clinical and Translational Research, Summa Health System, Akron, OH 44304, USA
| |
Collapse
|
81
|
Xu D, Miller SD, Koh S. Immune mechanisms in epileptogenesis. Front Cell Neurosci 2013; 7:195. [PMID: 24265605 PMCID: PMC3821015 DOI: 10.3389/fncel.2013.00195] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 10/08/2013] [Indexed: 01/03/2023] Open
Abstract
Epilepsy is a chronic brain disorder that affects 1% of the human population worldwide. Immune responses are implicated in seizure induction and the development of epilepsy. Pre-clinical and clinical evidence have accumulated to suggest a positive feedback cycle between brain inflammation and epileptogenesis. Prolonged or recurrent seizures and brain injuries lead to upregulation of proinflammatory cytokines and activated immune responses to further increase seizure susceptibility, promote neuronal excitability, and induce blood-brain barrier breakdown. This review focuses on the potential role of innate and adaptive immune responses in the pathogenesis of epilepsy. Both human studies and animal models that help delineate the contributions of brain inflammation in epileptogenesis will be discussed. We highlight the critical role of brain-resident immune mediators and emphasize the contribution of brain-infiltrating peripheral leukocytes. Additionally, we propose possible immune mechanisms that underlie epileptogenesis. Several proinflammatory pathways are discussed, including the interleukin-1 receptor/toll-like receptor signaling cascade, the pathways activated by damage-associated molecular patterns, and the cyclooxygenase-2/prostaglandin pathway. Finally, development of better therapies that target the key constituents and processes identified in these mechanisms are considered, for instance, engineering antagonizing agents that effectively block these pathways in an antigen-specific manner.
Collapse
Affiliation(s)
- Dan Xu
- Department of Microbiology-Immunology and Interdepartmental Immunobiology, Feinberg School of Medicine, Northwestern UniversityChicago IL, USA
- Department of Pediatrics, Division of Neurobiology, Children’s Research Center, Lurie Children’s Hospital of ChicagoChicago IL, USA
| | - Stephen D. Miller
- Department of Microbiology-Immunology and Interdepartmental Immunobiology, Feinberg School of Medicine, Northwestern UniversityChicago IL, USA
| | - Sookyong Koh
- Department of Pediatrics, Division of Neurobiology, Children’s Research Center, Lurie Children’s Hospital of ChicagoChicago IL, USA
- Department of Pediatrics, Feinberg School of Medicine, Northwestern UniversityChicago IL, USA
| |
Collapse
|
82
|
Mediation of glucose-induced anorexia by central nervous system interleukin 1 signaling. Behav Brain Res 2013; 256:512-9. [DOI: 10.1016/j.bbr.2013.08.050] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Revised: 08/23/2013] [Accepted: 08/28/2013] [Indexed: 01/07/2023]
|
83
|
Gonzalez P, Machado I, Vilcaes A, Caruso C, Roth GA, Schiöth H, Lasaga M, Scimonelli T. Molecular mechanisms involved in interleukin 1-beta (IL-1β)-induced memory impairment. Modulation by alpha-melanocyte-stimulating hormone (α-MSH). Brain Behav Immun 2013; 34:141-50. [PMID: 23968970 DOI: 10.1016/j.bbi.2013.08.007] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Revised: 08/05/2013] [Accepted: 08/14/2013] [Indexed: 11/26/2022] Open
Abstract
Pro-inflammatory cytokines can affect cognitive processes such as learning and memory. Particularly, interleukin-1β (IL-1β) influences the consolidation of hippocampus-dependent memories. We previously reported that administration of IL-1β in dorsal hippocampus impaired contextual fear memory consolidation. Different mechanisms have been implicated in the action of IL-1β on long-term potentiation (LTP), but the processes by which this inhibition occurs in vivo remain to be elucidated. We herein report that intrahippocampal injection of IL-1β induced a significant increase in p38 phosphorylation after contextual fear conditioning. Also, treatment with SB203580, an inhibitor of p38, reversed impairment induced by IL-1β on conditioned fear behavior, indicating that this MAPK would be involved in the effect of the cytokine. We also showed that IL-1β administration produced a decrease in glutamate release from dorsal hippocampus synaptosomes and that treatment with SB203580 partially reversed this effect. Our results indicated that IL-1β-induced impairment in memory consolidation could be mediated by a decrease in glutamate release. This hypothesis is sustained by the fact that treatment with d-cycloserine (DCS), a partial agonist of the NMDA receptor, reversed the effect of IL-1β on contextual fear memory. Furthermore, we demonstrated that IL-1β produced a temporal delay in ERK phosphorylation and that DCS administration reversed this effect. We also observed that intrahippocampal injection of IL-1β decreased BDNF expression after contextual fear conditioning. We previously demonstrated that α-MSH reversed the detrimental effect of IL-1β on memory consolidation. The present results demonstrate that α-MSH administration did not modify the decrease in glutamate release induced by IL-1β. However, intrahippocampal injection of α-MSH prevented the effect on ERK phosphorylation and BDNF expression induced by IL-1β after contextual fear conditioning. Therefore, in the present study we determine possible molecular mechanisms involved in the impairment induced by IL-1β on fear memory consolidation. We also established how this effect could be modulated by α-MSH.
Collapse
Affiliation(s)
- P Gonzalez
- IFEC-CONICET, Depto. Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Argentina
| | | | | | | | | | | | | | | |
Collapse
|
84
|
Lackovicova L, Gaykema RP, Banovska L, Kiss A, Goehler LE, Mravec B. The time-course of hindbrain neuronal activity varies according to location during either intraperitoneal or subcutaneous tumor growth in rats: single Fos and dual Fos/dopamine β-hydroxylase immunohistochemistry. J Neuroimmunol 2013; 260:37-46. [PMID: 23673146 DOI: 10.1016/j.jneuroim.2013.04.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Revised: 03/22/2013] [Accepted: 04/10/2013] [Indexed: 01/28/2023]
Abstract
Neuronal activity in the nucleus of the solitary tract, ventrolateral medulla, area postrema, and parabrachial nucleus was studied in rats with intraperitoneal or subcutaneous tumors on the 7th, 14th, 21st, and 28th day after injection of fibrosarcoma tumor cells. We found that the number of Fos and dopamine β-hydroxylase immunopositive neurons differs between animals with intraperitoneal and subcutaneous tumors and also between tumor-bearing rats at different times following injection. Our data indicate that responses of the brainstem structures to peripheral tumor growth depend on the localization as well as the stage of the tumor growth.
Collapse
Affiliation(s)
- Lubica Lackovicova
- Institute of Experimental Endocrinology, Slovak Academy of Sciences, Vlarska 3, 83306 Bratislava, Slovakia
| | | | | | | | | | | |
Collapse
|
85
|
Schéle E, Benrick A, Grahnemo L, Egecioglu E, Anesten F, Pálsdóttir V, Jansson JO. Inter-relation between interleukin (IL)-1, IL-6 and body fat regulating circuits of the hypothalamic arcuate nucleus. J Neuroendocrinol 2013; 25:580-9. [PMID: 23414303 DOI: 10.1111/jne.12033] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Revised: 02/06/2013] [Accepted: 02/11/2013] [Indexed: 12/29/2022]
Abstract
Interleukin (IL)-1 and IL-6 are immune modulating cytokines that also affect metabolic function because both IL-1 receptor I deficient (IL-1RI⁻/⁻) and IL-6 deficient (IL-6⁻/⁻) mice develop late-onset obesity and leptin resistance. Both IL-1 and IL-6 appear to target the central nervous system (CNS) to increase energy expenditure. The hypothalamic arcuate nucleus (ARC) is a major relay between the periphery and CNS in body fat regulation (e.g. by being a target of leptin). The present study aimed to investigate the possible mechanisms responsible for the effects exerted by endogenous IL-1 and IL-6 on body fat at the level of the ARC, as well as possible interactions between IL-1 and IL-6. Therefore, we measured the gene expression of neuropeptides of the ARC involved in energy balance in IL-1RI⁻/⁻ and IL-6⁻/⁻ mice. We also investigated the interactions between expression of IL-1 and IL-6 in these mice, and mapped IL-6 receptor α (IL-6Rα) in the ARC. The expression of the obesity promoting peptide neuropeptide Y (NPY), found in the ARC, was increased in IL-1RI⁻/⁻ mice. The expression of NPY and agouti-related peptide (AgRP), known to be co-expressed with NPY in ARC neurones, was increased in cold exposed IL-6⁻/⁻ mice. IL-6Rα immunoreactivity was densely localised in the ARC, especially in the medial part, and was partly found in NPY positive cell bodies and also α-melanocyte-stimulating hormone positive cell bodies. The expression of hypothalamic IL-6 was decreased in IL-1RI⁻/⁻ mice, whereas IL-1ß expression was increased in IL-6⁻/⁻ mice. The results of the present study indicate that depletion of the activity of the fat suppressing cytokines IL-1 and IL-6 in knockout mice can increase the expression of the obesity promoting neuropeptide NPY in the ARC. Depletion of IL-1 activity suppresses IL-6 expression, and IL-6Rα-like immunoreactivity is present in neurones in the medial ARC, including neurones containing NPY. Therefore, IL-6, IL-1 and NPY/AgRP could interact at the level of the hypothalamic ARC in the regulation of body fat.
Collapse
Affiliation(s)
- E Schéle
- Institute of Neuroscience and Physiology/Endocrinology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | | | | | | | | | | | | |
Collapse
|
86
|
Vezzani A, Friedman A, Dingledine RJ. The role of inflammation in epileptogenesis. Neuropharmacology 2013; 69:16-24. [PMID: 22521336 PMCID: PMC3447120 DOI: 10.1016/j.neuropharm.2012.04.004] [Citation(s) in RCA: 373] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Revised: 03/19/2012] [Accepted: 04/02/2012] [Indexed: 12/20/2022]
Abstract
One compelling challenge in the therapy of epilepsy is to develop anti-epileptogenic drugs with an impact on the disease progression. The search for novel targets has focused recently on brain inflammation since this phenomenon appears to be an integral part of the diseased hyperexcitable brain tissue from which spontaneous and recurrent seizures originate. Although the contribution of specific proinflammatory pathways to the mechanism of ictogenesis in epileptic tissue has been demonstrated in experimental models, the role of these pathways in epileptogenesis is still under evaluation. We review the evidence conceptually supporting the involvement of brain inflammation and the associated blood-brain barrier damage in epileptogenesis, and describe the available pharmacological evidence where post-injury intervention with anti-inflammatory drugs has been attempted. Our review will focus on three main inflammatory pathways, namely the IL-1 receptor/Toll-like receptor signaling, COX-2 and the TGF-β signaling. The mechanisms underlying neuronal-glia network dysfunctions induced by brain inflammation are also discussed, highlighting novel neuromodulatory effects of classical inflammatory mediators such as cytokines and prostaglandins. The increase in knowledge about a role of inflammation in disease progression, may prompt the use of specific anti-inflammatory drugs for developing disease-modifying treatments. This article is part of the Special Issue entitled 'New Targets and Approaches to the Treatment of Epilepsy'.
Collapse
Affiliation(s)
- Annamaria Vezzani
- Laboratory Experimental Neurology, Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Via G. La Masa 19, 20156 Milano, Italy.
| | | | | |
Collapse
|
87
|
McCusker RH, Kelley KW. Immune-neural connections: how the immune system's response to infectious agents influences behavior. ACTA ACUST UNITED AC 2013; 216:84-98. [PMID: 23225871 DOI: 10.1242/jeb.073411] [Citation(s) in RCA: 313] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Humans and animals use the classical five senses of sight, sound, touch, smell and taste to monitor their environment. The very survival of feral animals depends on these sensory perception systems, which is a central theme in scholarly research on comparative aspects of anatomy and physiology. But how do all of us sense and respond to an infection? We cannot see, hear, feel, smell or taste bacterial and viral pathogens, but humans and animals alike are fully aware of symptoms of sickness that are caused by these microbes. Pain, fatigue, altered sleep pattern, anorexia and fever are common symptoms in both sick animals and humans. Many of these physiological changes represent adaptive responses that are considered to promote animal survival, and this constellation of events results in sickness behavior. Infectious agents display a variety of pathogen-associated molecular patterns (PAMPs) that are recognized by pattern recognition receptors (PRRs). These PRR are expressed on both the surface [e.g. Toll-like receptor (TLR)-4] and in the cytoplasm [e.g. nucleotide-binding oligomerization domain (Nod)-like receptors] of cells of the innate immune system, primarily macrophages and dendritic cells. These cells initiate and propagate an inflammatory response by stimulating the synthesis and release of a variety of cytokines. Once an infection has occurred in the periphery, both cytokines and bacterial toxins deliver this information to the brain using both humoral and neuronal routes of communication. For example, binding of PRR can lead to activation of the afferent vagus nerve, which communicates neuronal signals via the lower brain stem (nucleus tractus solitarius) to higher brain centers such as the hypothalamus and amygdala. Blood-borne cytokines initiate a cytokine response from vascular endothelial cells that form the blood-brain barrier (BBB). Cytokines can also reach the brain directly by leakage through the BBB via circumventricular organs or by being synthesized within the brain, thus forming a mirror image of the cytokine milieu in the periphery. Although all cells within the brain are capable of initiating cytokine secretion, microglia have an early response to incoming neuronal and humoral stimuli. Inhibition of proinflammatory cytokines that are induced following bacterial infection blocks the appearance of sickness behaviors. Collectively, these data are consistent with the notion that the immune system communicates with the brain to regulate behavior in a way that is consistent with animal survival.
Collapse
Affiliation(s)
- Robert H McCusker
- Integrative Immunology and Behavior Program, Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801-3873, USA.
| | | |
Collapse
|
88
|
Wei SG, Zhang ZH, Beltz TG, Yu Y, Johnson AK, Felder RB. Subfornical organ mediates sympathetic and hemodynamic responses to blood-borne proinflammatory cytokines. Hypertension 2013; 62:118-25. [PMID: 23670302 DOI: 10.1161/hypertensionaha.113.01404] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Proinflammatory cytokines play an important role in regulating autonomic and cardiovascular function in hypertension and heart failure. Peripherally administered proinflammatory cytokines, such as tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β), act on the brain to increase blood pressure, heart rate, and sympathetic nerve activity. These molecules are too large to penetrate the blood-brain barrier, and so the mechanisms by which they elicit these responses remain unknown. We tested the hypothesis that the subfornical organ (SFO), a forebrain circumventricular organ that lacks a blood-brain barrier, plays a major role in mediating the sympathetic and hemodynamic responses to circulating proinflammatory cytokines. Intracarotid artery injection of TNF-α (200 ng) or IL-1β (200 ng) dramatically increased mean blood pressure, heart rate, and renal sympathetic nerve activity in rats with sham lesions of the SFO (SFO-s). These excitatory responses to intracarotid artery TNF-α and IL-1β were significantly attenuated in SFO-lesioned (SFO-x) rats. Similarly, the increases in mean blood pressure, heart rate, and renal sympathetic nerve activity in response to intravenous injections of TNF-α (500 ng) or IL-1β (500 ng) in SFO-s rats were significantly reduced in the SFO-x rats. Immunofluorescent staining revealed a dense distribution of the p55 TNF-α receptor and the IL-1 receptor accessory protein, a subunit of the IL-1 receptor, in the SFO. These data suggest that SFO is a predominant site in the brain at which circulating proinflammatory cytokines act to elicit cardiovascular and sympathetic responses.
Collapse
Affiliation(s)
- Shun-Guang Wei
- Department of Internal Medicine, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA, USA
| | | | | | | | | | | |
Collapse
|
89
|
Williamson LL, Bilbo SD. Chemokines and the hippocampus: a new perspective on hippocampal plasticity and vulnerability. Brain Behav Immun 2013; 30:186-94. [PMID: 23376170 DOI: 10.1016/j.bbi.2013.01.077] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Revised: 01/07/2013] [Accepted: 01/15/2013] [Indexed: 12/13/2022] Open
Abstract
The hippocampus is critical for several aspects of learning and memory and is unique among other cortical regions in structure, function and the potential for plasticity. This remarkable region recapitulates development throughout the lifespan with enduring neurogenesis and well-characterized plasticity. The structure and traits of the hippocampus that distinguish it from other brain regions, however, may be the same reasons that this important brain region is particularly vulnerable to insult and injury. The immune system within the brain responds to insult and injury, and the hippocampus and the immune system are extensively interconnected. Immune signaling molecules, cytokines and chemokines (chemotactic cytokines), are well known for their functions during insult or injury. They are also increasingly implicated in normal hippocampal neurogenesis (e.g., CXCR4 on newborn neurons), cellular plasticity (e.g., interleukin-6 in LTP maintenance), and learning and memory (e.g., interleukin-1β in fear conditioning). We provide evidence from the small but growing literature that neuroimmune interactions and immune signaling molecules, especially chemokines, may be a primary underlying mechanism for the coexistence of plasticity and vulnerability within the hippocampus. We also highlight the evidence that the hippocampus exhibits a remarkable resilience in response to diverse environmental events (e.g., enrichment, exercise), which all may converge onto common neuroimmune mechanisms.
Collapse
Affiliation(s)
- Lauren L Williamson
- Duke University, Genome Science Research Building 2, 210 Research Dr., Box 91050, Durham, NC 27710, United States.
| | | |
Collapse
|
90
|
Dragunow M. Meningeal and choroid plexus cells--novel drug targets for CNS disorders. Brain Res 2013; 1501:32-55. [PMID: 23328079 DOI: 10.1016/j.brainres.2013.01.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Accepted: 01/07/2013] [Indexed: 12/13/2022]
Abstract
The meninges and choroid plexus perform many functions in the developing and adult human central nervous system (CNS) and are composed of a number of different cell types. In this article I focus on meningeal and choroid plexus cells as targets for the development of drugs to treat a range of traumatic, ischemic and chronic brain disorders. Meningeal cells are involved in cortical development (and their dysfunction may be involved in cortical dysplasia), fibrotic scar formation after traumatic brain injuries (TBI), brain inflammation following infections, and neurodegenerative disorders such as Multiple Sclerosis (MS) and Alzheimer's disease (AD) and other brain disorders. The choroid plexus regulates the composition of the cerebrospinal fluid (CSF) as well as brain entry of inflammatory cells under basal conditions and after injuries. The meninges and choroid plexus also link peripheral inflammation (occurring in the metabolic syndrome and after infections) to CNS inflammation which may contribute to the development and progression of a range of CNS neurological and psychiatric disorders. They respond to cytokines generated systemically and secrete cytokines and chemokines that have powerful effects on the brain. The meninges may also provide a stem cell niche in the adult brain which could be harnessed for brain repair. Targeting meningeal and choroid plexus cells with therapeutic agents may provide novel therapies for a range of human brain disorders.
Collapse
Affiliation(s)
- Mike Dragunow
- Department of Pharmacology and Centre for Brain Research, The University of Auckland, Auckland, New Zealand.
| |
Collapse
|
91
|
Murta V, Pitossi FJ, Ferrari CC. CNS response to a second pro-inflammatory event depends on whether the primary demyelinating lesion is active or resolved. Brain Behav Immun 2012; 26:1102-15. [PMID: 22824737 DOI: 10.1016/j.bbi.2012.07.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Revised: 07/12/2012] [Accepted: 07/12/2012] [Indexed: 11/16/2022] Open
Abstract
Interleukin-1β (IL-1β) is considered to be one of the most important mediators in the pathogenesis of inflammatory diseases, particularly in neurodegenerative diseases such as multiple sclerosis (MS). MS is a chronic inflammatory disease characterized by demyelination and remyelination events, with unpredictable relapsing and remitting episodes that seldom worsen MS lesions. We proposed to study the effect of a unique component of the inflammatory process, IL-1β, and evaluate its effect in repeated episodes, similar to the relapsing-remitting MS pathology. Using adenoviral vectors, we developed a model of focal demyelination/remyelination triggered by the chronic expression of IL-1β. The long-term expression of IL-1β in the striatum produced blood-brain barrier (BBB) breakdown, demyelination, microglial/macrophage activation, and neutrophil infiltration but no overt neuronal degeneration. This demyelinating process was followed by complete remyelination of the area. This simple model allows us to study demyelination and remyelination independently of the autoimmune and adaptive immune components. Re-exposure to this cytokine when the first inflammatory response was still unresolved generated a lesion with decreased neuroinflammation, demyelination, axonal injury and glial response. However, a second long-term expression of IL-1β when the first lesion was resolved could not be differentiated from the first event. In this study, we demonstrated that the response to a second inflammatory stimulus varies depending on whether the initial lesion is still active or has been resolved. Considering that anti-inflammatory treatments have shown little improvement in MS patients, studies about the behavior of specific components of the inflammatory process should be taken into account to develop new therapeutic tools.
Collapse
Affiliation(s)
- Veronica Murta
- Leloir Institute Foundation, Institute for Biochemical Investigations, CONICET, Buenos Aires, Argentina.
| | | | | |
Collapse
|
92
|
Simões AP, Duarte JA, Agasse F, Canas PM, Tomé AR, Agostinho P, Cunha RA. Blockade of adenosine A2A receptors prevents interleukin-1β-induced exacerbation of neuronal toxicity through a p38 mitogen-activated protein kinase pathway. J Neuroinflammation 2012; 9:204. [PMID: 22901528 PMCID: PMC3439355 DOI: 10.1186/1742-2094-9-204] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2011] [Accepted: 07/30/2012] [Indexed: 12/20/2022] Open
Abstract
Background and purpose Blockade of adenosine A2A receptors (A2AR) affords robust neuroprotection in a number of brain conditions, although the mechanisms are still unknown. A likely candidate mechanism for this neuroprotection is the control of neuroinflammation, which contributes to the amplification of neurodegeneration, mainly through the abnormal release of pro-inflammatory cytokines such as interleukin(IL)-1β. We investigated whether A2AR controls the signaling of IL-1β and its deleterious effects in cultured hippocampal neurons. Methods Hippocampal neuronal cultures were treated with IL-1β and/or glutamate in the presence or absence of the selective A2AR antagonist, SCH58261 (50 nmol/l). The effect of SCH58261 on the IL-1β-induced phosphorylation of the mitogen-activated protein kinases (MAPKs) c-Jun N-terminal kinase (JNK) and p38 was evaluated by western blotting and immunocytochemistry. The effect of SCH58261 on glutamate-induced neurodegeneration in the presence or absence of IL-1β was evaluated by nucleic acid and by propidium iodide staining, and by lactate dehydrogenase assay. Finally, the effect of A2AR blockade on glutamate-induced intracellular calcium, in the presence or absence of IL-1β, was studied using single-cell calcium imaging. Results IL-1β (10 to 100 ng/ml) enhanced both JNK and p38 phosphorylation, and these effects were prevented by the IL-1 type 1 receptor antagonist IL-1Ra (5 μg/ml), in accordance with the neuronal localization of IL-1 type 1 receptors, including pre-synaptically and post-synaptically. At 100 ng/ml, IL-1β failed to affect neuronal viability but exacerbated the neurotoxicity induced by treatment with 100 μmol/l glutamate for 25 minutes (evaluated after 24 hours). It is likely that this resulted from the ability of IL-1β to enhance glutamate-induced calcium entry and late calcium deregulation, both of which were unaffected by IL-1β alone. The selective A2AR antagonist, SCH58261 (50 nmol/l), prevented both the IL-1β-induced phosphorylation of JNK and p38, as well as the IL-1β-induced deregulation of calcium and the consequent enhanced neurotoxicity, whereas it had no effect on glutamate actions. Conclusions These results prompt the hypothesis that the neuroprotection afforded by A2AR blockade might result from this particular ability of A2AR to control IL-1β-induced exacerbation of excitotoxic neuronal damage, through the control of MAPK activation and late calcium deregulation.
Collapse
Affiliation(s)
- Ana Patrícia Simões
- Center for Neurosciences of Coimbra, Institute of Biochemistry, Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal.
| | | | | | | | | | | | | |
Collapse
|
93
|
Guo L, Zhu N, Guo Z, Li GK, Chen C, Sang N, Yao QC. Particulate matter (PM10) exposure induces endothelial dysfunction and inflammation in rat brain. JOURNAL OF HAZARDOUS MATERIALS 2012; 213-214:28-37. [PMID: 22365138 DOI: 10.1016/j.jhazmat.2012.01.034] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Revised: 12/05/2011] [Accepted: 01/11/2012] [Indexed: 05/04/2023]
Abstract
Epidemiological studies suggest that particulate matter (PM(10)) inhalation was associated with adverse effects on brain-related health, however, existing experimental data lacked relevant evidences. In this study, we treated Wistar rats with PM(10) at different concentrations (0.3, 1, 3 and 10 mg/kg body weight (bw)), and investigated endothelial dysfunction and inflammatory responses in the brain. The results indicate that mild pathological abnormal occurred after 15-day exposure (five times with 3 days each), followed by the changes of endothelial mediators (ET-1 and eNOS) and inflammatory markers (IL-1β, TNF-α, COX-2, iNOS and ICAM-1). Also, the sample up-regulated bax/bcl-2 ratio and p53 expression, and induced neuronal apoptosis. It implicates that PM(10) exerted injuries to mammals' brain, and the mechanisms might be involved in endothelial dysfunction and inflammatory responses.
Collapse
Affiliation(s)
- Lin Guo
- College of Environment and Resource, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | | | | | | | | | | | | |
Collapse
|
94
|
Interleukin-1 receptor accessory protein organizes neuronal synaptogenesis as a cell adhesion molecule. J Neurosci 2012; 32:2588-600. [PMID: 22357843 DOI: 10.1523/jneurosci.4637-11.2012] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Interleukin-1 receptor accessory protein (IL-1RAcP) is the essential component of receptor complexes mediating immune responses to interleukin-1 family cytokines. IL-1RAcP in the brain exists in two isoforms, IL-1RAcP and IL-1RAcPb, differing only in the C-terminal region. Here, we found robust synaptogenic activities of IL-1RAcP in cultured cortical neurons. Knockdown of IL-1RAcP isoforms in cultured cortical neurons suppressed synapse formation as indicated by decreases of active zone protein Bassoon puncta and dendritic protrusions. IL-1RAcP recovered the accumulation of presynaptic Bassoon puncta, while IL-1RAcPb rescued both Bassoon puncta and dendritic protrusions. Consistently, the expression of IL-1RAcP in cortical neurons enhances the accumulation of Bassoon puncta and that of IL-1RAcPb stimulated both Bassoon puncta accumulation and spinogenesis. IL-1RAcP interacted with protein tyrosine phosphatase (PTP) δ through the extracellular domain. Mini-exon peptides in the Ig-like domains of PTPδ splice variants were critical for their efficient binding to IL-1RAcP. The synaptogenic activities of IL-1RAcP isoforms were diminished in cortical neurons from PTPδ knock-out mice. Correspondingly, PTPδ required IL-1RAcPb to induce postsynaptic differentiation. Thus, IL-1RAcPb bidirectionally regulated synapse formation of cortical neurons. Furthermore, the spine densities of cortical and hippocampal pyramidal neurons were reduced in IL-1RAcP knock-out mice lacking both isoforms. These results suggest that IL-1RAcP isoforms function as trans-synaptic cell adhesion molecules in the brain and organize synapse formation. Thus, IL-1RAcP represents an interesting molecular link between immune systems and synapse formation in the brain.
Collapse
|
95
|
Wei SG, Zhang ZH, Yu Y, Weiss RM, Felder RB. Central actions of the chemokine stromal cell-derived factor 1 contribute to neurohumoral excitation in heart failure rats. Hypertension 2012; 59:991-8. [PMID: 22493069 DOI: 10.1161/hypertensionaha.111.188086] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The ample expression of chemokines and their receptors by neurons in the brain suggests that they play a functional role beyond the coordination of inflammatory and immune responses. Growing evidence implicates brain chemokines in the regulation of neuronal activity and neurohormonal release. This study examined the potential role of brain chemokines in regulating hemodynamic, sympathetic, and neuroendocrine mechanisms in rats with ischemia-induced heart failure (HF). Immunohistochemical analysis revealed that the chemokine stromal cell-derived factor 1 (SDF-1)/CXCL12 was highly expressed in the hypothalamic paraventricular nucleus and subfornical organ and that SDF-1 expression was significantly increased in HF rats compared with sham-operated (SHAM) control rats. ICV injection of SDF-1 induced substantial and long-lasting increases in blood pressure, heart rate, and renal sympathetic nerve activity in both SHAM and HF rats, but responses were exaggerated in HF rats. Bilateral microinjection of SDF-1 into the paraventricular nucleus also elicited exaggerated increases in blood pressure, heart rate, and renal sympathetic nerve activity in the HF rats. A 4-hour ICV infusion of SDF-1 increased plasma levels of arginine vasopressin, adrenocorticotropic hormone, and norepinephrine in normal rats, responses that were prevented by pretreatment with ICV SDF-1 short-hairpin RNA (shRNA). ICV administration of SDF-1 shRNA also reduced plasma arginine vasopressin, adrenocorticotropic hormone, and norepinephrine levels in HF rats. These data suggest that the chemokine SDF-1, acting within the brain, plays an important role in regulating sympathetic drive, neuroendocrine release, and hemodynamic function in normal and pathophysiological conditions and so may contribute to the neural and humoral activation in HF.
Collapse
Affiliation(s)
- Shun-Guang Wei
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | | | | | | | | |
Collapse
|
96
|
Galic MA, Riazi K, Pittman QJ. Cytokines and brain excitability. Front Neuroendocrinol 2012; 33:116-25. [PMID: 22214786 PMCID: PMC3547977 DOI: 10.1016/j.yfrne.2011.12.002] [Citation(s) in RCA: 321] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2011] [Revised: 12/07/2011] [Accepted: 12/15/2011] [Indexed: 01/21/2023]
Abstract
Cytokines are molecules secreted by peripheral immune cells, microglia, astrocytes and neurons in the central nervous system. Peripheral or central inflammation is characterized by an upregulation of cytokines and their receptors in the brain. Emerging evidence indicates that pro-inflammatory cytokines modulate brain excitability. Findings from both the clinical literature and from in vivo and in vitro laboratory studies suggest that cytokines can increase seizure susceptibility and may be involved in epileptogenesis. Cellular mechanisms that underlie these effects include upregulation of excitatory glutamatergic transmission and downregulation of inhibitory GABAergic transmission.
Collapse
Affiliation(s)
- Michael A Galic
- Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, Department of Physiology and Pharmacology, Faculty of Medicine, University of Calgary, Health Sciences Centre, 3330 Hospital Dr. NW, Calgary, Alberta, Canada T2N 4N1
| | | | | |
Collapse
|
97
|
Abstract
The invasion of inflammatory cells occurring after ischemic or traumatic brain injury (TBI) has a detrimental effect on neuronal survival and functional recovery after injury. We have recently demonstrated that not only the blood-brain barrier, but also the blood-cerebrospinal fluid (CSF) barrier (BCSFB), has a role in posttraumatic recruitment of neutrophils. Here, we show that TBI results in a rapid increase in synthesis and release into the CSF of a major chemoattractant for monocytes, CCL2, by the choroid plexus epithelium, a site of the BCSFB. Using an in vitro model of the BCSFB, we also show that CCL2 is released across the apical and basolateral membranes of the choroidal epithelium, a pattern of chemokine secretion that promotes leukocyte migration across epithelial barriers. Immunohistochemical and electron microscopic analyses of choroidal tissue provide evidence for the movement of monocytes, sometimes in tandem with neutrophils, along the paracellular pathways between adjacent epithelial cells. These data further support the pathophysiological role of BCSFB in promoting the recruitment of inflammatory cells to the injured brain.
Collapse
|
98
|
Balan KV, Kc P, Hoxha Z, Mayer CA, Wilson CG, Martin RJ. Vagal afferents modulate cytokine-mediated respiratory control at the neonatal medulla oblongata. Respir Physiol Neurobiol 2011; 178:458-64. [PMID: 21397055 PMCID: PMC3150618 DOI: 10.1016/j.resp.2011.03.003] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2011] [Revised: 02/20/2011] [Accepted: 03/02/2011] [Indexed: 01/17/2023]
Abstract
Perinatal sepsis and inflammation trigger lung and brain injury in preterm infants, and associated apnea of prematurity. We hypothesized that endotoxin exposure in the immature lung would upregulate proinflammatory cytokine mRNA expression in the medulla oblongata and be associated with impaired respiratory control. Lipopolysaccharide (LPS, 0.1mg/kg) or saline was administered intratracheally to rat pups and medulla oblongatas were harvested for quantifying expression of mRNA for proinflammatory cytokines. LPS-exposure significantly increased medullary mRNA for IL-1β and IL-6, and vagotomy blunted this increase in IL-1β, but not IL-6. Whole-body flow plethysmography revealed that LPS-exposed pups had an attenuated ventilatory response to hypoxia both before and after carotid sinus nerve transection. Immunochemical expression of IL-1β within the nucleus of the solitary tract and area postrema was increased after LPS-exposure. In summary, intratracheal endotoxin-exposure in rat pups is associated with upregulation of proinflammatory cytokines in the medulla oblongata that is vagally mediated for IL-1β and associated with an impaired hypoxic ventilatory response.
Collapse
Affiliation(s)
- Kannan V Balan
- Department of Pediatrics, Case Western Reserve University, 11100 Euclid Avenue, Cleveland, OH 44106, USA
| | | | | | | | | | | |
Collapse
|
99
|
Thanos CG, Bintz BE, Goddard M, Boekelheide K, Hall S, Emerich DF. Functional modulation of choroid plexus epithelial clusters in vitro for tissue repair applications. Cell Transplant 2011; 20:1659-72. [PMID: 21396169 DOI: 10.3727/096368911x564985] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
One of the primary obstacles in the restoration or repair of damaged tissues is the temporospatial orchestration of biological and physiological events. Cellular transplantation is an important component of tissue repair as grafted cells can serve as replacement cells or as a source of secreted factors. But few, if any, primary cells can perform more than a single tissue repair function. Epithelial cells, derived from the choroid plexus (CP), are an exception to this rule, as transplanted CP is protective and regenerative in animal models as diverse as CNS degeneration and dermal wound repair. They secrete a myriad of proteins with therapeutic potential as well as matrix and adhesion factors, and contain responsive cytoskeletal components potentially capable of precise manipulation of cellular and extracellular niches. Here we isolated CP from neonatal porcine lateral ventricles and cultured the cells under a variety of conditions to specifically modulate tissue morphology (2D vs. 3D) and protein expression. Using qRT-PCR analysis, transmission electron microscopy, and gene microarray studies we demonstrate a fine level of control over CP epithelial cell clusters opening further opportunities for exploration of the therapeutic potential of this unique tissue source.
Collapse
Affiliation(s)
- C G Thanos
- CytoSolv, Inc., Providence, RI 02905, USA.
| | | | | | | | | | | |
Collapse
|
100
|
Abstract
Stress is a state of physiological or psychological strain caused by adverse stimuli; responses to stress include activation of the sympathetic nervous system, glucocorticoid secretion and emotional behaviors. Prostaglandin E(2) (PGE(2)), acting through its four receptor subtypes (EP1, EP2, EP3 and EP4), is involved in these stress responses. Studies of EP-selective drugs and mice lacking specific EPs have identified the neuronal pathways regulated by PGE(2). In animals with febrile illnesses, PGE(2) acts on neurons expressing EP3 in the preoptic hypothalamus. In illness-induced activation of the hypothalamic-pituitary-adrenal axis, EP1 and EP3 regulate distinct neuronal pathways that converge at the paraventricular hypothalamus. During psychological stress, EP1 suppresses impulsive behaviors via the midbrain dopaminergic systems. PGE(2) promotes illness-induced memory impairment, yet also supports hippocampus-dependent memory formation and synaptic plasticity via EP2 in physiological conditions. In response to illness, PGE(2) is synthesized by enzymes induced in various cell types inside and outside the brain, whereas constitutively expressed enzymes in neurons and/or microglia synthesize PGE(2) in response to psychological stress. Dependent on the type of stress stimuli, PGE(2) released from different cell types activates distinct EP receptors, which mobilize multiple neuronal pathways, resulting in stress responses.
Collapse
Affiliation(s)
- Tomoyuki Furuyashiki
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Yoshida-konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan.
| | | |
Collapse
|