101
|
Ryan KJ, Griffin ÉW, Connor TJ. Complementary anti-inflammatory actions of the β2-adrenoceptor agonist clenbuterol and the glucocorticoid dexamethasone in rat brain. J Neuroimmunol 2011; 232:209-16. [DOI: 10.1016/j.jneuroim.2010.10.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2010] [Revised: 09/14/2010] [Accepted: 10/04/2010] [Indexed: 10/18/2022]
|
102
|
An Y, Chen Q, Quan N. Interleukin-1 exerts distinct actions on different cell types of the brain in vitro. J Inflamm Res 2011; 2011:11-20. [PMID: 22022205 PMCID: PMC3196613 DOI: 10.2147/jir.s15357] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Interleukin-1 (IL-1) is a critical neuroinflammatory mediator in the central nervous system (CNS). In this study, we investigated the effect of IL-1 on inducing inflammation-related gene expression in three astrocyte, two microglial, and one brain endothelial cell line. Interleukin-1 beta (IL-1β) is found to be produced by the two microglial cell lines constitutively, but these cells do not respond to IL-1β stimulation. The three astrocyte cell lines responded to IL-1β stimulation by expressing MCP-1, CXCL-1, and VCAM-1, but different subtypes of astrocytes exhibited different expression profiles after IL-1β stimulation. The brain endothelial cells showed strongest response to IL-1β by producing MCP-1, CXCL-1, VCAM-1, ICAM-1, IL-6, and COX-2 mRNA. The induction of endothelial COX-2 mRNA is shown to be mediated by p38 MAPK pathway, whereas the induction of other genes is mediated by the NF-κB pathway. These results demonstrate that IL-1 exerts distinct cell type-specific action in CNS cells and suggest that IL-1-mediated neuroinflammation is the result of the summation of multiple responses from different cell types in the CNS to IL-1.
Collapse
Affiliation(s)
- Ying An
- Department of Oral Biology, Ohio State University, Columbus, OH, USA
| | | | | |
Collapse
|
103
|
Li Q, Powell N, Zhang H, Belevych N, Ching S, Chen Q, Sheridan J, Whitacre C, Quan N. Endothelial IL-1R1 is a critical mediator of EAE pathogenesis. Brain Behav Immun 2011; 25:160-7. [PMID: 20854891 PMCID: PMC2991628 DOI: 10.1016/j.bbi.2010.09.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2010] [Revised: 09/10/2010] [Accepted: 09/14/2010] [Indexed: 10/19/2022] Open
Abstract
Interleukin-1 (IL-1) has been implicated in the disease progression of multiple sclerosis (MS). In the animal model of MS, experimental autoimmune encephalomyelitis (EAE), the induction of disease is significantly attenuated in mice lacking the type I IL-1 receptor (IL-1R1). In this study, we created a transgenic mouse (eIL-1R1 kd) in which IL-1R1 expression is knocked down specifically in endothelial cells. Induction of EAE in eIL-1R1 kd mice results in a decrease in incidence, severity and delayed onset of EAE. In addition, eIL-1R1 kd mice show significant decrease in VCAM-1 expression and diminished CD45(+) and CD3(+) infiltrating leukocytes in the spinal cord in animals challenged with EAE. Further, IL-1 and IL-23 stimulate IL-17 production by splenocytes from both wild type and the eIL-1R1 kd animals. Similarly, IL-1 and IL-23 synergistically stimulate splenocytes proliferation in these two strains of animals. After immunization with MOG(79-96), although eIL-1R1 kd mice displayed greatly reduced clinical scores, their splenocytes produced IL-17 and proliferated in response to a second MOG challenge, similar to wild type animals. These findings indicate a critical role for endothelial IL-1R1 in mediating the pathogenesis of EAE, and describe a new model that can be used to study endothelial IL-1R1.
Collapse
Affiliation(s)
- Qiming Li
- Institute of Behavior Medicine, 3132 Postle Hall, 305 West 12th Avenue, Ohio State University, Columbus, OH 43210-1094, USA
| | - Nicole Powell
- Institute of Behavior Medicine, 3132 Postle Hall, 305 West 12th Avenue, Ohio State University, Columbus, OH 43210-1094, USA
| | - Hao Zhang
- Institute of Behavior Medicine, 3132 Postle Hall, 305 West 12th Avenue, Ohio State University, Columbus, OH 43210-1094, USA
| | - Natalya Belevych
- Institute of Behavior Medicine, 3132 Postle Hall, 305 West 12th Avenue, Ohio State University, Columbus, OH 43210-1094, USA
| | - San Ching
- Institute of Behavior Medicine, 3132 Postle Hall, 305 West 12th Avenue, Ohio State University, Columbus, OH 43210-1094, USA
| | - Qun Chen
- Institute of Behavior Medicine, 3132 Postle Hall, 305 West 12th Avenue, Ohio State University, Columbus, OH 43210-1094, USA
| | - John Sheridan
- Institute of Behavior Medicine, 3132 Postle Hall, 305 West 12th Avenue, Ohio State University, Columbus, OH 43210-1094, USA
| | - Caroline Whitacre
- Institute of Behavior Medicine, 3132 Postle Hall, 305 West 12th Avenue, Ohio State University, Columbus, OH 43210-1094, USA
| | - Ning Quan
- Institute of Behavior Medicine, 3132 Postle Hall, 305 West 12th Avenue, Ohio State University, Columbus, OH 43210-1094, USA
| |
Collapse
|
104
|
Sinno MH, Coquerel Q, Boukhettala N, Coëffier M, Gallas S, Terashi M, Ibrahim A, Breuillé D, Déchelotte P, Fetissov SO. Chemotherapy-induced anorexia is accompanied by activation of brain pathways signaling dehydration. Physiol Behav 2010; 101:639-48. [DOI: 10.1016/j.physbeh.2010.09.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2010] [Revised: 08/17/2010] [Accepted: 09/22/2010] [Indexed: 02/07/2023]
|
105
|
Neurons and glial cells of the rat organum vasculosum laminae terminalis directly respond to lipopolysaccharide and pyrogenic cytokines. Brain Res 2010; 1363:93-106. [DOI: 10.1016/j.brainres.2010.09.083] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2010] [Revised: 09/16/2010] [Accepted: 09/22/2010] [Indexed: 02/06/2023]
|
106
|
Caruso C, Sanchez M, Durand D, de la Cruz Perez M, Gonzalez PV, Lasaga M, Scimonelli TN. α-Melanocyte-stimulating hormone modulates lipopolysaccharide plus interferon-γ-induced tumor necrosis factor-α expression but not tumor necrosis factor-α receptor expression in cultured hypothalamic neurons. J Neuroimmunol 2010; 227:52-9. [DOI: 10.1016/j.jneuroim.2010.06.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Revised: 06/09/2010] [Accepted: 06/10/2010] [Indexed: 01/09/2023]
|
107
|
Beynon AL, Coogan AN. DIURNAL, AGE, AND IMMUNE REGULATION OF INTERLEUKIN-1β AND INTERLEUKIN-1 TYPE 1 RECEPTOR IN THE MOUSE SUPRACHIASMATIC NUCLEUS. Chronobiol Int 2010; 27:1546-63. [DOI: 10.3109/07420528.2010.501927] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Amy L. Beynon
- Neuroscience and Molecular Psychiatry, Institute of Life Science, School of Medicine, Swansea University, Swansea, United Kingdom
| | - Andrew N. Coogan
- Neuroscience and Molecular Psychiatry, Institute of Life Science, School of Medicine, Swansea University, Swansea, United Kingdom
- Department of Psychology, National University of Ireland Maynooth, Maynooth, Co. Kildare, Republic of Ireland
| |
Collapse
|
108
|
Grossberg AJ, Scarlett JM, Marks DL. Hypothalamic mechanisms in cachexia. Physiol Behav 2010; 100:478-89. [PMID: 20346963 PMCID: PMC2927357 DOI: 10.1016/j.physbeh.2010.03.011] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2010] [Revised: 03/10/2010] [Accepted: 03/12/2010] [Indexed: 12/12/2022]
Abstract
The role of nutrition and balanced metabolism in normal growth, development, and health maintenance is well known. Patients affected with either acute or chronic diseases often show disorders of nutrient balance. In some cases, a devastating state of malnutrition known as cachexia arises, brought about by a synergistic combination of a dramatic decrease in appetite and an increase in metabolism of fat and lean body mass. Other common features that are not required for the diagnosis include decreases in voluntary movement, insulin resistance, and anhedonia. This combination is found in a number of disorders including cancer, cystic fibrosis, AIDS, rheumatoid arthritis, renal failure, and Alzheimer's disease. The severity of cachexia in these illnesses is often the primary determining factor in both quality of life, and in eventual mortality. Indeed, body mass retention in AIDS patients has a stronger association with survival than any other current measure of the disease. This has led to intense investigation of cachexia and the proposal of numerous hypotheses regarding its etiology. Most authors suggest that cytokines released during inflammation and malignancy act on the central nervous system to alter the release and function of a number of neurotransmitters, thereby altering both appetite and metabolic rate. This review will discuss the salient features of cachexia in human diseases, and review the mechanisms whereby inflammation alters the function of key brain regions to produce stereotypical illness behavior. The paper represents an invited review by a symposium, award winner or keynote speaker at the Society for the Study of Ingestive Behavior [SSIB] Annual Meeting in Portland, July 2009.
Collapse
Affiliation(s)
- Aaron J Grossberg
- Department of Pediatrics, Oregon Health & Science University, Portland, OR 97239, USA
| | | | | |
Collapse
|
109
|
McNamee EN, Griffin ÉW, Ryan KM, Ryan KJ, Heffernan S, Harkin A, Connor TJ. Noradrenaline acting at β-adrenoceptors induces expression of IL-1β and its negative regulators IL-1ra and IL-1RII, and drives an overall anti-inflammatory phenotype in rat cortex. Neuropharmacology 2010; 59:37-48. [DOI: 10.1016/j.neuropharm.2010.03.014] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2010] [Revised: 03/23/2010] [Accepted: 03/24/2010] [Indexed: 10/19/2022]
|
110
|
Smith PM, Ferguson AV. Circulating signals as critical regulators of autonomic state--central roles for the subfornical organ. Am J Physiol Regul Integr Comp Physiol 2010; 299:R405-15. [PMID: 20463185 DOI: 10.1152/ajpregu.00103.2010] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
To maintain homeostasis autonomic control centers in the hypothalamus and medulla must respond appropriately to both external and internal stimuli. Although protected behind the blood-brain barrier, neurons in these autonomic control centers are known to be influenced by changing levels of important signaling molecules in the systemic circulation (e.g., osmolarity, glucose concentrations, and regulatory peptides). The subfornical organ belongs to a group of specialized central nervous system structures, the circumventricular organs, which are characterized by the lack of the normal blood-brain barrier, such that circulating lipophobic substances may act on neurons within this region and via well-documented efferent neural projections to hypothalamic autonomic control centers, influence autonomic function. This review focuses on the role of the subfornical organ in sensing peripheral signals and transmitting this information to autonomic control centers in the hypothalamus.
Collapse
Affiliation(s)
- Pauline M Smith
- Dept. of Physiology, Queen's Univ., Kingston, Ontario, Canada K7L 3N6
| | | |
Collapse
|
111
|
Rummel C, Inoue W, Poole S, Luheshi GN. Leptin regulates leukocyte recruitment into the brain following systemic LPS-induced inflammation. Mol Psychiatry 2010; 15:523-34. [PMID: 19773811 DOI: 10.1038/mp.2009.98] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The appetite suppressing hormone leptin has emerged as an important modulator of immune function and is now considered to be a critical link between energy balance and host defense responses to pathogens. These 'adaptive' responses can, in situations of severe and sustained systemic inflammation, lead to adverse effects including brain damage that is partly mediated by neutrophil recruitment into the brain. We examined the contribution of leptin to this process in leptin-deficient (ob/ob), -resistant (db/db) and wild-type (WT) mice injected intraperitoneally with a septic dose of lipopolysaccharide (LPS). This treatment induced a dramatic increase in the number of neutrophils entering the brain of WT mice, an effect that was almost totally abolished in the mutant mice and correlated with a significant reduction in the mRNA levels of interleukin-1beta, intracellular adhesion molecule-1 and neutrophil-specific chemokines. These effects were reversed with leptin replenishment in ob/ob mice leading to recovery of neutrophil recruitment into the brain. Moreover, 48 h food deprivation in WT mice, which decreased circulating leptin levels, attenuated the LPS-induced neutrophil recruitment as did a single injection of an anti-leptin antiserum 4 h before LPS treatment in WT mice. These results provide the first demonstration that leptin has a critical role in leukocyte recruitment to the brain following severe systemic inflammation with possible implications for individuals with altered leptin levels such as during obesity or starvation.
Collapse
Affiliation(s)
- C Rummel
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | | | | | | |
Collapse
|
112
|
Abstract
The innate immune system of the brain is principally composed of microglial cells and astrocytes, which, once activated, protect neurons against insults (infectious agents, lesions, etc.). Activated glial cells produce inflammatory cytokines that act specifically through receptors expressed by the brain. The functional consequences of brain cytokine action (also called neuroinflammation) are alterations in cognition, mood and behaviour, a hallmark of altered well-being. In addition, proinflammatory cytokines play a key role in depression and neurodegenerative diseases linked to aging. Polyunsaturated fatty acids (PUFA) are essential nutrients and essential components of neuronal and glial cell membranes. PUFA from the diet regulate both prostaglandin and proinflammatory cytokine production. n-3 fatty acids are anti-inflammatory while n-6 fatty acids are precursors of prostaglandins. Inappropriate amounts of dietary n-6 and n-3 fatty acids could lead to neuroinflammation because of their abundance in the brain and reduced well-being. Depending on which PUFA are present in the diet, neuroinflammation will, therefore, be kept at a minimum or exacerbated. This could explain the protective role of n-3 fatty acids in neurodegenerative diseases linked to aging.
Collapse
Affiliation(s)
- Sophie Layé
- Psychoneuroimmunology, Nutrition and Genetic (PsyNuGen), UMR INRA 1286, CNRS 5226, University Bordeaux 2, Bordeaux, France.
| |
Collapse
|
113
|
Bémeur C, Desjardins P, Butterworth RF. Antioxidant and anti-inflammatory effects of mild hypothermia in the attenuation of liver injury due to azoxymethane toxicity in the mouse. Metab Brain Dis 2010; 25:23-9. [PMID: 20198438 DOI: 10.1007/s11011-010-9186-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2009] [Accepted: 01/06/2010] [Indexed: 12/19/2022]
Abstract
Previous studies have demonstrated protective effects of mild hypothermia following acetaminophen (APAP)-induced acute liver failure (ALF). However, effects of this treatment in ALF due to other toxins have not yet been fully investigated. In the present study, the effects of mild hypothermia in relation to liver pathology, hepatic and cerebral glutathione, plasma ammonia concentrations, progression of encephalopathy, cerebral edema, and plasma proinflammatory cytokines were assessed in mice with ALF resulting from azoxymethane (AOM) hepatotoxicity, a well characterized model of toxic liver injury. Male C57BL/6 mice were treated with AOM (100 microg/g; i.p.) or saline and sacrificed at coma stages of encephalopathy in parallel with AOM mice maintained mildly hypothermic (35 degrees C). AOM treatment led to hepatic damage, significant increase in plasma transaminase activity, decreased hepatic glutathione levels, and brain GSH/GSSG ratios as well as selective increases in expression of plasma proinflammatory cytokines. Mild hypothermia resulted in reduced hepatic damage, improvement in neurological function, normalization of glutathione levels, and selective attenuation in expression of circulating proinflammatory cytokines. These findings demonstrate that the beneficial effects of mild hypothermia in experimental AOM-induced ALF involve both antioxidant and anti-inflammatory mechanisms.
Collapse
Affiliation(s)
- Chantal Bémeur
- Neuroscience Research Unit, St-Luc Hospital (CHUM), University of Montreal, 1058 St-Denis Street, Montreal, Quebec, Canada, H2X 3J4
| | | | | |
Collapse
|
114
|
Nitric oxide and interleukin-1β mediate noradrenergic induced corticotrophin-releasing hormone release in organotypic cultures of rat paraventricular nucleus. Neuroscience 2010; 165:1191-202. [DOI: 10.1016/j.neuroscience.2009.12.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2009] [Revised: 11/27/2009] [Accepted: 12/01/2009] [Indexed: 11/18/2022]
|
115
|
Serrats J, Schiltz JC, García-Bueno B, van Rooijen N, Reyes TM, Sawchenko PE. Dual roles for perivascular macrophages in immune-to-brain signaling. Neuron 2010; 65:94-106. [PMID: 20152116 PMCID: PMC2873837 DOI: 10.1016/j.neuron.2009.11.032] [Citation(s) in RCA: 207] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/20/2009] [Indexed: 11/25/2022]
Abstract
Cytokines produced during infection/inflammation activate adaptive central nervous system (CNS) responses, including acute stress responses mediated by the hypothalamo-pituitary-adrenal (HPA) axis. The mechanisms by which cytokines engage HPA control circuitry remain unclear, though stimulated release of prostanoids from neighboring vascular cells has been implicated in this regard. How specific vascular cell types, endothelial cells (ECs) versus perivascular cells (PVCs; a subset of brain-resident macrophages), participate in this response remains unsettled. We exploited the phagocytic activity of PVCs to deplete them in rats by central injection of a liposome-encapsulated proapoptotic drug. This manipulation abrogated CNS and hormonal indices of HPA activation under immune challenge conditions (interleukin-1) that activated prostanoid synthesis only in PVCs, while enhancing these responses to stimuli (lipopolysaccharide) that engaged prostanoid production by ECs as well. Thus, PVCs provide both prostanoid-mediated drive to the HPA axis and an anti-inflammatory action that constrains endothelial and overall CNS responses to inflammatory insults.
Collapse
Affiliation(s)
- Jordi Serrats
- Laboratory of Neuronal Structure and Function, The Salk Institute for Biological Studies and the Clayton Medical Research Foundation, La Jolla, CA 92037
| | - Jennifer C. Schiltz
- Laboratory of Neuronal Structure and Function, The Salk Institute for Biological Studies and the Clayton Medical Research Foundation, La Jolla, CA 92037
| | - Borja García-Bueno
- Laboratory of Neuronal Structure and Function, The Salk Institute for Biological Studies and the Clayton Medical Research Foundation, La Jolla, CA 92037
| | - Nico van Rooijen
- Dept. Molecular Cell Biology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands
| | - Teresa M. Reyes
- Laboratory of Neuronal Structure and Function, The Salk Institute for Biological Studies and the Clayton Medical Research Foundation, La Jolla, CA 92037
| | - Paul E. Sawchenko
- Laboratory of Neuronal Structure and Function, The Salk Institute for Biological Studies and the Clayton Medical Research Foundation, La Jolla, CA 92037
| |
Collapse
|
116
|
Gautron L, Layé S. Neurobiology of inflammation-associated anorexia. Front Neurosci 2010; 3:59. [PMID: 20582290 PMCID: PMC2858622 DOI: 10.3389/neuro.23.003.2009] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2009] [Accepted: 12/16/2009] [Indexed: 12/23/2022] Open
Abstract
Compelling data demonstrate that inflammation-associated anorexia directly results from the action of pro-inflammatory factors, primarily cytokines and prostaglandins E2, on the nervous system. For instance, the aforementioned pro-inflammatory factors can stimulate the activity of peripheral sensory neurons, and induce their own de novo synthesis and release into the brain parenchyma and cerebrospinal fluid. Ultimately, it results in the mobilization of a specific neural circuit that shuts down appetite. The present article describes the different cell groups and neurotransmitters involved in inflammation-associated anorexia and examines how they interact with neural systems regulating feeding such as the melanocortin system. A better understanding of the neurobiological mechanisms underlying inflammation-associated anorexia will help to develop appetite stimulants for cancer and AIDS patients.
Collapse
Affiliation(s)
- Laurent Gautron
- The University of Texas Southwestern Medical Center Dallas, TX, USA
| | | |
Collapse
|
117
|
Abstract
The growing spark of interest in research concerning the molecular links between the nervous, endocrine and immune systems has caused an explosion of new knowledge concerning the fine mechanisms that orchestrate the integrated response to an immune challenge. For instance, elevation in plasma glucocorticoid (GC) levels is one of the most powerful and well-controlled feedback mechanisms on the proinflammatory signal transduction machinery taking place across the organism. Circulating inflammatory molecules have the ability to target their cognate receptors at the levels of blood-brain barrier, the latter in return produces specific prostaglandins (PGs). This chapter presents the brain circuits involved in the activation of the hypothalamic-pituitary-adrenal (HPA) axis by endogenously produced prostaglandin E(2) (PGE(2)) during systemic innate immune insults.
Collapse
|
118
|
Hollis JH, Lemus M, Evetts MJ, Oldfield BJ. Central interleukin-10 attenuates lipopolysaccharide-induced changes in food intake, energy expenditure and hypothalamic Fos expression. Neuropharmacology 2009; 58:730-8. [PMID: 20045008 DOI: 10.1016/j.neuropharm.2009.12.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2009] [Revised: 12/13/2009] [Accepted: 12/18/2009] [Indexed: 10/20/2022]
Abstract
Lipopolysaccharide (LPS) is often used to mimic acute infection and induces hypophagia, the selective partitioning of fat for energy, and fever. Interleukin-10 (IL-10) is an anti-inflammatory cytokine expressed in the brain which attenuates LPS-induced hypophagia; however the potential sites of interaction within the brain have not been investigated. Hypothalamic orexin (ORX) and melanin-concentrating hormone (MCH) regulate energy expenditure and food intake although the regulation of these neuropeptides through the interactions between central IL-10 and the inflammatory consequences of peripheral LPS have not been investigated. The present study in the rat investigated during the dark phase of the light-dark cycle the ability of central IL-10 (250 ng, i.c.v.) to attenuate the changes in food intake, energy substrate partitioning, and central Fos expression within the hypothalamus to peripheral LPS (100 microg/kg, i.p.); Fos expression changes specifically within ORX and MCH neurons were also investigated. Central IL-10 attenuated the peripheral LPS-induced hypophagia, reduction in motor activity, fever and reduction in respiratory exchange ratio. Central IL-10 also attenuated peripheral LPS-induced increases in Fos expression within ORX neurons and decreases in Fos expression within unidentified cells of the caudal arcuate nucleus. In contrast, both IL-10 and LPS injection independently decreased Fos expression within MCH neurons. The present study provides further insight into the interactions within the brain between the anti-inflammatory cytokine IL-10, the inflammatory consequences of LPS, and neuropeptides known to regulate energy expenditure and food intake.
Collapse
Affiliation(s)
- Jacob H Hollis
- Department of Physiology, Monash University, Victoria, Australia.
| | | | | | | |
Collapse
|
119
|
McNamee EN, Ryan KM, Kilroy D, Connor TJ. Noradrenaline induces IL-1ra and IL-1 type II receptor expression in primary glial cells and protects against IL-1beta-induced neurotoxicity. Eur J Pharmacol 2009; 626:219-28. [PMID: 19818755 DOI: 10.1016/j.ejphar.2009.09.054] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2009] [Revised: 09/18/2009] [Accepted: 09/28/2009] [Indexed: 10/20/2022]
Abstract
The pro-inflammatory cytokine interleukin-1beta (IL-1beta) plays a key role in initiating an immune response within the central nervous system (CNS), and is thought to be a significant contributor to the neurodegenerative process. The actions of IL-1beta can be regulated by interleukin-1 receptor antagonist (IL-1ra), which prevents IL-1beta from acting on the IL-1 type I receptor (IL-1RI). Another negative regulator of the IL-1 system is the IL-1 type II receptor (IL-1RII); a decoy receptor that serves to sequester IL-1. Consequently, pharmacological strategies that tip the balance in favour of IL-1ra and IL-1RII may be of therapeutic benefit. Evidence suggests that the neurotransmitter noradrenaline elicits anti-inflammatory actions in the CNS, and consequently may play an endogenous neuroprotective role. Here we report that noradrenaline induces production of IL-1ra and IL-1RII from primary rat mixed glial cells. In contrast, noradrenaline did not alter IL-1beta expression, or expression of IL-1RI or the IL-1 type I receptor accessory protein (IL-1RAcp); both of which are required for IL-1 signalling. Our results demonstrate that the ability of noradrenaline to induce IL-1ra and IL-1RII is mediated via beta-adrenoceptor activation and downstream activation of protein kinase A and extracellular signal-regulated kinase (ERK). In parallel with its ability to increase IL-1ra and IL-1RII, noradrenaline prevented neurotoxicity in cortical primary neurons induced by conditioned medium from IL-1beta treated mixed glial cells. These data indicate that noradrenaline negatively regulates IL-1 system in glial cells and has neuroprotective properties in situations where IL-1 contributes to pathology.
Collapse
Affiliation(s)
- Eoin N McNamee
- Neuroimmunology Research Group, Department of Physiology, School of Medicine & Trinity College Institute of Neuroscience, Trinity College, Dublin 2, Ireland
| | | | | | | |
Collapse
|
120
|
Serrats J, Sawchenko PE. How T-cell-dependent and -independent challenges access the brain: vascular and neural responses to bacterial lipopolysaccharide and staphylococcal enterotoxin B. Brain Behav Immun 2009; 23:1038-52. [PMID: 19524662 PMCID: PMC2751606 DOI: 10.1016/j.bbi.2009.06.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2009] [Revised: 05/30/2009] [Accepted: 06/08/2009] [Indexed: 12/12/2022] Open
Abstract
Bacterial lipopolysaccharide (LPS) is widely used to study immune influences on the CNS, and cerebrovascular prostaglandin (PG) synthesis is implicated in mediating LPS influences on some acute phase responses. Other bacterial products, such as staphylococcal enterotoxin B (SEB), impact target tissues differently in that their effects are T-lymphocyte-dependent, yet both LPS and SEB recruit a partially overlapping set of subcortical central autonomic cell groups. We sought to compare neurovascular responses to the two pathogens, and the mechanisms by which they may access the brain. Rats received iv injections of LPS (2 microg/kg), SEB (1mg/kg) or vehicle and were sacrificed 0.5-3h later. Both challenges engaged vascular cells as early 0.5h, as evidenced by induced expression of the vascular early response gene (Verge), and the immediate-early gene, NGFI-B. Cyclooxygenase-2 (COX-2) expression was detected in both endothelial and perivascular cells (PVCs) in response to LPS, but only in PVCs of SEB-challenged animals. The non-selective COX inhibitor, indomethacin (1mg/kg, iv), blocked LPS-induced activation in a subset of central autonomic structures, but failed to alter SEB-driven responses. Liposome mediated ablation of PVCs modulated the CNS response to LPS, did not affect the SEB-induced activational profile. By contrast, disruptions of interoceptive signaling by area postrema lesions or vagotomy (complete or hepatic) markedly attenuated SEB-, but not LPS-, stimulated central activational responses. Despite partial overlap in their neuronal and vascular response profiles, LPS and SEB appear to use distinct mechanisms to access the brain.
Collapse
Affiliation(s)
- Jordi Serrats
- Laboratory of Neuronal Structure and Function, The Salk Institute for Biological Studies and The Clayton Medical Research Foundation, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | |
Collapse
|
121
|
Summy-Long JY, Hu S. Peripheral osmotic stimulation inhibits the brain's innate immune response to microdialysis of acidic perfusion fluid adjacent to supraoptic nucleus. Am J Physiol Regul Integr Comp Physiol 2009; 297:R1532-45. [PMID: 19759333 DOI: 10.1152/ajpregu.00340.2009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
During the brain's innate immune response microglia, astroglia and ependymal cells resolve/repair damaged tissue and control infection. Released interleukin-1beta (IL-1beta) reaching cerebroventricles stimulates circumventricular organs (CVOs; subfornical organ, SFO; organum vasculosum lamina terminalis, OVLT), the median preoptic nucleus (MePO), and magnocellular and parvocellular neurons in the supraoptic (SON) and paraventricular (PVN) nuclei. Hypertonic saline (HS) also activates these osmosensory CVOs and neuroendocrine systems, but, in contrast to IL-1beta, inhibits the peripheral immune response. To examine whether the brain's innate immune response is attenuated by osmotic stimulation, sterile acidic perfusion fluid was microdialyzed (2 microl/min) in the SON area of conscious rats for 6 h with sterile HS (1.5 M NaCl) injected subcutaneously (15 ml/kg) at 5 h. Immunohistochemistry identified cytokine sources (IL-1beta(+); OX-42(+) microglia) and targets (IL-1R(+); inducible cyclooxygenase, COX-2(+); c-Fos(+)) near the probe, in CVOs, MePO, ependymal cells, periventricular hypothalamus, SON, and PVN. Inserting the probe stimulated magnocellular neurons (c-Fos(+); SON; PVN) via the MePO (c-Fos(+)), a response enhanced by HS. Microdialysis activated microglia (OX-42(+); amoeboid/hypertrophied; IL-1beta(+)) in the adjacent SON and bilaterally in perivascular areas of the PVN, periventricular hypothalamus and ependyma, coincident with c-Fos expression in ependymal cells and COX-2 in the vasculature. These microglial responses were attenuated by HS, coincident with activating parvocellular and magnocellular neuroendocrine systems and elevating circulating IL-1beta, oxytocin, and vasopressin. Acidosis-induced cellular injury from microdialysis activated the brain's innate immune response by a mechanism inhibited by peripheral osmotic stimulation.
Collapse
Affiliation(s)
- Joan Y Summy-Long
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, Pennsylvania 17033, USA
| | | |
Collapse
|
122
|
Harrison NA, Brydon L, Walker C, Gray MA, Steptoe A, Dolan RJ, Critchley HD. Neural origins of human sickness in interoceptive responses to inflammation. Biol Psychiatry 2009; 66:415-22. [PMID: 19409533 PMCID: PMC2885492 DOI: 10.1016/j.biopsych.2009.03.007] [Citation(s) in RCA: 254] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2008] [Revised: 01/26/2009] [Accepted: 02/23/2009] [Indexed: 11/25/2022]
Abstract
BACKGROUND Inflammation is associated with psychological, emotional, and behavioral disturbance, known as sickness behavior. Inflammatory cytokines are implicated in coordinating this central motivational reorientation accompanying peripheral immunologic responses to pathogens. Studies in rodents suggest an afferent interoceptive neural mechanism, although comparable data in humans are lacking. METHODS In a double-blind, randomized crossover study, 16 healthy male volunteers received typhoid vaccination or saline (placebo) injection in two experimental sessions. Profile of Mood State questionnaires were completed at baseline and at 2 and 3 hours. Two hours after injection, participants performed a high-demand color word Stroop task during functional magnetic resonance imaging. Blood samples were performed at baseline and immediately after scanning. RESULTS Typhoid but not placebo injection produced a robust inflammatory response indexed by increased circulating interleukin-6 accompanied by a significant increase in fatigue, confusion, and impaired concentration at 3 hours. Performance of the Stroop task under inflammation activated brain regions encoding representations of internal bodily state. Spatial and temporal characteristics of this response are consistent with interoceptive information flow via afferent autonomic fibers. During performance of this task, activity within interoceptive brain regions also predicted individual differences in inflammation-associated but not placebo-associated fatigue and confusion. Maintenance of cognitive performance, despite inflammation-associated fatigue, led to recruitment of additional prefrontal cortical regions. CONCLUSIONS These findings suggest that peripheral infection selectively influences central nervous system function to generate core symptoms of sickness and reorient basic motivational states.
Collapse
Affiliation(s)
- Neil A. Harrison
- Wellcome Trust, Centre for Neuroimaging, Institute of Neurology, University College London, London, United Kingdom,Institute of Cognitive Neuroscience, University College London, London, United Kingdom,Address correspondence to Neil Harrison, M.B.B.S., Ph.D., UCL Institute of Cognitive Neuroscience, Alexandra House, University College London, 17 Queen Square, London WC1N 3AR, UK
| | - Lena Brydon
- Department of Epidemiology and Public Health, University College London, London, United Kingdom
| | - Cicely Walker
- Department of Epidemiology and Public Health, University College London, London, United Kingdom
| | - Marcus A. Gray
- Brighton and Sussex Medical School, University of Sussex Campus, Falmer, Brighton, United Kingdom
| | - Andrew Steptoe
- Department of Epidemiology and Public Health, University College London, London, United Kingdom
| | - Raymond J. Dolan
- Wellcome Trust, Centre for Neuroimaging, Institute of Neurology, University College London, London, United Kingdom
| | - Hugo D. Critchley
- Brighton and Sussex Medical School, University of Sussex Campus, Falmer, Brighton, United Kingdom
| |
Collapse
|
123
|
Szmydynger-Chodobska J, Strazielle N, Zink BJ, Ghersi-Egea JF, Chodobski A. The role of the choroid plexus in neutrophil invasion after traumatic brain injury. J Cereb Blood Flow Metab 2009; 29:1503-16. [PMID: 19471279 PMCID: PMC2736364 DOI: 10.1038/jcbfm.2009.71] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Traumatic brain injury (TBI) frequently results in neuroinflammation, which includes the invasion of neutrophils. After TBI, neutrophils infiltrate the choroid plexus (CP), a site of the blood-cerebrospinal fluid (CSF) barrier (BCSFB), and accumulate in the CSF space near the injury, from where these inflammatory cells may migrate to brain parenchyma. We have hypothesized that the CP functions as an entry point for neutrophils to invade the injured brain. Using the controlled cortical impact model of TBI in rats and an in vitro model of the BCSFB, we show that the CP produces CXC chemokines, such as cytokine-induced neutrophil chemoattractant (CINC)-1 or CXCL1, CINC-2alpha or CXCL3, and CINC-3 or CXCL2. These chemokines are secreted both apically and basolaterally from the choroidal epithelium, a prerequisite for neutrophil migration across epithelial barriers. Consistent with these findings, we also provide electron microscopic evidence that neutrophils infiltrate the choroidal stroma and subsequently reach the intercellular space between choroidal epithelial cells. This is the first detailed analysis of the BCSFB function related to neutrophil trafficking. Our observations support the role of this barrier in posttraumatic neutrophil invasion.
Collapse
Affiliation(s)
- Joanna Szmydynger-Chodobska
- Department of Emergency Medicine, The Warren Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA.
| | | | | | | | | |
Collapse
|
124
|
Nilsberth C, Hamzic N, Norell M, Blomqvist A. Peripheral lipopolysaccharide administration induces cytokine mRNA expression in the viscera and brain of fever-refractory mice lacking microsomal prostaglandin E synthase-1. J Neuroendocrinol 2009; 21:715-21. [PMID: 19500218 DOI: 10.1111/j.1365-2826.2009.01888.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
We examined the expression of interleukin (IL)-1beta, IL-6 and tumour necrosis factor (TNF) alpha in mice lacking microsomal prostaglandin E synthase-1 (mPGES-1), which neither produce prostaglandin E(2), nor mount a febrile response upon immune challenge. Intraperitoneal lipopolysaccharide (LPS) injection resulted in a strongly induced expression of all three cytokines in the brain and viscera, similar to wild-type animals. Several brain regions additionally showed modest induction of receptors for these cytokines in both genotypes. Telemetric recordings of body temperature showed that the mPGES-1 deficient mice remained afebrile upon LPS challenge, in contrast to the prominent fever displayed by the wild-type mice. These data demonstrate that LPS-induced cytokine expression occurs independently of prostaglandin E(2), and imply that endogenously expressed IL-1beta, IL-6, and TNFalpha are not pyrogenic per se, supporting the role of prostaglandin E(2) as the final and obligatory mediator of LPS-induced fever.
Collapse
Affiliation(s)
- C Nilsberth
- Linköping University, Faculty of Health Sciences, Department of Clinical and Experimental Medicine, Division of Cell Biology, Linköping, Sweden.
| | | | | | | |
Collapse
|
125
|
Abstract
After defining hyperthermia and fever, this article describes the complete chain of events leading to the genesis of fever, starting with the lipopolysaccharide-induced formation of endogenous pyrogens (cytokines), their interactions with relevant targets in the brain, the induction of enzymes responsible for the formation of prostaglandin E2, the activation of descending neuronal pathways via the EP3 receptor, and the stimulation of thermogenesis via this pathway to support the febrile shift of the thermoregulatory set point. This article also summarizes an alternative hypothesis to account for a rapid induction of the early phase of lipopolysaccharide-induced fever before the release of larger amounts of cytokines into the bloodstream. Other topics discussed include malignant hypothermia, drug-induced hypothermia, and the heat stroke syndrome.
Collapse
|
126
|
Abstract
The psychologic and behavioral components of sickness represent, together with fever response and associated neuroendocrine changes, a highly organized strategy of the organism to fight infection. This strategy, referred to as sickness behavior, is triggered by the proinflammatory cytokines produced by activated cells of the innate immune system in contact with specific pathogen-associated molecular patterns (PAMPs). Interleukin-1 and other cytokines act on the brain via (1) a neural route represented by the primary afferent neurons that innervate the body site where the infectious process takes place and (2) a humoral pathway that involves the production of proinflammatory cytokines. This article presents the current knowledge on the way this communication system is organized and regulated and the implications of these advances for understanding brain physiology and pathology.
Collapse
Affiliation(s)
- Robert Dantzer
- Integrative Immunology and Behavior Program, University of Illinois at Urbana-Champaign, 212 ERML, 1201 W Gregory Drive, Urbana, IL 61801, USA.
| |
Collapse
|
127
|
Koo JW, Duman RS. Interleukin-1 receptor null mutant mice show decreased anxiety-like behavior and enhanced fear memory. Neurosci Lett 2009; 456:39-43. [PMID: 19429130 DOI: 10.1016/j.neulet.2009.03.068] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2008] [Revised: 03/20/2009] [Accepted: 03/22/2009] [Indexed: 10/21/2022]
Abstract
IL-1beta is a proinflammatory cytokine that contributes to psychological stress responses and has been implicated in various psychiatric disorders most notably depression. Preclinical studies also demonstrate that IL-1beta modulates anxiety- and fear-related behaviors, although these findings are difficult to assess because IL-1beta infusions influence locomotor activity and nociception. Here we demonstrate that IL-1RI null mice exhibit a behavioral phenotype consistent with a decrease in anxiety-related behaviors. This includes significant effects in the elevated plus maze, light-dark, and novelty-induced hypophagia tests compared to wild-type mice, with no differences in locomotor activity. With regard to fear conditioning, IL-1RI null mice showed more freezing in auditory and contextual fear conditioning tests, and there was no effect on pain sensitivity. Taken together, the results indicate that the IL-1beta/IL-1RI signaling pathway induces anxiety-related behaviors and impairs fear memory.
Collapse
Affiliation(s)
- Ja Wook Koo
- Department of Neuroscience, Mount Sinai School of Medicine, 1425 Madison Avenue, New York, NY 10029, United States
| | | |
Collapse
|
128
|
Sirivelu M, Shin A, Perez G, MohanKumar P, MohanKumar S. Effect of L-dopa on interleukin-1 beta-induced suppression of luteinizing hormone secretion in intact female rats. Hum Reprod 2009; 24:718-25. [PMID: 19054775 PMCID: PMC2646791 DOI: 10.1093/humrep/den434] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2008] [Revised: 10/24/2008] [Accepted: 11/05/2008] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND The cytokine, interleukin-1 beta (IL-1 beta), increases during immune stress and is known to suppress the preovulatory luteinizing hormone (LH) surge in female rats by decreasing hypothalamic norepinephrine (NE). We hypothesized that IL-1 beta could produce this effect by decreasing NE biosynthesis. METHODS Female Sprague-Dawley rats were implanted with a push-pull cannula in the medial preoptic area (MPA) of the hypothalamus and a catheter in the jugular vein. They were treated i.p. with the vehicle or 5 microg of IL-1 beta, the NE precursor, L-dopa, or a combination of L-dopa and IL-1 beta at 1300 hours on the day of proestrus. They were subjected to push-pull perfusion and serial blood sampling. Perfusates were analyzed for NE levels and serum samples for LH. RESULTS IL-1 beta treatment blocked the increase in NE levels in the MPA and the LH surge. Treatment with L-dopa was able to partially restore both NE and LH levels during the afternoon of proestrus. IL-1 beta treatment caused failure of ovulation and this effect was also reversed by L-dopa. CONCLUSIONS These results suggest that IL-1 beta could decrease NE levels in the MPA to suppress reproductive functions and L-dopa can be used to counter this effect.
Collapse
Affiliation(s)
- M.P. Sirivelu
- Comparative Medicine and Integrative Biology, College of Veterinary Medicine, Michigan State University, B-440 Life Sciences Building, East Lansing, MI 48824, USA
| | - A.C. Shin
- Neuroscience Graduate Programs, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - G.I. Perez
- Comparative Medicine and Integrative Biology, College of Veterinary Medicine, Michigan State University, B-440 Life Sciences Building, East Lansing, MI 48824, USA
- Department of Physiology, College of Veterinary Medicine, Michigan State University, B-440 Life Sciences Building, East Lansing, MI 48824, USA
| | - P.S. MohanKumar
- Comparative Medicine and Integrative Biology, College of Veterinary Medicine, Michigan State University, B-440 Life Sciences Building, East Lansing, MI 48824, USA
- Neuroscience Graduate Programs, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA
- Department of Pathobiology & Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, B-440 Life Sciences Building, East Lansing, MI 48824, USA
| | - S.M.J. MohanKumar
- Comparative Medicine and Integrative Biology, College of Veterinary Medicine, Michigan State University, B-440 Life Sciences Building, East Lansing, MI 48824, USA
- Neuroscience Graduate Programs, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA
- Department of Pharmacology and Toxicology, College of Veterinary Medicine, Michigan State University, B-440 Life Sciences Building, East Lansing, MI 48824, USA
| |
Collapse
|
129
|
Chen Q, Zhang H, Li Q, An Y, Herkenham M, Lai W, Popovich P, Agarwal S, Quan N. Three Promoters Regulate Tissue- and Cell Type-specific Expression of Murine Interleukin-1 Receptor Type I. J Biol Chem 2009; 284:8703-13. [PMID: 19196714 DOI: 10.1074/jbc.m808261200] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The type 1 interleukin-1 receptor (IL-1R1) mediates diverse functions of interleukin-1 (IL-1) in the nervous, immune, and neuroendocrine systems. It has been suggested previously that the versatile functions of IL-1 may in part be conferred by the multiple promoters of IL-1R1 that have been identified for the human IL-1R1 gene. Promoters for murine IL-1R1 (mIL-1R1) gene have not been studied in detail. We performed 5'-rapid amplification of cDNA ends to determine the transcription start sites (TSS) in mIL-1R1, using mRNAs derived from 24 different tissues. The results revealed three putative TSSs of mIL-1R1. Three full-length cDNAs containing these distinct TSSs were recovered in screens of cloned cDNA libraries. Translation of these cDNAs produced IL-1R1 proteins that were verified by Western blot analysis. IL-1 stimulation of the individual IL-1R1 proteins resulted in the activation of NF-kappaB. Promoter-reporter assay for genomic DNA sequences immediately upstream of the three TSSs validated that the sequences possess promoter activity in a cell type-specific manner. These promoters are termed P1, P2, and P3 of the mIL-1R1, in 5' to 3' order. Quantitative PCR analysis of P1-, P2-, and P3-specific mIL-1R1 mRNAs showed that there is tissue-specific distribution of these mRNAs in vivo, and there are distinct patterns of P1, P2, and P3 mRNA expression in different cell lines. In the brain, P3 mRNA is expressed preferentially in the dentate gyrus. Further, glucocorticoids differentially regulate these promoters in a cell type-specific manner. Together, these results suggest that the different IL-1R1 promoters contribute to the discrete and diverse actions of IL-1.
Collapse
Affiliation(s)
- Qun Chen
- Department of Oral Biology, Ohio State University, Columbus, Ohio 43210, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
130
|
Konsman JP, Veeneman J, Combe C, Poole S, Luheshi GN, Dantzer R. Central nervous action of interleukin-1 mediates activation of limbic structures and behavioural depression in response to peripheral administration of bacterial lipopolysaccharide. Eur J Neurosci 2009; 28:2499-510. [PMID: 19087175 DOI: 10.1111/j.1460-9568.2008.06549.x] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Although receptors for the pro-inflammatory cytokine interleukin-1 have long been known to be expressed in the brain, their role in fever and behavioural depression observed during the acute phase response (APR) to tissue infection remains unclear. This may in part be due to the fact that interleukin-1 in the brain is bioactive only several hours after peripheral administration of bacterial lipopolysaccharide (LPS). To study the role of cerebral interleukin-1 action in temperature and behavioural changes, and activation of brain structures during the APR, interleukin-1 receptor antagonist (IL-1ra; 100 microg) was infused into the lateral brain ventricle 4 h after intraperitoneal (i.p.) LPS injection (250 microg/kg) in rats. I.p. LPS administration induced interleukin-1beta (IL-1beta) production in systemic circulation as well as in brain circumventricular organs and the choroid plexus. Intracerebroventricular (i.c.v.) infusion of IL-1ra 4 h after i.p. LPS injection attenuated the reduction in social interaction, a cardinal sign of behavioural depression during sickness, and c-Fos expression in the amygdala and bed nucleus of the stria terminalis. However, LPS-induced fever, rises in plasma corticosterone, body weight loss and c-Fos expression in the hypothalamus and caudal brainstem were not altered by i.c.v. infusion of IL-1ra. These findings, together with our previous observations showing that i.c.v. infused IL-1ra diffuses throughout perivascular spaces, where macrophages express interleukin-1 receptors, can be interpreted to suggest that circulating or locally produced brain IL-1beta acts on these cells to bring about behavioural depression and activation of limbic structures during the APR after peripheral LPS administration.
Collapse
Affiliation(s)
- J P Konsman
- PsychoNeuroImmunologie, Nutrition et Génétique, CNRS UMR 5526/INRA UMR 1286, Université Victor Ségalen Bordeaux 2, Bordeaux 33076, France.
| | | | | | | | | | | |
Collapse
|
131
|
Mitchell K, Yang HYT, Berk JD, Tran JH, Iadarola MJ. Monocyte chemoattractant protein-1 in the choroid plexus: a potential link between vascular pro-inflammatory mediators and the CNS during peripheral tissue inflammation. Neuroscience 2009; 158:885-95. [PMID: 19032979 PMCID: PMC2668531 DOI: 10.1016/j.neuroscience.2008.10.047] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2008] [Revised: 10/17/2008] [Accepted: 10/28/2008] [Indexed: 12/22/2022]
Abstract
During peripheral tissue inflammation, inflammatory processes in the CNS can be initiated by blood-borne pro-inflammatory mediators. The choroid plexus, the site of cerebrospinal fluid (CSF) production, is a highly specialized interface between the vascular system and CNS, and thus, this structure may be an important element in communication between the vascular compartment and the CNS during peripheral tissue inflammation. We investigated the potential participation of the choroid plexus in this process during peripheral tissue inflammation by examining expression of the small inducible cytokine A2 (SCYA2) gene which codes for monocyte chemoattractant protein-1 (MCP-1). MCP-1 protein was previously reported to be induced in a variety of cells during peripheral tissue inflammation. In the basal state, SCYA2 is highly expressed in the choroid plexus as compared with other rat CNS tissues. During hind paw inflammation, SCYA2 expression was significantly elevated in choroid plexus, whereas it remained unchanged in a variety of brain regions. The SCYA2-expressing cells were strongly associated with the choroid plexus as vascular depletion of blood cells by whole-body saline flush did not significantly alter SCYA2 expression in the choroid plexus. In situ hybridization suggested that the SCYA2-expressing cells were localized to the choroid plexus stroma. To elucidate potential molecular mechanisms of SCYA2 increase, we examined genes in the nuclear factor-kappa B (NF-kappaB) signaling cascade including tumor necrosis factor-alpha (TNF-alpha), interleukin-1beta (IL-1beta) and inhibitor of kappa B alpha (IkappaBalpha) in choroid tissue. Given that we also detected increased levels of MCP-1 protein by ELISA, we sought to identify potential downstream targets of MCP-1 and observed altered expression levels of mRNAs encoding tight junction proteins TJP2 and claudin 5. Finally, we detected a substantial up-regulation of the transcript encoding endothelial leukocyte adhesion molecule 1 (E-selectin), a molecule which could participate in leukocyte recruitment to the choroid plexus along with MCP-1. Together, these results suggest that profound changes occur in the choroid plexus during peripheral tissue inflammation, likely initiated by blood-borne inflammatory mediators, which may modify events in CNS.
Collapse
Affiliation(s)
- K. Mitchell
- Neurobiology and Pain Therapeutics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Building 49, Room 1C20, 49 Convent Drive, MSC 4410, Bethesda, MD 20892-4410, USA
| | - H.-Y. T. Yang
- Neurobiology and Pain Therapeutics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Building 49, Room 1C20, 49 Convent Drive, MSC 4410, Bethesda, MD 20892-4410, USA
| | - J. D. Berk
- Neurobiology and Pain Therapeutics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Building 49, Room 1C20, 49 Convent Drive, MSC 4410, Bethesda, MD 20892-4410, USA
| | - J. H. Tran
- Neurobiology and Pain Therapeutics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Building 49, Room 1C20, 49 Convent Drive, MSC 4410, Bethesda, MD 20892-4410, USA
| | - M. J. Iadarola
- Neurobiology and Pain Therapeutics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Building 49, Room 1C20, 49 Convent Drive, MSC 4410, Bethesda, MD 20892-4410, USA
| |
Collapse
|
132
|
Tumor necrosis factor-α, interleukin-1β and nitric oxide induce calcium transients in distinct populations of cells cultured from the rat area postrema. J Neuroimmunol 2009; 206:44-51. [DOI: 10.1016/j.jneuroim.2008.10.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2008] [Revised: 10/22/2008] [Accepted: 10/23/2008] [Indexed: 01/23/2023]
|
133
|
Russell JA, Douglas AJ, Brunton PJ. Reduced Hypothalamo-pituitary-adrenal Axis Stress Responses in Late Pregnancy. Ann N Y Acad Sci 2008; 1148:428-38. [DOI: 10.1196/annals.1410.032] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
134
|
Marty V, El Hachmane M, Amédée T. Dual modulation of synaptic transmission in the nucleus tractus solitarius by prostaglandin E2 synthesized downstream of IL-1beta. Eur J Neurosci 2008; 27:3132-50. [PMID: 18598258 DOI: 10.1111/j.1460-9568.2008.06296.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The activation of the innate immune system induces the production of blood-borne proinflammatory cytokines like interleukin-1beta (IL-1beta), which in turn triggers brain-mediated adaptative responses referred to as sickness behaviour. These responses involve the modulation of neural networks in key regions of the brain. The nucleus tractus solitarius (NTS) of the brainstem is a key nucleus for immune-to-brain signalling. It is the main site of termination of vagal afferents and is adjacent to the area postrema, a circumventricular organ allowing blood-borne action of circulating IL-1beta. Although it is well described that IL-1beta activates cerebral endothelial and glial cells, it is still unknown if and how IL-1beta or downstream-synthesized molecules impact NTS synaptic function. In this study we report that IL-1beta did not modulate NTS synaptic transmission per se, whereas prostaglandin E(2) (PGE(2)), which is produced downstream of IL-1beta, produced opposite effects on spontaneous and evoked release. On the one hand, PGE(2) facilitated glutamatergic transmission between local NTS neurons by enhancing the frequency of spontaneous excitatory postsynaptic currents through a presynaptic receptor different from the classical EP1-4 subtypes. On the other hand, PGE(2) also depressed evoked excitatory input from vagal afferent terminals through presynaptic EP3 receptors coupled to G-proteins linked to adenylyl cyclase and protein kinase A activity. Our data show that IL-1beta-induced PGE(2) can modulate evoked and spontaneous release in the NTS differentially through different mechanisms. These data unravel some molecular mechanisms by which innate immune stimuli could signal to, and be integrated within, the brainstem to produce central adaptative responses.
Collapse
Affiliation(s)
- Vincent Marty
- Psychoneuroimmunology, Nutrition and Genetics (PsyNuGen), UMR 5226 CNRS-UB2/UMR 1286 INRA-UB2, Université Bordeaux 2, Institut François Magendie, 146 Rue Léo-Saignat, F-33077 Bordeaux, France
| | | | | |
Collapse
|
135
|
Scarlett JM, Zhu X, Enriori PJ, Bowe DD, Batra AK, Levasseur PR, Grant WF, Meguid MM, Cowley MA, Marks DL. Regulation of agouti-related protein messenger ribonucleic acid transcription and peptide secretion by acute and chronic inflammation. Endocrinology 2008; 149:4837-45. [PMID: 18583425 PMCID: PMC2582916 DOI: 10.1210/en.2007-1680] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Agouti-related protein (AgRP) is an orexigenic neuropeptide produced by neurons in the hypothalamic arcuate nucleus (ARC) that is a key component of central neural circuits that control food intake and energy expenditure. Disorders in energy homeostasis, characterized by hypophagia and increased metabolic rate, frequently develop in animals with either acute or chronic diseases. Recently, studies have demonstrated that proopiomelanocortin-expressing neurons in the ARC are activated by the proinflammatory cytokine IL-1beta. In the current study, we sought to determine whether inflammatory processes regulate the expression of AgRP mRNA and to characterize the response of AgRP neurons to IL-1beta. Here, we show by real-time RT-PCR and in situ hybridization analysis that AgRP mRNA expression in rodents is increased in models of acute and chronic inflammation. AgRP neurons were found to express the type I IL-1 receptor, and the percentage of expression was significantly increased after peripheral administration of lipopolysaccharide. Furthermore, we demonstrate that IL-1beta inhibits the release of AgRP from hypothalamic explants. Collectively, these data indicate that proinflammatory signals decrease the secretion of AgRP while increasing the transcription of the AgRP gene. These observations suggest that AgRP neurons may participate with ARC proopiomelanocortin neurons in mediating the anorexic and metabolic responses to acute and chronic disease processes.
Collapse
MESH Headings
- Agouti-Related Protein/genetics
- Agouti-Related Protein/metabolism
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Arcuate Nucleus of Hypothalamus/cytology
- Arcuate Nucleus of Hypothalamus/physiology
- Brain Tissue Transplantation
- Chronic Disease
- Disease Models, Animal
- Inflammation/chemically induced
- Inflammation/immunology
- Inflammation/physiopathology
- Interleukin-1beta/metabolism
- Ketorolac/pharmacology
- Kidney Failure, Chronic/immunology
- Kidney Failure, Chronic/physiopathology
- Lipopolysaccharides/pharmacology
- Male
- Mice
- Mice, Inbred C57BL
- Neoplasms/immunology
- Neoplasms/physiopathology
- Neurons/physiology
- Prostaglandins/metabolism
- Proto-Oncogene Proteins c-fos/genetics
- RNA, Messenger/genetics
- Rats
- Rats, Inbred F344
- Rats, Sprague-Dawley
- Receptors, Interleukin-1/genetics
- Transcription, Genetic/immunology
Collapse
Affiliation(s)
- Jarrad M Scarlett
- Center for the Study of Weight Regulation and Associated Disorders, Department of Pediatrics, Oregon Health & Science University Child Development and Rehabiliation Center Portland, Portland, OR 97239, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
136
|
Khatami R, von Büdingen HC, Bassetti CL. Sleep–Wake Disturbances in Neurologic Autoimmune Disorders. Sleep Med Clin 2008. [DOI: 10.1016/j.jsmc.2008.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
137
|
Coogan AN, Wyse CA. Neuroimmunology of the circadian clock. Brain Res 2008; 1232:104-12. [PMID: 18703032 DOI: 10.1016/j.brainres.2008.07.087] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2008] [Revised: 03/31/2008] [Accepted: 07/11/2008] [Indexed: 12/12/2022]
Abstract
Circadian timekeeping is a ubiquitous feature of all eukaryotes which allows for the imposition of a biologically appropriate temporal architecture on an animal's physiology, behavior and metabolism. There is growing evidence that in mammals the processes of circadian timing are under the influence of the immune system. Such a role for the neuroimmune regulation of the circadian clock has inferences for phenomena such as sickness behavior. Conversely, there is also accumulating evidence for a circadian influence on immune function, raising the likelihood that there is a bidirectional communication between the circadian and immune systems. In this review, we examine the evidence for these interactions, including circadian rhythmicity in models of disease and immune challenge, distribution of cytokines and their receptors in the suprachiasmatic nucleus of the hypothalamus, the site of the master circadian pacemaker, and the evidence for endogenous circadian timekeeping in immune cells.
Collapse
Affiliation(s)
- Andrew N Coogan
- Neuroscience and Molecular Psychiatry, Institute of Life Science, School of Medicine, Swansea University, Swansea, SA2 8PP, UK.
| | | |
Collapse
|
138
|
Rivest S, Lacroix S, Vallières L, Nadeau S, Zhang J, Laflamme N. How the Blood Talks to the Brain Parenchyma and the Paraventricular Nucleus of the Hypothalamus During Systemic Inflammatory and Infectious Stimuli. ACTA ACUST UNITED AC 2008. [DOI: 10.1111/j.1525-1373.2000.22304.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
139
|
Quan N. Immune-to-brain signaling: how important are the blood-brain barrier-independent pathways? Mol Neurobiol 2008; 37:142-52. [PMID: 18563639 PMCID: PMC2780323 DOI: 10.1007/s12035-008-8026-z] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2008] [Accepted: 05/09/2008] [Indexed: 10/21/2022]
Abstract
A conceptual obstacle for understanding immune-to-brain signaling is the issue of the blood-brain barrier (BBB). In the last 30 years, several pathways have been investigated to address the question of how peripheral immune signals are transmitted into the brain. These pathways can be categorized into two types: BBB-dependent pathways and BBB-independent pathways. BBB-dependent pathways involve the BBB as a relay station or porous barrier, whereas BBB-independent pathways use neuronal routes that bypass the BBB. Recently, a complete BBB-dependent ascending pathway for immune-to-brain signaling has been described. Details of BBB-independent pathways are still under construction. In this review, I will summarize the current progress in unraveling immune-to-brain signaling pathways. In addition, I will provide a critical analysis of the literature to point to areas where our knowledge of the immunological afferent signaling to the central nervous system is still sorely lacking.
Collapse
Affiliation(s)
- Ning Quan
- Institute of Behavior Medicine, Ohio State University, 4179 Postle Hall, 305 W. 12th Ave, Columbus, OH 43210-1094, USA.
| |
Collapse
|
140
|
Grill M, Heinemann A, Hoefler G, Peskar BA, Schuligoi R. Effect of endotoxin treatment on the expression and localization of spinal cyclooxygenase, prostaglandin synthases, and PGD2 receptors. J Neurochem 2008; 104:1345-57. [PMID: 18028337 DOI: 10.1111/j.1471-4159.2007.05078.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Systemic inflammation leads to increased expression of spinal cyclooxygenase (COX)-2 and to a subsequent increase of prostaglandin (PG) biosynthesis, which contribute to the development of hyperalgesia and allodynia. In this study, endotoxin caused a sequential induction of membrane bound prostaglandin E synthase-1 and lipocalin-type PGD synthase (L-PGDS) in the mouse spinal cord. L-PGDS expression was detected in the leptomeninges, oligodendrocytes, and interestingly, in discrete perivascular cells. Endotoxin-caused increase was most prominent in oligodendrocytes. Endotoxin-induced COX-2 and membrane bound prostaglandin E synthase-1 were restricted to the leptomeninges and perivascular cells. COX-1 was not influenced by endotoxin. We found COX-1 expressed in microglia, some of them in close proximity to L-PGDS-positive oligodendrocytes and co-localization of COX-1 with L-PGDS in perivascular and leptomeningeal cells under control conditions. It can be assumed, that PGD2 biosynthesis under control conditions is mediated via COX-1 and that during inflammation, increased PGD2 is dependent on COX-2. We found the PGD2 receptors DP1 and chemoattractant receptor homologous molecule expressed on T helper type 2 cells (CRTH2) localized in neurons of the dorsal, and motoneurons in the ventral horn. The localization of the PGD2 receptors DP1 and CRTH in spinal cord neurons, particularly in neurons of lamina I and II involved in the processing of nociceptive stimuli, supports a role of PGD2 under inflammatory conditions.
Collapse
MESH Headings
- Animals
- Cytokines/biosynthesis
- Cytokines/genetics
- Cytokines/metabolism
- Dose-Response Relationship, Drug
- Gene Expression Regulation, Enzymologic/drug effects
- Gene Expression Regulation, Enzymologic/physiology
- Inflammation Mediators/metabolism
- Injections, Intraperitoneal
- Lipopolysaccharides/administration & dosage
- Male
- Mice
- Mice, Inbred BALB C
- Prostaglandin D2/metabolism
- Prostaglandin-Endoperoxide Synthases/biosynthesis
- Prostaglandin-Endoperoxide Synthases/genetics
- Prostaglandin-Endoperoxide Synthases/metabolism
- Receptors, Immunologic/biosynthesis
- Receptors, Immunologic/genetics
- Receptors, Immunologic/metabolism
- Receptors, Prostaglandin/biosynthesis
- Receptors, Prostaglandin/genetics
- Receptors, Prostaglandin/metabolism
- Spinal Cord/drug effects
- Spinal Cord/enzymology
- Spinal Cord/metabolism
- Time Factors
Collapse
Affiliation(s)
- Magdalena Grill
- Institute of Experimental and Clinical Pharmacology, Medical University Graz, Graz, Austria
| | | | | | | | | |
Collapse
|
141
|
Whitaker KW, Reyes TM. Central blockade of melanocortin receptors attenuates the metabolic and locomotor responses to peripheral interleukin-1beta administration. Neuropharmacology 2008; 54:509-20. [PMID: 18082228 PMCID: PMC2323450 DOI: 10.1016/j.neuropharm.2007.10.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2007] [Revised: 10/08/2007] [Accepted: 10/26/2007] [Indexed: 01/28/2023]
Abstract
Loss of appetite and cachexia is an obstacle in the treatment of chronic infection and cancer. Proinflammatory cytokines released from activated immune cells and acting in the central nervous system (CNS) are prime candidates for mediating these metabolic changes, potentially affecting both energy intake as well as energy expenditure. The effect of intravenous administration of two proinflammatory cytokines, interleukin (IL)-1beta (15 microg/kg) and tumor necrosis factor (TNF)-alpha (10 microg/kg), on food and water intake, locomotor activity, oxygen consumption (VO2), and respiratory exchange ratio (RER) was evaluated. The two cytokines elicited a comparable decrease in food intake and activated similar numbers of cells in the paraventricular nucleus of the hypothalamus (PVH), a region that plays a critical role in the regulation of appetite and metabolism (determined via expression of the immediate early gene, c-fos). However, only IL-1beta reduced locomotion and RER, and increased VO2, while TNF-alpha was without effect. To examine the role of the melanocortins in mediating IL-1beta- induced metabolic changes, animals were pretreated centrally with a melanocortin receptor antagonist, HS014. Pretreatment with HS014 blocked the effect of IL-1beta on food intake and RER at later time points (beyond 8 h post injection), as well as the hypoactivity and increased metabolic rate. Further, HS014 blocked the induction of Fos-ir in the PVH. These data highlight the importance of the melanocortin system, particularly within the PVH, in mediating a broad range of metabolic responses to IL-1beta.
Collapse
Affiliation(s)
- Keith W Whitaker
- Department of Biochemistry, Scripps Florida, Jupiter, FL 33458, USA
| | | |
Collapse
|
142
|
Harry GJ, Funk JA, Lefebvre d'Hellencourt C, McPherson CA, Aoyama M. The type 1 interleukin 1 receptor is not required for the death of murine hippocampal dentate granule cells and microglia activation. Brain Res 2008; 1194:8-20. [PMID: 18191113 PMCID: PMC2274920 DOI: 10.1016/j.brainres.2007.11.076] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2007] [Revised: 11/18/2007] [Accepted: 11/26/2007] [Indexed: 02/06/2023]
Abstract
Alterations in inflammatory process, neuronal death, and glia response have been observed under manipulation of interleukin-1 (IL-1) and subsequent signaling through the type 1 IL-1 receptor (IL-1R1). To investigate the influence of IL-1R1 activation in the pathophysiology of a chemical-induced injury to the murine hippocampus, we examined the level and pattern of neuronal death and neuroinflammation in male weanling mice exposed to trimethyltin hydroxide (2.0 mg TMT/kg, i.p.). Dentate granule cell death occurred at 6 h post-TMT as detected by active caspase 3 immunostaining and presence of lectin positive microglia. The severity of neuronal death and microglia response increased by 12-24 h with elevations in mRNA levels for TNFalpha and IL-1alpha. In IL-1R1 null (IL-1R1-/-) mice, the pattern and severity of neuronal death at 24 or 72 h post-TMT was similar as compared to wildtype (WT) mice. In both groups, mRNA levels for TNFalpha and MIP-1alpha were elevated, no significant change was seen in either IL-1alpha or IL-1beta, and the early activation of microglia, including their ability to progress to a phagocytic phenotype, was maintained. Compared to WT mice, IL-1R1-/- mice displayed a limited glial fibrillary acidic protein (GFAP) astrocytic response, as well as a preferential induction in mRNA levels of Fas signaling components. Cumulatively, these results indicate that IL-1R1 activation is not necessary for TMT-induced death of dentate granule neurons or local activation of microglia; however, IL-1R1 signaling is involved in mediating the structural response of astrocytes to injury and may regulate apoptotic mechanisms via Fas signaling components.
Collapse
Affiliation(s)
- G Jean Harry
- Neurotoxicology Group, Laboratory of Neurobiology, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC 27709, USA.
| | | | | | | | | |
Collapse
|
143
|
Bhatt S, Bhatt R, Zalcman SS, Siegel A. Role of IL-1 beta and 5-HT2 receptors in midbrain periaqueductal gray (PAG) in potentiating defensive rage behavior in cat. Brain Behav Immun 2008; 22:224-33. [PMID: 17890051 PMCID: PMC2276628 DOI: 10.1016/j.bbi.2007.07.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2007] [Revised: 07/17/2007] [Accepted: 07/28/2007] [Indexed: 11/26/2022] Open
Abstract
Feline defensive rage, a form of aggressive behavior that occurs in response to a threat can be elicited by electrical stimulation of the medial hypothalamus or midbrain periaqueductal gray (PAG). Our laboratory has recently begun a systematic examination of the role of cytokines in the regulation of rage and aggressive behavior. It was shown that the cytokine, interleukin-2 (IL-2), differentially modulates defensive rage when microinjected into the medial hypothalamus and PAG by acting through separate neurotransmitter systems. The present study sought to determine whether a similar relationship exists with respect to interleukin 1-beta (IL-1 beta), whose receptor activation in the medial hypothalamus potentiates defensive rage. Thus, the present study identified the effects of administration of IL-1 beta into the PAG upon defensive rage elicited from the medial hypothalamus. Microinjections of IL-1 beta into the dorsal PAG significantly facilitated defensive rage behavior elicited from the medial hypothalamus in a dose and time dependent manner. In addition, the facilitative effects of IL-1 beta were blocked by pre-treatment with anti-IL-1 beta receptor antibody, while IL-1 beta administration into the PAG had no effect upon predatory attack elicited from the lateral hypothalamus. The findings further demonstrated that IL-1 beta's effects were mediated through 5-HT(2) receptors since pretreatment with a 5-HT(2C) receptors antagonist blocked the facilitating effects of IL-1 beta. An extensive pattern of labeling of IL-1 beta and 5-HT(2C) receptors in the dorsal PAG supported these findings. The present study demonstrates that IL-beta in the dorsal PAG, similar to the medial hypothalamus, potentiates defensive rage behavior and is mediated through a 5-HT(2C) receptor mechanism.
Collapse
Affiliation(s)
- Suresh Bhatt
- Department of Neurology & Neurosciences, New Jersey Medical School, University of Medicine and Dentistry of New Jersey, 185 South Orange Avenue, Newark, NJ 07103, USA
| | - Rekha Bhatt
- Department of Neurology & Neurosciences, New Jersey Medical School, University of Medicine and Dentistry of New Jersey, 185 South Orange Avenue, Newark, NJ 07103, USA
| | - Steven S Zalcman
- Department of Psychiatry, New Jersey Medical School, University of Medicine and Dentistry of New Jersey, 185 South Orange Avenue, Newark, NJ 07103, USA
| | - Allan Siegel
- Department of Neurology & Neurosciences, New Jersey Medical School, University of Medicine and Dentistry of New Jersey, 185 South Orange Avenue, Newark, NJ 07103, USA
- Department of Psychiatry, New Jersey Medical School, University of Medicine and Dentistry of New Jersey, 185 South Orange Avenue, Newark, NJ 07103, USA
| |
Collapse
|
144
|
Becskei C, Riediger T, Hernádfalvy N, Arsenijevic D, Lutz TA, Langhans W. Inhibitory effects of lipopolysaccharide on hypothalamic nuclei implicated in the control of food intake. Brain Behav Immun 2008; 22:56-64. [PMID: 17624718 DOI: 10.1016/j.bbi.2007.06.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2007] [Revised: 06/01/2007] [Accepted: 06/02/2007] [Indexed: 10/23/2022] Open
Abstract
The arcuate nucleus (Arc) and the lateral hypothalamic area (LHA), two key hypothalamic nuclei regulating feeding behavior, express c-Fos, a marker of neuronal activation in fasted animals. This is reversed by refeeding. In the present study we tested whether an anorectic dose of lipopolysaccharide (LPS), the cell wall component of Gram-negative bacteria, also inhibits fasting-induced c-Fos expression in these hypothalamic nuclei. This would suggest that they are involved in anorexia during bacterial infections as well. We also studied whether LPS modulates the activity of orexin-A positive (OX+) LHA neurons. Food deprived BALB/c mice were injected with LPS or saline and were sacrificed 4 or 6h later. Four hours after injection, LPS reduced the number of c-Fos positive cells in the Arc and in the LHA, but had no effect on c-Fos in OX+ neurons. Six hours after injection, LPS reduced c-Fos expression in the LHA, both in the OX- and OX+ neurons, but not in the Arc. These results show that LPS modulates neuronal activity in the Arc and LHA similar to feeding-related stimuli, suggesting that the observed effects might contribute to the anorectic effect of LPS. Thus, physiological satiety signals released during refeeding and anorexia during bacterial infection seem to engage similar neuronal substrates.
Collapse
Affiliation(s)
- Csilla Becskei
- Institute of Veterinary Physiology and Zurich Centre of Human Integrative Physiology, University of Zurich, Winterthurerstrasse 260, 8057 Zurich, Switzerland.
| | | | | | | | | | | |
Collapse
|
145
|
Conti B, Tabarean I, Sanchez-Alavez M, Davis C, Brownell S, Behrens M, Bartfai T. Cytokine Receptors in the Brain. CYTOKINES AND THE BRAIN 2008. [DOI: 10.1016/s1567-7443(07)10002-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
146
|
Price CJ, Hoyda TD, Ferguson AV. The area postrema: a brain monitor and integrator of systemic autonomic state. Neuroscientist 2007; 14:182-94. [PMID: 18079557 DOI: 10.1177/1073858407311100] [Citation(s) in RCA: 183] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The area postrema is a medullary structure lying at the base of the fourth ventricle. The area postrema's privileged location outside of the blood-brain barrier make this sensory circumventricular organ a vital player in the control of autonomic functions by the central nervous system. By virtue of its lack of tight junctions between endothelial cells in this densely vascularized structure and the presence of fenestrated capillaries, peptide and other physiological signals borne in the blood have direct access to neurons that project to brain areas with important roles in the autonomic control of many physiological systems, including the cardiovascular system and systems controlling feeding and metabolism. However, the area postrema is not simply a conduit through which signals flow into the brain, but it is now being recognized as the initial site of integration for these signals as they enter the circuitry of the central nervous system.
Collapse
|
147
|
Brunton PJ, Russell JA. Attenuated hypothalamo-pituitary-adrenal axis responses to immune challenge during pregnancy: the neurosteroid opioid connection. J Physiol 2007; 586:369-75. [PMID: 17991694 DOI: 10.1113/jphysiol.2007.146233] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
In late pregnancy maternal hypothalamo-pituitary-adrenal (HPA) axis responses to emotional and physical stressors are attenuated. This is expected to minimize the detrimental programming effects of glucocorticoid exposure on the fetuses. We have utilized a model of immune challenge, systemic administration of interleukin-1beta (IL-1beta), to investigate the underlying mechanisms. Intravenous IL-1beta activates corticotropin-releasing hormone (CRH) neurones in the parvocellular division of the paraventricular nucleus (pPVN) via noradrenergic (A2 cell group) neurones in the nucleus tractus solitarii (NTS). Despite comparable activation of these brainstem neurones by IL-1beta in virgin and in late pregnant rats, pPVN CRH neurones are activated only in virgin rats. As a consequence IL-1beta fails to evoke ACTH and corticosterone secretion in late pregnant rats, in contrast to virgin rats. Suppressed responsiveness of the CRH neurones, and hence the HPA axis, following IL-1beta in late pregnancy is explained by presynaptic inhibition of noradrenaline release in the pPVN, due to increased endogenous enkephalin and mu-opioid receptor production in brainstem NTS neurones. The factor that signals to the brain the pregnancy status of the animal and stimulates opioid production in the brainstem is allopregnanolone, a neurosteroid metabolite of progesterone. The supporting evidence for these mechanisms is discussed.
Collapse
Affiliation(s)
- Paula J Brunton
- Laboratory of Neuroendocrinology, Centre for Integrative Physiology, Hugh Robson Building, University of Edinburgh, George Square, Edinburgh EH8 9XD, UK
| | | |
Collapse
|
148
|
Ruud J, Blomqvist A. Identification of rat brainstem neuronal structures activated during cancer-induced anorexia. J Comp Neurol 2007; 504:275-86. [PMID: 17640050 DOI: 10.1002/cne.21407] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In cancer-related anorexia, body weight loss is paradoxically associated with reduced appetite, which is contrary to the situation during starvation, implying that the normal coupling of food intake to energy expenditure is disarranged. Here we examined brainstem mechanisms that may underlie suppression of food intake in a rat model of cancer anorexia. Cultured Morris 7777 hepatoma cells were injected subcutaneously in Buffalo rats, resulting in slowly growing tumor and reduced food intake and body weight loss after about 10 days. The brainstem was examined for induced expression of the transcription factors Fos and FosB as signs of neuronal activation. The results showed that anorexia and retarded body weight growth were associated with Fos protein expression in the area postrema, the general visceral region of the nucleus of the solitary tract, and the external lateral parabrachial nucleus, structures that also display Fos after peripheral administration of satiating or anorexigenic stimuli. The magnitude of the Fos expression was specifically related to the size of induced tumor, and not associated with weight loss per se, because it was not present in pair-fed or food-deprived rats. It also appeared to be independent of proinflammatory cytokines, as determined by the absence of increased cytokine levels in plasma and induced cytokine and cyclooxygenase expression in the brain. The findings thus provide evidence that cancer-associated anorexia and weight loss in this model is associated with activation of brainstem circuits involved in the suppression of food intake, and suggest that this occurs by inflammatory-independent mechanisms.
Collapse
Affiliation(s)
- Johan Ruud
- Division of Cell Biology, Department of Biomedicine and Surgery, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| | | |
Collapse
|
149
|
Ching S, Zhang H, Belevych N, He L, Lai W, Pu XA, Jaeger LB, Chen Q, Quan N. Endothelial-specific knockdown of interleukin-1 (IL-1) type 1 receptor differentially alters CNS responses to IL-1 depending on its route of administration. J Neurosci 2007; 27:10476-86. [PMID: 17898219 PMCID: PMC6673171 DOI: 10.1523/jneurosci.3357-07.2007] [Citation(s) in RCA: 124] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2006] [Revised: 08/08/2007] [Accepted: 08/09/2007] [Indexed: 12/22/2022] Open
Abstract
Interleukin-1 (IL-1) has been implicated as a critical mediator of neuroimmune communication. In the brain, the functional receptor for IL-1, type 1 IL-1 receptor (IL-1R1), is localized primarily to the endothelial cells. In this study, we created an endothelial-specific IL-1R1 knockdown model to test the role of endothelial IL-1R1 in mediating the effects of IL-1. Neuronal activation in the hypothalamus was measured by c-fos expression in the paraventricular nucleus and the ventromedial preoptic area. In addition, two specific sickness symptoms, febrile response and reduction of locomotor activity, were studied. Intracerebroventricular injection of IL-1 induced leukocyte infiltration into the CNS, activation of hypothalamic neurons, fever, and reduced locomotor activity in normal mice. Endothelial-specific knockdown of IL-1R1 abrogated all these responses. Intraperitoneal injection of IL-1 also induced neuronal activation in the hypothalamus, fever, and reduced locomotor activity, without inducing leukocyte infiltration into the brain. Endothelial-specific knockdown of IL-1R1 suppressed intraperitoneal IL-1-induced fever, but not the induction of c-fos in hypothalamus. When IL-1 was given intravenously, endothelial knockdown of IL-1R1 abolished intravenous IL-1-induced CNS activation and the two monitored sickness symptoms. In addition, endothelial-specific knockdown of IL-1R1 blocked the induction of cyclooxygenase-2 expression induced by all three routes of IL-1 administration. These results show that the effects of intravenous and intracerebroventricular IL-1 are mediated by endothelial IL-1R1, whereas the effects of intraperitoneal IL-1 are partially dependent on endothelial IL-1R1.
Collapse
Affiliation(s)
| | | | | | | | | | - Xin-an Pu
- Center for Neurobiology, Ohio State University, Columbus, Ohio 43210-1094, and
| | - Laura B. Jaeger
- Department of Pharmacology and Physiology, St. Louis University, St. Louis, Missouri 63106
| | | | | |
Collapse
|
150
|
Scarlett JM, Jobst EE, Enriori PJ, Bowe DD, Batra AK, Grant WF, Cowley MA, Marks DL. Regulation of central melanocortin signaling by interleukin-1 beta. Endocrinology 2007; 148:4217-25. [PMID: 17525125 DOI: 10.1210/en.2007-0017] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Anorexia and involuntary weight loss are common and debilitating complications of a number of chronic diseases and inflammatory states. Proinflammatory cytokines, including IL-1 beta, are hypothesized to mediate these responses through direct actions on the central nervous system. However, the neural circuits through which proinflammatory cytokines regulate food intake and energy balance remain to be characterized. Here we report that IL-1 beta activates the central melanocortin system, a key neuronal circuit in the regulation of energy homeostasis. Proopiomelanocortin (POMC) neurons in the arcuate nucleus of the hypothalamus (ARC) were found to express the type I IL-1 receptor. Intracerebroventricular injection of IL-1 beta induced the expression of Fos protein in ARC POMC neurons but not in POMC neurons in the commissural nucleus of the tractus solitarius. We further show that IL-1 beta increases the frequency of action potentials of ARC POMC neurons and stimulates the release of alpha-MSH from hypothalamic explants in a dose-dependent fashion. Collectively, our data support a model in which IL-1 beta increases central melanocortin signaling by activating a subpopulation of hypothalamic POMC neurons and stimulating their release of alpha-MSH.
Collapse
Affiliation(s)
- Jarrad M Scarlett
- Center for the Study of Weight Regulation and Associated Disorders, Oregon Health and Science University, 3181 Southwest Sam Jackson Park Road, Portland, Oregon 97239, USA
| | | | | | | | | | | | | | | |
Collapse
|