101
|
Alegría-Herrera E, Herrera-Ruiz M, Román-Ramos R, Zamilpa A, Santillán-Urquiza MA, Aguilar MI, Avilés-Flores M, Fuentes-Mata M, Jiménez-Ferrer E. Effect of Ocimum basilicum, Ocimum selloi, and Rosmarinic Acid on Cerebral Vascular Damage in a Chronic Hypertension Model. Biol Pharm Bull 2019; 42:201-211. [PMID: 30713252 DOI: 10.1248/bpb.b18-00574] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The main objective of treatment against hypertension is not only to reduce blood pressure levels, but also to reduce vascular risk in general. In the present work, administering angiotensin II (AGII; 0.2 µg/kg intraperitoneally (i.p.) for 12 weeks) activates the hypothalamic-pituitary-adrenal (HPA) axis, which caused an increase in corticosterone levels, as well as in proinflammatory cytokines (interleukin 1β (IL-1β), interleukin 6 (IL-6), and tumor necrosis factor alpha (TNF-α)) and macrophage chemotactic protein 1 (MCP-1), and decreased anti-inflammatory cytokines (interleukin 10 (IL-10) and interleukin 4 (IL-4)). On observing the behavior in the different models, an anxiogenic effect (elevated plus maze (EPM)) and cognitive impairment (water Morris maze (WMM)) was observed in animals with AGII. By administering organic extracts from Ocimum basilicum (Oba-EtOAc) and Ocimum selloi (Ose-EtOAc), and some doses of rosmarinic acid (RA) (6 weeks per os (p.o.)), the damage caused by AGII was stopped by re-establishing corticosterone serum levels and by decreasing the proinflammatory cytokines and MCP-1.
Collapse
Affiliation(s)
- Elian Alegría-Herrera
- Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social (IMSS)
- Doctorado en Ciencias Biológicas y de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Iztapalapa
- Departamento Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Iztapalapa
| | - Maribel Herrera-Ruiz
- Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social (IMSS)
| | - Rubén Román-Ramos
- Departamento Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Iztapalapa
| | - Alejandro Zamilpa
- Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social (IMSS)
| | | | - María Isabel Aguilar
- Laboratorio 111, Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México
| | | | | | | |
Collapse
|
102
|
Pitocco D, Scavone G, Di Leo M, Vitiello R, Rizzi A, Tartaglione L, Costantini F, Flex A, Galli M, Caputo S, Ghirlanda G, Pontecorvi A. Charcot Neuroarthropathy: From the Laboratory to the Bedside. Curr Diabetes Rev 2019; 16:62-72. [PMID: 31057120 DOI: 10.2174/1573399815666190502121945] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 03/26/2019] [Accepted: 04/17/2019] [Indexed: 01/11/2023]
Abstract
BACKGROUND The diabetic Charcot foot syndrome is a serious and potentially limbthreatening lower-extremity complication of diabetes. INTRODUCTION The present review provides a concise account of the advances made over the last twentyfive years in understanding the pathogenesis and management of Charcot neuroarthropathy (CN). METHODS In this study, the widely known pathogenetic mechanisms underpinning CN are brought into focus, particularly the role of RANKL/RANK/OPG system and advanced glycation end production in the pathogenesis of CN. Furthermore, other potential triggering factors, namely nitric oxide, endothelial dysfunction, macro calcifications and body weight that influence CN have also been discussed. RESULTS The wide range of diagnostic tools available to clinicians for accurate staging of this pathology has been examined, particularly radiological and nuclear medicine imaging. Additionally, the difficult differential diagnosis between osteomyelitis and CN is also elucidated. CONCLUSION The review concludes with the comprehensive summary of the major promising therapeutic strategies, including conservative treatment involving orthopedic devices, pharmacological approach, and the most common surgical techniques currently employed in the diagnosis and treatment of this acute disease.
Collapse
Affiliation(s)
- Dario Pitocco
- Diabetes Care Unit, Institute of Endocrinology, Catholic University of Sacred Heart, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Giuseppe Scavone
- Diabetes Care Unit, Institute of Endocrinology, Catholic University of Sacred Heart, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Mauro Di Leo
- Diabetes Care Unit, Institute of Endocrinology, Catholic University of Sacred Heart, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Raffaele Vitiello
- Diabetes Care Unit, Institute of Endocrinology, Catholic University of Sacred Heart, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Alessandro Rizzi
- Diabetes Care Unit, Institute of Endocrinology, Catholic University of Sacred Heart, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Linda Tartaglione
- Diabetes Care Unit, Institute of Endocrinology, Catholic University of Sacred Heart, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Federica Costantini
- Diabetes Care Unit, Institute of Endocrinology, Catholic University of Sacred Heart, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Andrea Flex
- Institute of Internal Medicine, Catholic University of Sacred Heart, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Marco Galli
- Institute of Orthopedic Surgery, Catholic University of Sacred Heart, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Salvatore Caputo
- Diabetes Care Unit, Institute of Endocrinology, Catholic University of Sacred Heart, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Giovanni Ghirlanda
- Diabetes Care Unit, Institute of Endocrinology, Catholic University of Sacred Heart, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Alfredo Pontecorvi
- Diabetes Care Unit, Institute of Endocrinology, Catholic University of Sacred Heart, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| |
Collapse
|
103
|
Diz-Chaves Y, Toba L, Fandiño J, González-Matías LC, Garcia-Segura LM, Mallo F. The GLP-1 analog, liraglutide prevents the increase of proinflammatory mediators in the hippocampus of male rat pups submitted to maternal perinatal food restriction. J Neuroinflammation 2018; 15:337. [PMID: 30518432 PMCID: PMC6282252 DOI: 10.1186/s12974-018-1370-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 11/18/2018] [Indexed: 12/17/2022] Open
Abstract
Background Perinatal maternal malnutrition is related to altered growth of tissues and organs. The nervous system development is very sensitive to environmental insults, being the hippocampus a vulnerable structure, in which altered number of neurons and granular cells has been observed. Moreover, glial cells are also affected, and increased expression of proinflammatory mediators has been observed. We studied the effect of Glucagon-like peptide-1 receptor (GLP-1R) agonists, liraglutide, which have very potent metabolic and neuroprotective effects, in order to ameliorate/prevent the glial alterations present in the hippocampus of the pups from mothers with food restriction during pregnancy and lactation (maternal perinatal food restriction—MPFR). Methods Pregnant Sprague-Dawley rats were randomly assigned to 50% food restriction (FR; n = 12) or ad libitum controls (CT, n = 12) groups at day of pregnancy 12 (GD12). From GD14 to parturition, pregnant FR and CT rats were treated with liraglutide (100 μg/kg) or vehicle. At postnatal day 21 and before weaning, 48 males and 45 females (CT and MPFR) were sacrificed. mRNA expression levels of interleukin-1β (IL1β), interleukin-6 (IL-6), nuclear factor-κβ, major histocompatibility complex-II (MHCII), interleukin 10 (IL10), arginase 1 (Arg1), and transforming growth factor (TGFβ) were assessed in the hippocampus by quantitative real-time polymerase chain reaction. Iba1 and GFAP-immunoreactivity were assessed by immunocytochemistry. Results The mRNA expression IL1β, IL6, NF-κB, and MHCII increased in the hippocampus of male but not in female pups from MPFR. In addition, there was an increase in the percentage of GFAP and Iba1-immupositive cells in the dentate gyrus compared to controls, indicating an inflammatory response in the brain. On the other hand, liraglutide treatment prevented the neuroinflammatory process, promoting the production of anti-inflammatory molecules such as IL10, TGFβ, and arginase 1, and decreasing the number and reactivity of microglial cells and astrocytes in the hippocampus of male pups. Conclusion Therefore, the GLP-1 analog, liraglutide, emerges as neuroprotective drug that minimizes the harmful effects of maternal food restriction, decreasing neuroinflammation in the hippocampus in a very early stage.
Collapse
Affiliation(s)
- Y Diz-Chaves
- Laboratory of Endocrinology, Biomedical Research Center (CINBIO), University of Vigo, Campus As Lagoas-Marcosende, E-36310, Vigo (Pontevedra), Spain.
| | - L Toba
- Laboratory of Endocrinology, Biomedical Research Center (CINBIO), University of Vigo, Campus As Lagoas-Marcosende, E-36310, Vigo (Pontevedra), Spain
| | - J Fandiño
- Laboratory of Endocrinology, Biomedical Research Center (CINBIO), University of Vigo, Campus As Lagoas-Marcosende, E-36310, Vigo (Pontevedra), Spain
| | - L C González-Matías
- Laboratory of Endocrinology, Biomedical Research Center (CINBIO), University of Vigo, Campus As Lagoas-Marcosende, E-36310, Vigo (Pontevedra), Spain
| | - L M Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Avenida Doctor Arce 37, E-28002, Madrid, Spain.,Centro de Investigación en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - F Mallo
- Laboratory of Endocrinology, Biomedical Research Center (CINBIO), University of Vigo, Campus As Lagoas-Marcosende, E-36310, Vigo (Pontevedra), Spain
| |
Collapse
|
104
|
Al-Hakeim HK, Twayej AJ, Al- Dujaili AH. Reduction in serum IL-1β, IL-6, and IL-18 levels and Beck Depression Inventory-II score by combined sertraline and ketoprofen administration in major depressive disorder: A clinical trial. ACTA ACUST UNITED AC 2018. [DOI: 10.1016/j.npbr.2018.10.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
105
|
Stampanoni Bassi M, Gilio L, Maffei P, Dolcetti E, Bruno A, Buttari F, Centonze D, Iezzi E. Exploiting the Multifaceted Effects of Cannabinoids on Mood to Boost Their Therapeutic Use Against Anxiety and Depression. Front Mol Neurosci 2018; 11:424. [PMID: 30515077 PMCID: PMC6256035 DOI: 10.3389/fnmol.2018.00424] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 10/31/2018] [Indexed: 12/27/2022] Open
Abstract
The endocannabinoid system (ECS) has been recently recognized as a prominent promoter of the emotional homeostasis, mediating the effects of different environmental signals including rewarding and stressing stimuli. The ECS modulates the rewarding effects of environmental stimuli, influencing synaptic transmission in the dopaminergic projections to the limbic system, and mediates the neurophysiological and behavioral consequences of stress. Notably, the individual psychosocial context is another key element modulating the activity of the ECS. Finally, inflammation represents an additional factor that could alter the cannabinoid signaling in the CNS inducing a "sickness behavior," characterized by anxiety, anhedonia, and depressive symptoms. The complex influences of the ECS on both the environmental and internal stimuli processing, make the cannabinoid-based drugs an appealing option to treat different psychiatric conditions. Although ample experimental evidence shows beneficial effects of ECS modulation on mood, scarce clinical indication limits the use of cannabis-based treatments. To better define the possible clinical indications of cannabinoid-based drugs in psychiatry, a number of issues should be better addressed, including genetic variability and psychosocial factors possibly affecting the individual response. In particular, better knowledge of the multifaceted effects of cannabinoids could help to understand how to boost their therapeutic use in anxiety and depression treatment.
Collapse
Affiliation(s)
| | - Luana Gilio
- Unit of Neurology and Neurorehabilitation, IRCCS Neuromed, Pozzilli, Italy.,Laboratory of Synaptic Immunopathology, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Pierpaolo Maffei
- Unit of Neurology and Neurorehabilitation, IRCCS Neuromed, Pozzilli, Italy
| | - Ettore Dolcetti
- Laboratory of Synaptic Immunopathology, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Antonio Bruno
- Laboratory of Synaptic Immunopathology, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Fabio Buttari
- Unit of Neurology and Neurorehabilitation, IRCCS Neuromed, Pozzilli, Italy
| | - Diego Centonze
- Unit of Neurology and Neurorehabilitation, IRCCS Neuromed, Pozzilli, Italy.,Laboratory of Synaptic Immunopathology, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Ennio Iezzi
- Unit of Neurology and Neurorehabilitation, IRCCS Neuromed, Pozzilli, Italy
| |
Collapse
|
106
|
Krueger JM, Nguyen JT, Dykstra-Aiello CJ, Taishi P. Local sleep. Sleep Med Rev 2018; 43:14-21. [PMID: 30502497 DOI: 10.1016/j.smrv.2018.10.001] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 10/04/2018] [Accepted: 10/08/2018] [Indexed: 12/20/2022]
Abstract
The historic sleep regulatory paradigm invokes "top-down" imposition of sleep on the brain by sleep regulatory circuits. While remaining conceptually useful, many sleep phenomena are difficult to explain using that paradigm, including, unilateral sleep, sleep-walking, and poor performance after sleep deprivation. Further, all animals sleep after non-lethal brain lesions, regardless of whether the lesion includes sleep regulatory circuits, suggesting that sleep is a fundamental property of small viable neuronal/glial networks. That small areas of the brain can exhibit non-rapid eye movement sleep-like states is summarized. Further, sleep-like states in neuronal/glial cultures are described. The local sleep states, whether in vivo or in vitro, share electrophysiological properties and molecular regulatory components with whole animal sleep and exhibit sleep homeostasis. The molecular regulatory components of sleep are also involved in plasticity and inflammation. Like sleep, these processes, are initiated by local cell-activity dependent events, yet have at higher levels of tissue organization whole body functions. While there are large literatures dealing with local initiation and regulation of plasticity and inflammation, the literature surrounding local sleep is in its infancy and clinical applications of the local sleep concept are absent. Regardless, the local use-dependent sleep paradigm can advise and advance future research and clinical applications.
Collapse
Affiliation(s)
- James M Krueger
- Department of Integrative Physiology and Neurobiology, College of Veterinary Medicine, Spokane, WA, USA.
| | - Joseph T Nguyen
- Department of Integrative Physiology and Neurobiology, College of Veterinary Medicine, Spokane, WA, USA
| | - Cheryl J Dykstra-Aiello
- Department of Integrative Physiology and Neurobiology, College of Veterinary Medicine, Spokane, WA, USA
| | - Ping Taishi
- Department of Integrative Physiology and Neurobiology, College of Veterinary Medicine, Spokane, WA, USA
| |
Collapse
|
107
|
α-Tocopherol Ameliorates Redox Equilibrium and Reduces Inflammatory Response Caused by Chronic Variable Stress. BIOMED RESEARCH INTERNATIONAL 2018; 2018:7210783. [PMID: 30533439 PMCID: PMC6250045 DOI: 10.1155/2018/7210783] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 10/20/2018] [Accepted: 10/28/2018] [Indexed: 12/14/2022]
Abstract
Chronic exposure to stress factors contributes to the development of depression by generating excess of reactive oxygen species which leads to oxidative stress and inflammatory processes. The aim of the study was to assess the potential protective properties of α-tocopherol supplementation on the rats exposed to chronic variable stress (CVS). Male Wistar rats (50-55 days old, weighing 200-250 g) were divided into three groups (n=10): control, stressed, and stressed and receiving (+)-α-tocopherol solution in a dose of 100 mg/kg/day. Rats in the stressed groups were exposed to CVS for 40 days. Markers of redox disorders (glutathione reduced and oxidized levels, GSH/GSSG ratio, glutathione peroxidase, glutathione reductase activities, total antioxidant status, and lipid peroxidation) and inflammatory response (IL-1β, IL6, and TNF-α) were determined in the blood. Additionally, molecular biomarkers of depression (expression of Fkbp5 and Tph2) were studied in hippocampus. The biochemical analysis was inconclusive about the presence of oxidative stress in the blood of rats exposed to CVS. However, changes in all parameters suggest presence of redox equilibrium disorders. Similarly, activation of inflammatory processes was observed as a result of CVS. Molecular effects of environmental stress in hippocampus were also observed. Generally, α-tocopherol ameliorated redox equilibrium disorders, tempered inflammatory response, and protected from changes in determined molecular markers of depression.
Collapse
|
108
|
Dooley LN, Kuhlman KR, Robles TF, Eisenberger NI, Craske MG, Bower JE. The role of inflammation in core features of depression: Insights from paradigms using exogenously-induced inflammation. Neurosci Biobehav Rev 2018; 94:219-237. [PMID: 30201219 PMCID: PMC6192535 DOI: 10.1016/j.neubiorev.2018.09.006] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 07/28/2018] [Accepted: 09/06/2018] [Indexed: 12/25/2022]
Abstract
A wealth of evidence has implicated inflammation in the development of depression. Yet, the heterogeneous nature of depression has impeded efforts to understand, prevent, and treat the disease. The purpose of this integrative review is to summarize the connections between inflammation and established core features of depression that exhibit more homogeneity than the syndrome itself: exaggerated reactivity to negative information, altered reward processing, decreased cognitive control, and somatic syndrome. For each core feature, we first provide a brief overview of its relevance to depression and neurobiological underpinnings, and then review evidence investigating a potential role of inflammation. We focus primarily on findings from experimental paradigms of exogenously-induced inflammation. We conclude that inflammation likely plays a role in exaggerated reactivity to negative information, altered reward reactivity, and somatic symptoms. There is less evidence supporting an effect of inflammation on cognitive control as assessed by standard neuropsychological measures. Finally, we discuss implications for future research and recommendationsfor how to test the role of inflammation in the pathogenesis of heterogeneous psychiatric disorders.
Collapse
Affiliation(s)
| | - Kate R Kuhlman
- Department of Psychological Science, University of California Irvine, Irvine, CA 92697, USA; Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience & Human Behavior, University of California Los Angeles, Los Angeles, CA 90095, USA.
| | - Theodore F Robles
- Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience & Human Behavior, University of California Los Angeles, Los Angeles, CA 90095, USA; Department of Psychology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Naomi I Eisenberger
- Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience & Human Behavior, University of California Los Angeles, Los Angeles, CA 90095, USA; Department of Psychology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Michelle G Craske
- Department of Psychology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Julienne E Bower
- Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience & Human Behavior, University of California Los Angeles, Los Angeles, CA 90095, USA; Department of Psychology, University of California Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
109
|
Pandey GN, Rizavi HS, Zhang H, Bhaumik R, Ren X. Abnormal protein and mRNA expression of inflammatory cytokines in the prefrontal cortex of depressed individuals who died by suicide. J Psychiatry Neurosci 2018; 43:376-385. [PMID: 30371993 PMCID: PMC6203549 DOI: 10.1503/jpn.170192] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Depression and stress are major risk factors for suicidal behaviour, and some studies show abnormalities of proinflammatory cytokines in the serum and cerebrospinal fluid (CSF) of depressed and suicidal patients. However, it is not clear if similar abnormalities of cytokines are present in the brain of suicidal and depressed patients. METHODS We therefore determined the mRNA (using realtime polymerase chain reaction) and protein (using enzyme-linked immunosorbent assay and Western Blot) expression levels of interleukin (IL)-1β, IL-6, tumour necrosis factor (TNF)-α, lymphotoxin A, lymphotoxin B, IL-8, IL-10 and IL-13 in the prefrontal cortex (PFC) obtained from 24 depressed individuals who died by suicide and 24 nonpsychiatric controls. RESULTS We observed that the mRNA and protein levels of IL-1β, IL-6, TNF-α, and lymphotoxin A were significantly increased, and levels of anti-inflammatory cytokine IL-10, and of IL-1 receptor antagonist (IL-1RA) were significantly decreased in the PFC of depressed individuals who died by suicide compared with controls. There were no significant differences in the protein and mRNA levels of IL-8 and IL-13 in the PFC. LIMITATIONS The main limitation of this study is that some of the suicide group had been taking antidepressant medication at the time of death. CONCLUSION Our results suggest that alterations of cytokines may be associated with the pathophysiology of depressed suicide and there may be an imbalance between pro- and anti-inflammatory cytokines in people who die by suicide. The causes of these increases in the brain of people who die by suicide, therefore, need to be investigated further.
Collapse
Affiliation(s)
- Ghanshyam N. Pandey
- From the Department of Psychiatry, University of Illinois at Chicago, Chicago, Ill., USA
| | - Hooriyah S. Rizavi
- From the Department of Psychiatry, University of Illinois at Chicago, Chicago, Ill., USA
| | - Hui Zhang
- From the Department of Psychiatry, University of Illinois at Chicago, Chicago, Ill., USA
| | - Runa Bhaumik
- From the Department of Psychiatry, University of Illinois at Chicago, Chicago, Ill., USA
| | - Xinguo Ren
- From the Department of Psychiatry, University of Illinois at Chicago, Chicago, Ill., USA
| |
Collapse
|
110
|
Morrison FG, Miller MW, Wolf EJ, Logue MW, Maniates H, Kwasnik D, Cherry JD, Svirsky S, Restaino A, Hildebrandt A, Aytan N, Stein TD, Alvarez VE, McKee AC, Huber BR. Reduced interleukin 1A gene expression in the dorsolateral prefrontal cortex of individuals with PTSD and depression. Neurosci Lett 2018; 692:204-209. [PMID: 30366016 DOI: 10.1016/j.neulet.2018.10.027] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 09/13/2018] [Accepted: 10/13/2018] [Indexed: 12/13/2022]
Abstract
The inflammatory system has been implicated in the pathophysiology of a variety of psychiatric conditions. Individuals with PTSD, depression, and other fear- and anxiety-related disorders exhibit alterations in peripheral circulating inflammatory markers, suggesting dysregulation of the inflammatory system. The relationship between inflammation and PTSD has been investigated almost exclusively in the periphery, and has not been extensively explored in human postmortem brain tissue. Interleukins (ILs) represent a subtype of cytokines and are key signaling proteins in the immune and inflammatory systems. Based on prior research implicating IL signaling in PTSD and depression, we performed a preliminary investigation of IL gene expression in a region of the cortex involved in emotion regulation and PTSD, the dorsolateral prefrontal cortex (dlPFC), using tissue from the newly established VA National PTSD Brain Bank. Gene expression analyses were conducted on post-mortem tissue from the dlPFC from 50 donors: 13 controls, 12 PTSD cases, and 25 depressed cases. RNA was extracted from frozen dlPFC tissue, reverse transcribed to cDNA, and quantitative polymerase chain reaction (qPCR) was performed to assess gene expression of IL1A, IL1B, IL6, IL8, IL10, IL13, and IL15. We found a multiple-testing corrected significant decrease in IL1A expression in the dlPFC for PTSD and depression cases compared to controls (p < 0.005) with age at death, sex, race and RNA integrity number (RIN) included as covariates. To our knowledge this finding is the first demonstration of altered IL expression in brain tissue from deceased individuals with histories of PTSD and/or depression.
Collapse
Affiliation(s)
- Filomene G Morrison
- National Center for PTSD at VA Boston Healthcare System, United States; Department of Psychiatry, Boston University School of Medicine, United States
| | - Mark W Miller
- National Center for PTSD at VA Boston Healthcare System, United States; Department of Psychiatry, Boston University School of Medicine, United States
| | - Erika J Wolf
- National Center for PTSD at VA Boston Healthcare System, United States; Department of Psychiatry, Boston University School of Medicine, United States
| | - Mark W Logue
- National Center for PTSD at VA Boston Healthcare System, United States; Department of Psychiatry, Boston University School of Medicine, United States; Biomedical Genetics, Boston University School of Medicine, United States; Department of Biostatistics, Boston University School of Public Health, United States
| | - Hannah Maniates
- National Center for PTSD at VA Boston Healthcare System, United States
| | - David Kwasnik
- Department of Psychiatry, Boston University School of Medicine, United States
| | - Jonathan D Cherry
- Department of Neurology, Boston University School of Medicine, United States
| | - Sarah Svirsky
- Pathology and Laboratory Medicine, VA Boston Healthcare System, United States; Department of Neurology, Boston University School of Medicine, United States
| | - Anthony Restaino
- Department of Neurology, Boston University School of Medicine, United States
| | - Audrey Hildebrandt
- National Center for PTSD at VA Boston Healthcare System, United States; Pathology and Laboratory Medicine, VA Boston Healthcare System, United States
| | - Nurgül Aytan
- Department of Neurology, Boston University School of Medicine, United States
| | - Thor D Stein
- Pathology and Laboratory Medicine, VA Boston Healthcare System, United States; Department of Neurology, Boston University School of Medicine, United States; Department of Pathology, Boston University School of Medicine, United States
| | - Victor E Alvarez
- National Center for PTSD at VA Boston Healthcare System, United States; Pathology and Laboratory Medicine, VA Boston Healthcare System, United States; Department of Neurology, Boston University School of Medicine, United States
| | - Ann C McKee
- Pathology and Laboratory Medicine, VA Boston Healthcare System, United States; Department of Neurology, Boston University School of Medicine, United States; Department of Pathology, Boston University School of Medicine, United States
| | | | - Bertrand R Huber
- National Center for PTSD at VA Boston Healthcare System, United States; Pathology and Laboratory Medicine, VA Boston Healthcare System, United States; Department of Neurology, Boston University School of Medicine, United States.
| |
Collapse
|
111
|
Craddock TJA, Michalovicz LT, Kelly KA, Rice MA, Miller DB, Klimas NG, Morris M, O'Callaghan JP, Broderick G. A Logic Model of Neuronal-Glial Interaction Suggests Altered Homeostatic Regulation in the Perpetuation of Neuroinflammation. Front Cell Neurosci 2018; 12:336. [PMID: 30374291 PMCID: PMC6196274 DOI: 10.3389/fncel.2018.00336] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 09/12/2018] [Indexed: 11/21/2022] Open
Abstract
Aberrant inflammatory signaling between neuronal and glial cells can develop into a persistent sickness behavior-related disorders, negatively impacting learning, memory, and neurogenesis. While there is an abundance of literature describing these interactions, there still lacks a comprehensive mathematical model describing the complex feed-forward and feedback mechanisms of neural-glial interaction. Here we compile molecular and cellular signaling information from various studies and reviews in the literature to create a logically-consistent, theoretical model of neural-glial interaction in the brain to explore the role of neuron-glia homeostatic regulation in the perpetuation of neuroinflammation. Logic rules are applied to this connectivity diagram to predict the system's homeostatic behavior. We validate our model predicted homeostatic profiles against RNAseq gene expression profiles in a mouse model of stress primed neuroinflammation. A meta-analysis was used to calculate the significance of similarity between the inflammatory profiles of mice exposed to diisopropyl fluorophostphate (DFP) [with and without prior priming by the glucocorticoid stress hormone corticosterone (CORT)], with the equilibrium states predicted by the model, and to provide estimates of the degree of the neuroinflammatory response. Beyond normal homeostatic regulation, our model predicts an alternate self-perpetuating condition consistent with chronic neuroinflammation. RNAseq gene expression profiles from the cortex of mice exposed to DFP and CORT+DFP align with this predicted state of neuroinflammation, whereas the alignment to CORT alone was negligible. Simulations of putative treatment strategies post-exposure were shown to be theoretically capable of returning the system to a state of typically healthy regulation with broad-acting anti-inflammatory agents showing the highest probability of success. The results support a role for the brain's own homeostatic drive in perpetuating the chronic neuroinflammation associated with exposure to the organophosphate DFP, with and without CORT priming. The deviation of illness profiles from exact model predictions suggests the presence of additional factors or of lasting changes to the brain's regulatory circuitry specific to each exposure.
Collapse
Affiliation(s)
- Travis J A Craddock
- Department of Psychology & Neuroscience, Nova Southeastern University, Ft. Lauderdale, FL, United States.,Institute for Neuro-Immune Medicine, Nova Southeastern University, Ft. Lauderdale, FL, United States.,Department of Computer Science, Nova Southeastern University, Ft. Lauderdale, FL, United States.,Department of Clinical Immunology, Nova Southeastern University, Ft. Lauderdale, FL, United States
| | - Lindsay T Michalovicz
- Health Effects Laboratory Division, Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, Morgantown, WV, United States
| | - Kimberly A Kelly
- Health Effects Laboratory Division, Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, Morgantown, WV, United States
| | - Mark A Rice
- Institute for Neuro-Immune Medicine, Nova Southeastern University, Ft. Lauderdale, FL, United States
| | - Diane B Miller
- Health Effects Laboratory Division, Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, Morgantown, WV, United States
| | - Nancy G Klimas
- Institute for Neuro-Immune Medicine, Nova Southeastern University, Ft. Lauderdale, FL, United States.,Department of Clinical Immunology, Nova Southeastern University, Ft. Lauderdale, FL, United States.,Miami Veterans Affairs Medical Center, Miami, FL, United States
| | - Mariana Morris
- Institute for Neuro-Immune Medicine, Nova Southeastern University, Ft. Lauderdale, FL, United States.,Department of Clinical Immunology, Nova Southeastern University, Ft. Lauderdale, FL, United States
| | - James P O'Callaghan
- Health Effects Laboratory Division, Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, Morgantown, WV, United States
| | - Gordon Broderick
- Department of Psychology & Neuroscience, Nova Southeastern University, Ft. Lauderdale, FL, United States.,Institute for Neuro-Immune Medicine, Nova Southeastern University, Ft. Lauderdale, FL, United States.,Center for Clinical Systems Biology, Rochester General Hospital, Rochester, NY, United States
| |
Collapse
|
112
|
Frank MG, Fonken LK, Dolzani SD, Annis JL, Siebler PH, Schmidt D, Watkins LR, Maier SF, Lowry CA. Immunization with Mycobacterium vaccae induces an anti-inflammatory milieu in the CNS: Attenuation of stress-induced microglial priming, alarmins and anxiety-like behavior. Brain Behav Immun 2018; 73:352-363. [PMID: 29807129 PMCID: PMC6129419 DOI: 10.1016/j.bbi.2018.05.020] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 05/17/2018] [Accepted: 05/24/2018] [Indexed: 12/12/2022] Open
Abstract
Exposure to stressors induces anxiety- and depressive-like behaviors, which are mediated, in part, by neuroinflammatory processes. Recent findings demonstrate that treatment with the immunoregulatory and anti-inflammatory bacterium, Mycobacterium vaccae (M. vaccae), attenuates stress-induced exaggeration of peripheral inflammation and stress-induced anxiety-like behavioral responses. However, the effects of M. vaccae on neuroimmune processes have largely been unexplored. In the present study, we examined the effect of M. vaccae NCTC11659 on neuroimmune regulation, stress-induced neuroinflammatory processes and anxiety-like behavior. Adult male rats were immunized 3× with a heat-killed preparation of M. vaccae (0.1 mg, s.c.) or vehicle. M. vaccae induced an anti-inflammatory immunophenotype in hippocampus (increased interleukin (Il)4, Cd200r1, and Mrc1 mRNA expression) and increased IL4 protein 8 d after the last immunization. Central administration of recombinant IL4 recapitulated the effects of M. vaccae on Cd200r1 and Mrc1 mRNA expression. M. vaccae reduced basal levels of genes (Nlrp3 and Nfkbia) involved in microglial priming; thus, we explored the effects of M. vaccae on stress-induced hippocampal microglial priming and HMGB1, which mediates priming. We found that M. vaccae blocked stress-induced decreases in Cd200r1, increases in the alarmin HMGB1, and priming of the microglial response to immune challenge. Furthermore, M. vaccae prevented stress-induced increases in anxiety-like behavior. The present findings suggest that M. vaccae enhances immunomodulation in the CNS and mitigates the neuroinflammatory and behavioral effects of stress, which may underpin its capacity to impart a stress resilient phenotype.
Collapse
Affiliation(s)
- Matthew G Frank
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Laura K Fonken
- Division of Pharmacology and Toxicology, University of Texas at Austin, Austin, TX 78712, USA
| | - Samuel D Dolzani
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Jessica L Annis
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Philip H Siebler
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Dominic Schmidt
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Linda R Watkins
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Steven F Maier
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Christopher A Lowry
- Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA; Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; Department of Physical Medicine & Rehabilitation, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Center for Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Denver Veterans Affairs Medical Center (VAMC), Denver, CO 80220, USA; Military and Veteran Microbiome Consortium for Research and Education (MVM-CoRE), Denver, CO 80220, USA
| |
Collapse
|
113
|
Fonken LK, Frank MG, Gaudet AD, Maier SF. Stress and aging act through common mechanisms to elicit neuroinflammatory priming. Brain Behav Immun 2018; 73:133-148. [PMID: 30009999 PMCID: PMC6129421 DOI: 10.1016/j.bbi.2018.07.012] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 07/09/2018] [Accepted: 07/12/2018] [Indexed: 01/05/2023] Open
Abstract
Over the course of an animal's lifespan, there is a protracted breakdown in basic homeostatic functions. Stressors (both psychological and physiological) can accelerate this process and compromise multiple homeostatic mechanisms. For example, both stress and aging can modulate neuroinflammatory function and cause a primed phenotype resulting in a heightened neuroinflammatory profile upon immune activation. Microglia, the brain's resident myeloid cell, produce "silent" immune machinery in response to stress and aging that does not cause immediate immune activation; rather, these changes prime the cell for a subsequent immune insult. Primed microglia exhibit a hyperinflammatory response upon immune activation that can exacerbate pathology. In this review, we will explore parallels between stress- and aging-induced neuroinflammatory priming. First, we will provide a background on the basic principles of neuroimmunology. Next, we will discuss evidence that neuroinflammatory responses become primed in the context of both stress and aging. We will also describe cell-specific contributions to neuroinflammatory priming with a focus on microglia. Finally, common mechanisms underlying priming in the context of stress and aging will be discussed: these mechanisms include glucocorticoid signaling; accumulation of danger signals; dis-inhibition of microglia; and breakdown of circadian rhythms. Overall, there are multifarious parallels between stress- and aging-elicited neuroinflammatory priming, suggesting that stress may promote a form of premature aging. Further unravelling mechanisms underlying priming could lead to improved treatments for buffering against stress- and aging-elicited behavioral pathologies.
Collapse
Affiliation(s)
- Laura K. Fonken
- University of Texas at Austin, Division of Pharmacology and Toxicology, Austin, TX 78712 USA;,To whom correspondence should be addressed: Laura K. Fonken, Division of Pharmacology and Toxicology, University of Texas at Austin, 107 W. Dean Keeton, BME 3.510C, Austin, TX 78712 USA.
| | - Matthew G. Frank
- University of Colorado Boulder, Department of Psychology and Neuroscience, Boulder, CO 80309 USA
| | - Andrew D. Gaudet
- University of Colorado Boulder, Department of Psychology and Neuroscience, Boulder, CO 80309 USA
| | - Steven F. Maier
- University of Colorado Boulder, Department of Psychology and Neuroscience, Boulder, CO 80309 USA
| |
Collapse
|
114
|
Iqubal A, Sharma S, Sharma K, Bhavsar A, Hussain I, Iqubal MK, Kumar R. Intranasally administered pitavastatin ameliorates pentylenetetrazol-induced neuroinflammation, oxidative stress and cognitive dysfunction. Life Sci 2018; 211:172-181. [PMID: 30227132 DOI: 10.1016/j.lfs.2018.09.025] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 09/04/2018] [Accepted: 09/13/2018] [Indexed: 12/13/2022]
Abstract
AIM The present study aimed to evaluate the neuroprotective potential of intranasally administered pitavastatin in the PTZ-induced kindling model. MATERIALS AND METHODS Subconvulsant dose of PTZ (35 mg/kg, i.p) was administered on an alternate day until the development of kindling. Behavioural test, biochemical tests and inflammatory cytokines were estimated. Comparative molecular docking study of sodium valproate (VPA) and pitavastatin was performed to predict the binding affinity with GABAA and GABA transaminase. Intranasally administered pitavastatin (0.5 mg/kg and 1 mg/kg) and VPA (200 mg/kg) were used to investigate its protective effect. KEY FINDINGS Comparative in-silico study showed docking score of -4.56 and -2.86 against GABAA receptor whereas -5.56 and -1.86, against GABA transaminase. Root mean square deviation (RMSD) of 0.39A and 0.55A was found for pitavastatin and VPA, respectively. The present study showed the dose-dependent protective effect of intranasally administered pitavastatin and oral VPA against PTZ-induced seizure, cognitive impairment, oxidative stress, and neuroinflammation. SIGNIFICANCE Our findings suggest that the intranasally administered pitavastatin is potential therapeutic approach to managing PTZ-induced kindling and associated comorbid conditions via its antioxidant, anti-inflammatory, and anticonvulsant potential. Further, pitavastatin can modulate GABAA receptor and GABA transaminase enzyme to ameliorate seizure. Meanwhile, more extensive studies are required to establish the molecular mechanism underlying the neuroprotective effect of pitavastatin.
Collapse
Affiliation(s)
- Ashif Iqubal
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Sumit Sharma
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Kalicharan Sharma
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Ashish Bhavsar
- School of Pharmaceutical Science, RGPV, Bhopal MP-462036, India
| | - Ibrahim Hussain
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Mohammad Kashif Iqubal
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Ratendra Kumar
- Om Bioscience and Pharma College, Roorkee-Haridwar, Uttarakhand 249405, India.
| |
Collapse
|
115
|
Caroprese M, Ciliberti M, De Palo P, Santillo A, Sevi A, Albenzio M. Glucocorticoid effects on sheep peripheral blood mononuclear cell proliferation and cytokine production under in vitro hyperthermia. J Dairy Sci 2018; 101:8544-8551. [DOI: 10.3168/jds.2018-14471] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 05/15/2018] [Indexed: 12/28/2022]
|
116
|
Sharif K, Watad A, Coplan L, Lichtbroun B, Krosser A, Lichtbroun M, Bragazzi NL, Amital H, Afek A, Shoenfeld Y. The role of stress in the mosaic of autoimmunity: An overlooked association. Autoimmun Rev 2018; 17:967-983. [PMID: 30118900 DOI: 10.1016/j.autrev.2018.04.005] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 04/04/2018] [Indexed: 12/22/2022]
Abstract
Stress is defined as the pscyophysiological reaction in which the steady state is disturbed or threatened. Stress is not always perceived as a negative response. Stress results when environmental demands exceed an individuals' adaptive capacities. Autoimmune diseases are heterogeneous group of chronic diseases which occur secondary to loss of self antigen tolerance. The etiopathogenesis of autoimmune disease is uncertain. Genetic factors as well as environmental factors appear to interplay, leading to a cascade of events resulting in disease onset. Stress has been postulated to play a role in disease onset in the genetically susceptible patients. During the stress response, catecholamines and glucocorticoids are released from locus coeruleus and adrenal gland. These biomolecules exert control over various immune cells in the innate and adaptive arms of the immune system, thereby altering the cytokine profile released. The increase of IL-4 promotes T-helper 2 (Th2) cell differentiation, while the decrease in IL-12 and the increased IL-10 production reduce the number of T-helper 1 (Th1) cells. The relationship between stress and autoimmune diseases is intricate. Stress has been shown to be associated with disease onset, and disease exacerbations in rheumatoid arthritis, systemic lupus erythematosus, inflammatory bowel disease, multiple sclerosis, Graves' disease as well as other autoimmune conditions. In certain conditions such as psoriasis, stress has been implicated in delaying lesion clearance upon the application of standard treatment regimes. Finally, psychological therapy and cognitive behavioral therapy aimed to reduce stress levels was shown to be effective in influencing better outcomes in many autoimmune diseases. The purpose of this paper is to closer inspect the clinical evidence regarding the role of stress on influencing the various aspects of disease entities.
Collapse
Affiliation(s)
- Kassem Sharif
- Department of Medicine 'B', Israel; Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Abdulla Watad
- Department of Medicine 'B', Israel; Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Louis Coplan
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | | | - Alec Krosser
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | | | - Nicola Luigi Bragazzi
- School of Public Health, Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
| | - Howard Amital
- Department of Medicine 'B', Israel; Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Arnon Afek
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Incumbent of the Laura Schwarz-Kipp Chair for Research of Autoimmune Diseases, Tel-Aviv University, Head of The Mosaic of Autoimmunity Project, Saint Petersburg State University, Israel; Head of The Mosaic of Autoimmunity Project, Saint Petersburg State University, Russia.
| |
Collapse
|
117
|
Lisboa SF, Niraula A, Resstel LB, Guimaraes FS, Godbout JP, Sheridan JF. Repeated social defeat-induced neuroinflammation, anxiety-like behavior and resistance to fear extinction were attenuated by the cannabinoid receptor agonist WIN55,212-2. Neuropsychopharmacology 2018; 43:1924-1933. [PMID: 29786066 PMCID: PMC6046035 DOI: 10.1038/s41386-018-0064-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 03/19/2018] [Accepted: 04/02/2018] [Indexed: 11/10/2022]
Abstract
Psychosocial stress contributes to the development of psychiatric disorders. Repeated social defeat (RSD) is a murine stressor that causes a release of inflammatory monocytes into circulation. Moreover, RSD-induced anxiety-like behavior is dependent on the recruitment of these monocytes to the brain. Activation of the endocannabinoid (ECB) system may modulate both neuroendocrine and inflammatory responses mediated by stress. Therefore, we hypothesized that a cannabinoid receptor agonist would attenuate RSD-induced inflammation, anxiety, and stress sensitization. To test this hypothesis, mice received an injection of the synthetic cannabinoid1/2 receptor agonist, WIN55,212-2 (WIN; 1 mg/kg, intraperitoneally) daily for six consecutive days, 30 min before each exposure to RSD. Anxiety-like behavior, immune activation, neuroinflammation, and microglial reactivity were determined 14 h after RSD. RSD-induced anxiety-like behavior in the open field and in the EPM was reversed by WIN55,212-2. Moreover, WIN55,212-2 reduced the accumulation of inflammatory monocytes in circulation and brain after RSD and attenuated RSD-induced interleukin-1β (IL-1β) messenger RNA (mRNA) expression in microglia/macrophages. Increased ex vivo reactivity of microglia/monocytes to lipopolysaccharides (LPS) after RSD was also attenuated by WIN55,212-2. Next, fear expression, extinction, and recall were evaluated 24 and 48 h, respectively, after contextual fear conditioning, which took place 7 days after RSD. Here, RSD caused prolonged fear expression and impaired fear extinction recall, which was associated with increased IL-1β mRNA in the brain. Moreover, these stress-induced effects were reversed by WIN55,212-2. In conclusion, activation of cannabinoid receptors limited the immune and neuroinflammatory responses to RSD and reversed the short-term and long-term behavioral deficits associated with RSD.
Collapse
Affiliation(s)
- Sabrina Francesca Lisboa
- Department of Pharmacology, University of Sao Paulo (USP), Ribeirão Preto, São Paulo, 14049900, Brazil. .,Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), Medical School of Ribeirão Preto, University of Sao Paulo (USP), Ribeirão Preto, São Paulo, 14049900, Brazil.
| | - Anzela Niraula
- 0000 0001 2285 7943grid.261331.4Division of Biosciences, Ohio State University, Columbus, OH 43210 USA
| | - Leonardo Barbosa Resstel
- 0000 0004 1937 0722grid.11899.38Department of Pharmacology, University of Sao Paulo (USP), Ribeirão Preto, São Paulo 14049900 Brazil ,0000 0004 1937 0722grid.11899.38Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), Medical School of Ribeirão Preto, University of Sao Paulo (USP), Ribeirão Preto, São Paulo 14049900 Brazil
| | - Francisco Silveira Guimaraes
- 0000 0004 1937 0722grid.11899.38Department of Pharmacology, University of Sao Paulo (USP), Ribeirão Preto, São Paulo 14049900 Brazil ,0000 0004 1937 0722grid.11899.38Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), Medical School of Ribeirão Preto, University of Sao Paulo (USP), Ribeirão Preto, São Paulo 14049900 Brazil
| | - Jonathan P. Godbout
- 0000 0001 2285 7943grid.261331.4Division of Biosciences, Ohio State University, Columbus, OH 43210 USA ,0000 0001 2285 7943grid.261331.4Department of Neuroscience, Ohio State University, Columbus, OH 43210 USA ,0000 0001 2285 7943grid.261331.4Center for Brain and Spinal Cord Repair, Ohio State University, Columbus, OH 43210 USA
| | - John F. Sheridan
- 0000 0001 2285 7943grid.261331.4Division of Biosciences, Ohio State University, Columbus, OH 43210 USA ,0000 0001 2285 7943grid.261331.4Department of Neuroscience, Ohio State University, Columbus, OH 43210 USA ,0000 0001 2285 7943grid.261331.4Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH 43210 USA
| |
Collapse
|
118
|
Clark SM, Song C, Li X, Keegan AD, Tonelli LH. CD8 + T cells promote cytokine responses to stress. Cytokine 2018; 113:256-264. [PMID: 30033139 DOI: 10.1016/j.cyto.2018.07.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 06/19/2018] [Accepted: 07/12/2018] [Indexed: 11/27/2022]
Abstract
Psychological stress is known to have profound effects on immune function and to promote inflammatory conditions. Elevated circulating levels of cytokines associated with stress are known to increase the risk to several diseases, but little is known about this mechanism. This study assessed the role of T cells on cytokine levels after exposure to stress in the learned helplessness paradigm. Adoptive transfer of CD4+ T cells into Rag2-/- mice did not change cytokine levels to stress while CD8+ T cells resulted in an increase in TNF-α, IL-6 and IFN-γ in stressed Rag2-/- mice. Moreover, depletion of CD8+ T cells in WT mice abolished these cytokine responses to stress. Corticosterone and behavioral stress responsiveness was impaired in Rag2-/- mice reconstituted with CD8+ T cells. Notably, depletion of these cells in WT mice had no effect on behavior or corticosterone levels. Exposure to stress did not change the expression of canonical markers of T cell activation including CD62L and CD44 or modified intracellular cytokine content, suggesting that they are not the main producers of circulating cytokines in response to stress. These results show that CD8+ T cells promote TNF-α, IL-6 and IFN-γ responses to stress, possibly by stimulating non-lymphoid cells.
Collapse
Affiliation(s)
- Sarah M Clark
- Laboratory of Behavioral Neuroimmunology, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA; Research and Development Service, Department of Veterans Affairs, VA Maryland Health Care System, Baltimore, MD, USA
| | - Chang Song
- Laboratory of Behavioral Neuroimmunology, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Xin Li
- Laboratory of Behavioral Neuroimmunology, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Achsah D Keegan
- Center for Vascular and Inflammatory Diseases, Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA; Research and Development Service, Department of Veterans Affairs, VA Maryland Health Care System, Baltimore, MD, USA
| | - Leonardo H Tonelli
- Laboratory of Behavioral Neuroimmunology, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA; Research and Development Service, Department of Veterans Affairs, VA Maryland Health Care System, Baltimore, MD, USA.
| |
Collapse
|
119
|
Jones ME, Paniccia JE, Lebonville CL, Reissner KJ, Lysle DT. Chemogenetic Manipulation of Dorsal Hippocampal Astrocytes Protects Against the Development of Stress-enhanced Fear Learning. Neuroscience 2018; 388:45-56. [PMID: 30030056 DOI: 10.1016/j.neuroscience.2018.07.015] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 06/28/2018] [Accepted: 07/07/2018] [Indexed: 11/18/2022]
Abstract
Maladaptive behavioral outcomes following stress have been associated with immune dysregulation. For example, we have previously reported that stress-induced dorsal hippocampal interleukin-1β signaling is critical to the development of stress-enhanced fear learning (SEFL). In parallel, astroglial signaling has been linked to the development of post-traumatic stress disorder (PTSD)-like phenotypes and our most recent studies have revealed astrocytes as the predominant cellular source of stress-induced IL-1β. Here, we used chemogenetic technology and morphological analyses to further explore dorsal hippocampal astrocyte function in the context of SEFL. Using a glial-expressing DREADD construct (AAV8-GFAP-hM4Di(Gi)-mCherry), we show that dorsal hippocampal astroglial Gi activation is sufficient to attenuate SEFL. Furthermore, our data provide the first initial evidence to support the function of the glial-DREADD construct employed. Specifically, we find that CNO (clozapine-n-oxide) significantly attenuated colocalization of the Gi-coupled DREADD receptor and cyclic adenosine monophosphate (cAMP), indicating functional inhibition of cAMP production. Subsequent experiments examined dorsal hippocampal astrocyte volume, surface area, and synaptic contacts (colocalization with postsynaptic density 95 (PSD95)) following exposure to severe stress (capable of inducing SEFL). While severe stress did not alter dorsal hippocampal astrocyte volume or surface area, the severe stressor exposure reduced dorsal hippocampal PSD95 immunoreactivity and the colocalization analysis showed reduced PSD95 colocalized with astrocytes. Collectively, these data provide evidence to support the functional efficacy of the glial-expressing DREADD employed, and suggest that an astrocyte-specific manipulation, activation of astroglial Gi signaling, is sufficient to protect against the development of SEFL, a PTSD-like behavior.
Collapse
Affiliation(s)
- Meghan E Jones
- Department of Psychology and Neuroscience, Behavioral and Integrative Neuroscience Program, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Jacqueline E Paniccia
- Department of Psychology and Neuroscience, Behavioral and Integrative Neuroscience Program, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Christina L Lebonville
- Department of Psychology and Neuroscience, Behavioral and Integrative Neuroscience Program, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Kathryn J Reissner
- Department of Psychology and Neuroscience, Behavioral and Integrative Neuroscience Program, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Donald T Lysle
- Department of Psychology and Neuroscience, Behavioral and Integrative Neuroscience Program, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.
| |
Collapse
|
120
|
Swanson A, Wolf T, Sitzmann A, Willette AA. Neuroinflammation in Alzheimer's disease: Pleiotropic roles for cytokines and neuronal pentraxins. Behav Brain Res 2018; 347:49-56. [PMID: 29462653 PMCID: PMC5988985 DOI: 10.1016/j.bbr.2018.02.015] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 01/30/2018] [Accepted: 02/13/2018] [Indexed: 12/14/2022]
Abstract
Neuroinflammation is a potential factor speculated to underlie Alzheimer's disease (AD) etiopathogenesis and progression. The overwhelming focus in this area of research to date has been on the chronic upregulation of pro-inflammatory cytokines to understand how neuroinflammatory mechanisms contribute to neurodegeneration. Yet, it is important to understand the pleiotropic roles of these cytokines in modulating neuroinflammation in which they cannot be labeled as a strictly "good" or "bad" biomarker phenotype. As such, biomarkers with more precise functions are needed to better understand how neuroinflammation impacts the brain in AD. Neuronal pentraxins are a concentration- dependent group of pro- or anti- inflammatory cytokines. There is contradictory evidence of these pentraxins as being both neuroprotective and potentially detrimental in AD. Potential neuroprotective examples include their ability to predict AD-related outcomes such as cognition, memory function and synaptic refinement. This review will briefly outline the basis of AD and subsequently summarize findings for neuropathological mechanisms of neuroinflammation, roles for traditional pro-and anti-inflammatory cytokines, and data found thus far on the neuronal pentraxins.
Collapse
Affiliation(s)
- Ashley Swanson
- Department of Food Science and Human Nutrition, Iowa State University, 2312 Food Sciences Building, 536 Farm House Lane, Ames, IA 50011, United States.
| | - Tovah Wolf
- Department of Food Science and Human Nutrition, Iowa State University, 2312 Food Sciences Building, 536 Farm House Lane, Ames, IA 50011, United States.
| | - Alli Sitzmann
- Department of Psychology, Iowa State University, W112 Lagomarcino Hall, 901 Stange Road, Ames, IA 50011, United States.
| | - Auriel A Willette
- Department of Food Science and Human Nutrition, Iowa State University, 2312 Food Sciences Building, 536 Farm House Lane, Ames, IA 50011, United States; Department of Psychology, Iowa State University, W112 Lagomarcino Hall, 901 Stange Road, Ames, IA 50011, United States; Department of Biomedical Sciences, Iowa State University, 2008 Veterinary Medicine, Ames, IA 50011, United States; Department of Neurology, University of Iowa, 2007 Roy Carver Pavilion, 200 Hawkins Drive, Iowa City, IA 52242, United States.
| |
Collapse
|
121
|
Magnetic resonance imaging evidence of hippocampal structural changes in patients with primary biliary cholangitis. Clin Transl Gastroenterol 2018; 9:169. [PMID: 29977030 PMCID: PMC6033882 DOI: 10.1038/s41424-018-0038-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 05/11/2018] [Accepted: 06/07/2018] [Indexed: 12/13/2022] Open
Abstract
Introduction Behavioral symptoms are commonly reported by patients with primary biliary cholangitis (PBC). In other patient populations, symptoms are commonly associated with hippocampal volume reduction linked to neuroinflammation (inferred from regional iron deposition), as demonstrated by magnetic resonance imaging (MRI). We hypothesized that PBC patients would exhibit reduced volume and increased iron deposition of the hippocampus. Methods Seventeen female non-cirrhotic PBC patients and 17 age/gender-matched controls underwent 3-Tesla T1-weighted MRI and quantitative susceptibility mapping (QSM; an indicator of iron deposition). The hippocampus and its subfields were segmented from T1 images using Freesurfer, and susceptibility of the whole hippocampus was calculated from QSM images. Volume and susceptibility were compared between groups, and associations with PBC-40 score and disease indicators (years since diagnosis, Fibroscan value, alkaline phosphatase level, clinical response to ursodeoxycholic acid (UDCA)) were investigated. Results PBC patients exhibited significantly reduced hippocampal volume (p = 0.023) and increased susceptibility (p = 0.048). Subfield volumes were reduced for the subiculum, molecular layer, granule cell layer of the dentate gyrus and CA4 (p < 0.05). Fibroscan value was significantly correlated with PBC-40 (Spearman’s rho = 0.499; p = 0.041) and disease duration (Spearman’s rho = 0.568; p = 0.017). Discussion Our findings suggest hippocampal changes occur early in the disease course of PBC, similar in magnitude to those observed in major depressive disorder and neurodegenerative diseases. Translational impact Clinical management of PBC could include early interventional strategies that promote hippocampal neurogenesis that may beneficially impact behavioral symptoms and improve quality of life.
Collapse
|
122
|
Antidepressant Effect of Fraxinus rhynchophylla Hance Extract in a Mouse Model of Chronic Stress-Induced Depression. BIOMED RESEARCH INTERNATIONAL 2018; 2018:8249563. [PMID: 30065945 PMCID: PMC6051329 DOI: 10.1155/2018/8249563] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 05/11/2018] [Accepted: 05/21/2018] [Indexed: 12/29/2022]
Abstract
Prolonged exposure to stress can affect mood and cognition and lead to mood disorders. Research on stress-associated mood disorders is important in modern society as people are increasingly exposed to unavoidable stressors. We used a mouse model with 2 weeks of exposure to electric foot shock and restraint, to determine the effect of Fraxinus rhynchophylla Hance (FX) extract on chronic stress-induced depression. We measured the effect of FX extract using various physiological, behavioral, and biochemical measures. FX extract ameliorated chronic stress-induced body and relative liver weight loss and improved depressive-like behaviors in the open field and forced swim tests. In addition, plasma cortisol and serotonin levels in stress-induced mice following FX treatment were similar to normal mice, and the elevation of proinflammatory cytokines was prevented. Moreover, FX treatment increased the expression of phosphorylated cyclic adenosine-3′,5′-monophosphate response element-binding protein (pCREB)/brain-derived neurotrophic factor (BDNF). Further experiments confirmed the efficacy of FX extract by showing similar results using esculin and esculetin, compounds extracted from FX. Taken together, these results indicate that FX extract has an antidepressant effect on chronic stress-induced depression by associating signaling with neuroinflammation and neurogenesis.
Collapse
|
123
|
Lasselin J, Lekander M, Axelsson J, Karshikoff B. Sex differences in how inflammation affects behavior: What we can learn from experimental inflammatory models in humans. Front Neuroendocrinol 2018; 50:91-106. [PMID: 29935190 DOI: 10.1016/j.yfrne.2018.06.005] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 05/29/2018] [Accepted: 06/19/2018] [Indexed: 12/12/2022]
Abstract
Human models demonstrate that experimental activation of the innate immune system has profound effects on brain activation and behavior, inducing fatigue, worsened mood and pain sensitivity. It has been proposed that inflammation is a mechanism involved in the etiology and maintenance of depression, chronic pain and long-term fatigue. These diseases show a strong female overrepresentation, suggesting that a better understanding of sex differences in how inflammation drives behavior could help the development of individualized treatment interventions. For this purpose, we here review sex differences in studies using experimental inflammatory models to investigate changes in brain activity and behavior. We suggest a model in which inflammation accentuates sex differences in brain networks and pre-existing vulnerability factors. This effect could render women more vulnerable to the detrimental effects of immune-to-brain communication over time. We call for systematic and large scale investigations of vulnerability factors for women in the behavioral response to inflammation.
Collapse
Affiliation(s)
- Julie Lasselin
- Stress Research Institute, Stockholm University, Stockholm, Sweden; Division of Psychology, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; Institute of Medical Psychology and Behavioral Immunobiology, University Hospital Essen, Essen, Germany
| | - Mats Lekander
- Stress Research Institute, Stockholm University, Stockholm, Sweden; Division of Psychology, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; Osher Center for Integrative Medicine, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - John Axelsson
- Stress Research Institute, Stockholm University, Stockholm, Sweden; Division of Psychology, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Bianka Karshikoff
- Stress Research Institute, Stockholm University, Stockholm, Sweden; Division of Psychology, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; Osher Center for Integrative Medicine, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; Division of Pain Medicine, Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Palo Alto, USA.
| |
Collapse
|
124
|
Roerink ME, Roerink SHPP, Skoluda N, van der Schaaf ME, Hermus ARMM, van der Meer JWM, Knoop H, Nater UM. Hair and salivary cortisol in a cohort of women with chronic fatigue syndrome. Horm Behav 2018; 103:1-6. [PMID: 29807037 DOI: 10.1016/j.yhbeh.2018.05.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 05/19/2018] [Accepted: 05/22/2018] [Indexed: 01/21/2023]
Abstract
Hypocortisolism has been found in CFS patients in blood, urine, and saliva. It is unclear if hypocortisolism can also be demonstrated using long-term cortisol measurements, such as cortisol in hair. In addition, the interaction between the HPA axis and the immune system, both expected to play an important role in CFS, is unclear. The objective of the current study was to compare hair and salivary cortisol concentrations in a cohort of female CFS patients to those in healthy controls, and to test the effect of an interleukin-1 receptor antagonist (anakinra) on the HPA axis. Salivary cortisol concentrations of 107 CFS patients were compared to 59 healthy controls, with CFS patients showing a decreased cortisol awakening response (4.2 nmol/L ± 5.4 vs 6.1 nmol/L ± 6.3, p = 0.036). Total cortisol output during the day did not differ significantly in saliva, but there was a trend to lower hair cortisol in a subset of 46 patients compared to 46 controls (3.8 pg/mg ± 2.1 vs 4.3 pg/mg ± 1.8, p = 0.062). After four weeks of treatment with either daily anakinra (100 mg/day) or placebo, there was a slight decrease of hair cortisol concentrations in the anakinra group compared to an increase in the placebo group (p = 0.022). This study confirms the altered dynamics of the HPA axis in a group of CFS patients, and for the first time shows that this might also be present for long-term cortisol measures.
Collapse
Affiliation(s)
- Megan E Roerink
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands.
| | - Sean H P P Roerink
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Nadine Skoluda
- Department of Psychology, University of Vienna, Vienna, Austria
| | - Marieke E van der Schaaf
- The Donders Institute for Brain, Cognition and Behavior, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Ad R M M Hermus
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jos W M van der Meer
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Hans Knoop
- Department of Medical Psychology, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Urs M Nater
- Department of Psychology, University of Vienna, Vienna, Austria
| |
Collapse
|
125
|
Martínez AD, Ruelas L, Granger DA. Household fear of deportation in relation to chronic stressors and salivary proinflammatory cytokines in Mexican-origin families post-SB 1070. SSM Popul Health 2018; 5:188-200. [PMID: 30073186 PMCID: PMC6068082 DOI: 10.1016/j.ssmph.2018.06.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 05/29/2018] [Accepted: 06/13/2018] [Indexed: 02/07/2023] Open
Abstract
Sociologists recognize that immigration enforcement policies are forms of institutionalized racism that can produce adverse health effects in both undocumented and documented Latinos and Mexican-origin persons in the United States. Despite this important advancement, little research examines the relationship between fear of immigration enforcement and biobehavioral health in mixed-status Mexican-origin families. This study applies an embodiment of racism approach to examine how household fear of deportation (FOD) is related to differences in salivary proinflammatory cytokines (IL-1 β , IL-6, IL-8, and TNF α ) in healthy Mexican-origin families with at least one immigrant, living in Phoenix, AZ. Participants were 111 individuals (n=46 adults, 72% female; n=65 children, 49% female) from 30 low-income, mixed-status families. During a home visit, anthropometric measures and saliva were collected from each family member and a household survey was administered. Saliva was assayed for salivary IL-1 β , IL-6, IL-8, and TNF α . Random effects multilevel structural equation models estimated the relationship between household FOD and a salivary proinflammatory cytokine latent variable between families, while controlling for other chronic stressors (economic/occupational, immigration, parental, and family conflict). Household FOD ( β =0.68, p=0.04) and family conflict chronic stress ( β =1.96, p=0.03) were strongly related to elevated levels of proinflammatory cytokines between families. These results were consistent in non-mixed and mixed-status families. Future research is needed to characterize what aspects of living with an undocumented family member shape the physical health outcomes of persons with authorized status or US-citizenship.
Collapse
Affiliation(s)
- Airín D Martínez
- Yale University, The Ethnicity, Race and Migration & American Studies, 35 Broadway, Room 210, New Haven, CT 06510, USA.,University of Massachusetts-Amherst, School of Public Health & Health Sciences, Department of Health Promotion & Policy, USA
| | - Lillian Ruelas
- Arizona State University Admissions Services Tempe, AZ. 85281, USA
| | - Douglas A Granger
- Institute for Interdisciplinary Salivary Bioscience Research, University of California, Irvine, Irvine, CA, USA.,The Johns Hopkins University Bloomberg School of Public Health, School of Nursing, and School of Medicine, Baltimore, MD, USA.,Saliva Bioscience Laboratory, University of Nebraska, Lincoln, NE, USA
| |
Collapse
|
126
|
Cao S, Zhang Q, Wang C, Wu H, Jiao L, Hong Q, Hu C. LPS challenge increased intestinal permeability, disrupted mitochondrial function and triggered mitophagy of piglets. Innate Immun 2018; 24:221-230. [PMID: 29642727 PMCID: PMC6830921 DOI: 10.1177/1753425918769372] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 03/07/2018] [Accepted: 03/15/2018] [Indexed: 12/21/2022] Open
Abstract
Here we investigated the influence of LPS-induced gut injury on antioxidant homeostasis, mitochondrial (mt) function and the level of mitophagy in piglets. The results showed that LPS-induced intestinal injury decreased the transepithelial electrical resistance, increased the paracellular permeability of F1TC dextran 4 kDa, and decreased the expression of claudin-1, occludin and zonula occludens-1 in the jejunum compared with the control group. LPS decreased the activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px), and increased the content of malondialdehyde in the jejunum. Meanwhile, the expression of SOD-related genes ( Cu/Zn-SOD, Mn-SOD) and GSH-Px-related genes ( GPX-1, GPX-4) declined in LPS-challenged pigs compared with the control. LPS also increased TNF-α, IL-6, IL-8 and IL-1β mRNA expression. LPS induced mt dysfunction, as demonstrated by increased reactive oxygen species production and decreased membrane potential of intestinal mitochondria, intestinal content of mt DNA and activities of the intestinal mt respiratory chain. Furthermore, LPS induced an increase in expression of mitophagy related proteins, PTEN-induced putative kinase (PINK1) and Parkin in the intestinal mitochondria, as well as an enhancement of the ratio of light chain 3-II (LC3-II) to LC3-I content in the jejunal mucosa. These results suggested that LPS-induced intestinal injury accompanied by disrupted antioxidant homeostasis, caused mt dysfunction and triggered mitophagy.
Collapse
Affiliation(s)
- Shuting Cao
- Animal Science College, Zhejiang University, The Key
Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou,
China
| | - Qianhui Zhang
- Animal Science College, Zhejiang University, The Key
Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou,
China
| | - ChunChun Wang
- Animal Science College, Zhejiang University, The Key
Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou,
China
| | - Huan Wu
- Animal Science College, Zhejiang University, The Key
Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou,
China
| | - Lefei Jiao
- Animal Science College, Zhejiang University, The Key
Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou,
China
| | - Qihua Hong
- Animal Science College, Zhejiang University, The Key
Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou,
China
| | - Caihong Hu
- Animal Science College, Zhejiang University, The Key
Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou,
China
| |
Collapse
|
127
|
Vezzani A, Dingledine R, Rossetti AO. Immunity and inflammation in status epilepticus and its sequelae: possibilities for therapeutic application. Expert Rev Neurother 2018; 15:1081-92. [PMID: 26312647 DOI: 10.1586/14737175.2015.1079130] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Status epilepticus (SE) is a life-threatening neurological emergency often refractory to available treatment options. It is a very heterogeneous condition in terms of clinical presentation and causes, which besides genetic, vascular and other structural causes also include CNS or severe systemic infections, sudden withdrawal from benzodiazepines or anticonvulsants and rare autoimmune etiologies. Treatment of SE is essentially based on expert opinions and antiepileptic drug treatment per se seems to have no major impact on prognosis. There is, therefore, urgent need of novel therapies that rely upon a better understanding of the basic mechanisms underlying this clinical condition. Accumulating evidence in animal models highlights that inflammation ensuing in the brain during SE may play a determinant role in ongoing seizures and their long-term detrimental consequences, independent of an infection or auto-immune cause; this evidence encourages reconsideration of the treatment flow in SE patients.
Collapse
Affiliation(s)
- Annamaria Vezzani
- a 1 Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Milano, Italy
| | | | | |
Collapse
|
128
|
Xiao X, Putatunda R, Zhang Y, Soni PV, Li F, Zhang T, Xin M, Luo JJ, Bethea JR, Cheng Y, Hu W. Lymphotoxin β receptor-mediated NFκB signaling promotes glial lineage differentiation and inhibits neuronal lineage differentiation in mouse brain neural stem/progenitor cells. J Neuroinflammation 2018; 15:49. [PMID: 29463313 PMCID: PMC5819232 DOI: 10.1186/s12974-018-1074-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 01/22/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Lymphotoxin (LT) is a lymphokine mainly expressed in lymphocytes. LTα binds one or two membrane-associated LTβ to form LTα2β1 or LTα1β2 heterotrimers. The predominant LTα1β2 binds to LTβ receptor (LTβR) primarily expressed in epithelial and stromal cells. Most studies on LTβR signaling have focused on the organization, development, and maintenance of lymphoid tissues. However, the roles of LTβR signaling in the nervous system, particularly in neurogenesis, remain unknown. Here, we investigated the role of LTβR-mediated NFκB signaling in regulating neural lineage differentiation. METHODS The C57BL/6J wild-type and GFAP-dnIκBα transgenic mice were used. Serum-free embryoid bodies were cultured from mouse embryonic stem cells and further induced into neural stem/progenitor cells (NSCs/NPCs). Primary neurospheres were cultured from embryonic and adult mouse brains followed by monolayer culture for amplification/passage. NFκB activation was determined by adenovirus-mediated NFκB-firefly-luciferase reporter assay and p65/RelB/p52 nuclear translocation assay. LTβR mRNA expression was evaluated by quantitative RT-PCR and LTβR protein expression was determined by immunohistochemistry and Western blot analysis. Multilabeled immunocytochemistry or immunohistochemistry followed by fluorescent confocal microscopy and quantitative analysis of neural lineage differentiation were performed. Graphing and statistical analysis were performed with GraphPad Prism software. RESULTS In cultured NSCs/NPCs, LTα1β2 stimulation induced an activation of classical and non-classical NFκB signaling. The expression of LTβR-like immunoreactivity in GFAP+/Sox2+ NSCs was identified in well-established neurogenic zones of adult mouse brain. Quantitative RT-PCR and Western blot analysis validated the expression of LTβR in cultured NSCs/NPCs and brain neurogenic regions. LTβR expression was significantly increased during neural induction. LTα1β2 stimulation in cultured NSCs/NPCs promoted astroglial and oligodendrocytic lineage differentiation, but inhibited neuronal lineage differentiation. Astroglial NFκB inactivation in GFAP-dnIκBα transgenic mice rescued LTβR-mediated abnormal phenotypes of cultured NSCs/NPCs. CONCLUSION This study provides the first evidence for the expression and function of LTβR signaling in NSCs/NPCs. Activation of LTβR signaling promotes glial lineage differentiation. Our results suggest that neurogenesis is regulated by the adaptive immunity and inflammatory responses.
Collapse
Affiliation(s)
- Xiao Xiao
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
- Center for Metabolic Disease Research, Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, 3500 N Broad Street, Philadelphia, PA, 19140, USA
| | - Raj Putatunda
- Center for Metabolic Disease Research, Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, 3500 N Broad Street, Philadelphia, PA, 19140, USA
| | - Yonggang Zhang
- Center for Metabolic Disease Research, Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, 3500 N Broad Street, Philadelphia, PA, 19140, USA
| | - Priya V Soni
- Center for Metabolic Disease Research, Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, 3500 N Broad Street, Philadelphia, PA, 19140, USA
| | - Fang Li
- Center for Metabolic Disease Research, Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, 3500 N Broad Street, Philadelphia, PA, 19140, USA
| | - Ting Zhang
- Center for Metabolic Disease Research, Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, 3500 N Broad Street, Philadelphia, PA, 19140, USA
| | - Mingyang Xin
- Center for Metabolic Disease Research, Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, 3500 N Broad Street, Philadelphia, PA, 19140, USA
| | - Jin Jun Luo
- Department of Neurology, Temple University Lewis Katz School of Medicine, 3401 N Broad Street, Philadelphia, PA, USA
| | - John R Bethea
- Department of Biology, Drexel University, Philadelphia, PA, USA
| | - Yuan Cheng
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China.
| | - Wenhui Hu
- Center for Metabolic Disease Research, Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, 3500 N Broad Street, Philadelphia, PA, 19140, USA.
| |
Collapse
|
129
|
La Fratta I, Tatangelo R, Campagna G, Rizzuto A, Franceschelli S, Ferrone A, Patruno A, Speranza L, De Lutiis MA, Felaco M, Grilli A, Pesce M. The plasmatic and salivary levels of IL-1β, IL-18 and IL-6 are associated to emotional difference during stress in young male. Sci Rep 2018; 8:3031. [PMID: 29445205 PMCID: PMC5813044 DOI: 10.1038/s41598-018-21474-y] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 02/04/2018] [Indexed: 12/14/2022] Open
Abstract
Saliva collection is considered a non-invasive method to detect inflammatory markers in response to emotional states within natural social contexts. Numerous studies have prompted an important role of cytokines in modulating distinct aspects of social and emotional behavior. The aim of this study was to investigate the reliability of plasma and saliva as investigative tools for measure some inflammatory marker levels (CRP, IL-1β, IL-18, and IL-6). At the same time, the relationships between these markers and emotional states in response to a socio-cognitive stress (Academic Exam, AE), were considered. It was demonstrated that the plasma and saliva concentrations of all immune-mediators analyzed were significantly related across the socio-cognitive stress. In addition, when there was a close correlation to AE, the anger state, the IL-1β, the IL-18 salivary and plasmatic concentrations were significantly higher, while they decreased during the AE. On the other hand, the anxiety state and the IL-6 levels significantly increased throughout the AE. The IL-1β and IL-6 were positively associated to the anger and the anxiety state, respectively. In conclusion, our data highlight that different immune markers are similarly detectable in plasma and saliva during socio-cognitive stress. Also, they could be related to different emotional responses.
Collapse
Affiliation(s)
- I La Fratta
- Medicine and Health Science School, Via dei Vestini, 31-66100, Chieti, Italy
| | - R Tatangelo
- Medicine and Health Science School, Via dei Vestini, 31-66100, Chieti, Italy
| | - G Campagna
- Medicine and Health Science School, Via dei Vestini, 31-66100, Chieti, Italy
| | - A Rizzuto
- Medicine and Health Science School, Via dei Vestini, 31-66100, Chieti, Italy
| | - S Franceschelli
- Medicine and Health Science School, Via dei Vestini, 31-66100, Chieti, Italy
| | - A Ferrone
- Medicine and Health Science School, Via dei Vestini, 31-66100, Chieti, Italy
| | - A Patruno
- Medicine and Health Science School, Via dei Vestini, 31-66100, Chieti, Italy
| | - L Speranza
- Medicine and Health Science School, Via dei Vestini, 31-66100, Chieti, Italy
| | - M A De Lutiis
- Medicine and Health Science School, Via dei Vestini, 31-66100, Chieti, Italy
| | - M Felaco
- Medicine and Health Science School, Via dei Vestini, 31-66100, Chieti, Italy
| | - A Grilli
- Medicine and Health Science School, Via dei Vestini, 31-66100, Chieti, Italy
| | - M Pesce
- Medicine and Health Science School, Via dei Vestini, 31-66100, Chieti, Italy.
| |
Collapse
|
130
|
Vinkler M, Leon AE, Kirkpatrick L, Dalloul RA, Hawley DM. Differing House Finch Cytokine Expression Responses to Original and Evolved Isolates of Mycoplasma gallisepticum. Front Immunol 2018. [PMID: 29403495 DOI: 10.3389/fimmu.2018.00013/full] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The recent emergence of the poultry bacterial pathogen Mycoplasma gallisepticum (MG) in free-living house finches (Haemorhous mexicanus), which causes mycoplasmal conjunctivitis in this passerine bird species, resulted in a rapid coevolutionary arms-race between MG and its novel avian host. Despite extensive research on the ecological and evolutionary dynamics of this host-pathogen system over the past two decades, the immunological responses of house finches to MG infection remain poorly understood. We developed seven new probe-based one-step quantitative reverse transcription polymerase chain reaction assays to investigate mRNA expression of house finch cytokine genes (IL1B, IL6, IL10, IL18, TGFB2, TNFSF15, and CXCLi2, syn. IL8L). These assays were then used to describe cytokine transcription profiles in a panel of 15 house finch tissues collected at three distinct time points during MG infection. Based on initial screening that indicated strong pro-inflammatory cytokine expression during MG infection at the periorbital sites in particular, we selected two key house finch tissues for further characterization: the nictitating membrane, i.e., the internal eyelid in direct contact with MG, and the Harderian gland, the secondary lymphoid tissue responsible for regulation of periorbital immunity. We characterized cytokine responses in these two tissues for 60 house finches experimentally inoculated either with media alone (sham) or one of two MG isolates: the earliest known pathogen isolate from house finches (VA1994) or an evolutionarily more derived isolate collected in 2006 (NC2006), which is known to be more virulent. We show that the more derived and virulent isolate NC2006, relative to VA1994, triggers stronger local inflammatory cytokine signaling, with peak cytokine expression generally occurring 3-6 days following MG inoculation. We also found that the extent of pro-inflammatory interleukin 1 beta signaling was correlated with conjunctival MG loads and the extent of clinical signs of conjunctivitis, the main pathological effect of MG in house finches. These results suggest that the pathogenicity caused by MG infection in house finches is largely mediated by host pro-inflammatory immune responses, with important implications for the dynamics of host-pathogen coevolution.
Collapse
Affiliation(s)
- Michal Vinkler
- Faculty of Science, Department of Zoology, Charles University, Prague, Czechia
| | - Ariel E Leon
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, United States
| | - Laila Kirkpatrick
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, United States
| | - Rami A Dalloul
- Avian Immunobiology Laboratory, Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA, United States
| | - Dana M Hawley
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
131
|
Vinkler M, Leon AE, Kirkpatrick L, Dalloul RA, Hawley DM. Differing House Finch Cytokine Expression Responses to Original and Evolved Isolates of Mycoplasma gallisepticum. Front Immunol 2018; 9:13. [PMID: 29403495 PMCID: PMC5786573 DOI: 10.3389/fimmu.2018.00013] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 01/04/2018] [Indexed: 01/25/2023] Open
Abstract
The recent emergence of the poultry bacterial pathogen Mycoplasma gallisepticum (MG) in free-living house finches (Haemorhous mexicanus), which causes mycoplasmal conjunctivitis in this passerine bird species, resulted in a rapid coevolutionary arms-race between MG and its novel avian host. Despite extensive research on the ecological and evolutionary dynamics of this host-pathogen system over the past two decades, the immunological responses of house finches to MG infection remain poorly understood. We developed seven new probe-based one-step quantitative reverse transcription polymerase chain reaction assays to investigate mRNA expression of house finch cytokine genes (IL1B, IL6, IL10, IL18, TGFB2, TNFSF15, and CXCLi2, syn. IL8L). These assays were then used to describe cytokine transcription profiles in a panel of 15 house finch tissues collected at three distinct time points during MG infection. Based on initial screening that indicated strong pro-inflammatory cytokine expression during MG infection at the periorbital sites in particular, we selected two key house finch tissues for further characterization: the nictitating membrane, i.e., the internal eyelid in direct contact with MG, and the Harderian gland, the secondary lymphoid tissue responsible for regulation of periorbital immunity. We characterized cytokine responses in these two tissues for 60 house finches experimentally inoculated either with media alone (sham) or one of two MG isolates: the earliest known pathogen isolate from house finches (VA1994) or an evolutionarily more derived isolate collected in 2006 (NC2006), which is known to be more virulent. We show that the more derived and virulent isolate NC2006, relative to VA1994, triggers stronger local inflammatory cytokine signaling, with peak cytokine expression generally occurring 3-6 days following MG inoculation. We also found that the extent of pro-inflammatory interleukin 1 beta signaling was correlated with conjunctival MG loads and the extent of clinical signs of conjunctivitis, the main pathological effect of MG in house finches. These results suggest that the pathogenicity caused by MG infection in house finches is largely mediated by host pro-inflammatory immune responses, with important implications for the dynamics of host-pathogen coevolution.
Collapse
Affiliation(s)
- Michal Vinkler
- Faculty of Science, Department of Zoology, Charles University, Prague, Czechia
| | - Ariel E. Leon
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, United States
| | - Laila Kirkpatrick
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, United States
| | - Rami A. Dalloul
- Avian Immunobiology Laboratory, Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA, United States
| | - Dana M. Hawley
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
132
|
Jones ME, Lebonville CL, Paniccia JE, Balentine ME, Reissner KJ, Lysle DT. Hippocampal interleukin-1 mediates stress-enhanced fear learning: A potential role for astrocyte-derived interleukin-1β. Brain Behav Immun 2018; 67:355-363. [PMID: 28963000 PMCID: PMC5696098 DOI: 10.1016/j.bbi.2017.09.016] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Revised: 09/19/2017] [Accepted: 09/25/2017] [Indexed: 12/27/2022] Open
Abstract
Post-traumatic stress disorder (PTSD) is associated with immune dysregulation. We have previously shown that severe stress exposure in a preclinical animal model of the disorder, stress-enhanced fear learning (SEFL), is associated with an increase in hippocampal interleukin-1β (IL-1β) and that blocking central IL-1 after the severe stress prevents the development of SEFL. Here, we tested whether blocking hippocampal IL-1 signaling is sufficient to prevent enhanced fear learning and identified the cellular source of stress-induced IL-1β in this region. Experiment 1 tested whether intra-dorsal hippocampal (DH) infusions of interleukin-1 receptor antagonist (IL-1RA, 1.25µg per hemisphere) 24 and 48h after stress exposure prevents the development of enhanced fear learning. Experiment 2 used triple fluorescence immunohistochemistry to examine hippocampal alterations in IL-1β, glial fibrillary acidic protein (GFAP), an astrocyte-specific marker, and ionized calcium binding adaptor molecule -1 (Iba-1), a microglial-specific marker, 48h after exposure to the severe stressor of the SEFL paradigm. Intra-DH IL-1RA prevented SEFL and stress-induced IL-1β was primarily colocalized with astrocytes in the hippocampus. Further, hippocampal GFAP immunoreactivity was not altered, whereas hippocampal Iba-1 immunoreactivity was significantly attenuated following severe stress. These data suggest that hippocampal IL-1 signaling is critical to the development of SEFL and that astrocytes are a predominant source of stress-induced IL-1β.
Collapse
|
133
|
Trevisi E, Minuti A. Assessment of the innate immune response in the periparturient cow. Res Vet Sci 2017; 116:47-54. [PMID: 29223307 DOI: 10.1016/j.rvsc.2017.12.001] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 12/03/2017] [Indexed: 01/24/2023]
Abstract
The transition period is the most critical phase in the life of high yielding dairy cows. Within a few weeks, cows are submitted to many challenges (physiological, nutritional, psychological, management) that require prompt and effective adaptive responses. The immune system is involved in this process, and many changes of the cow's immune system components have been observed around calving. Cows are considered to be immunosuppressed in late lactation, and available data suggest that the immune system is dysregulated around parturition. Significant attention has been focused on modification of cellular functions (e.g. the reduction of phagocytosis and diapedesis), but growing interest concerns the components of the innate immune system, which often exhibits increased responses such as susceptibility to inflammatory events and the related acute phase response (APR). Systemic inflammation plays a significant role in early lactation, affects many liver functions and has been associated with the impairment of cow performance (i.e. reduced feed intake, milk yield, fertility, welfare). The assessment of variations in immune-metabolic indices offers opportunities to predict the onset of the health troubles and to anticipate the proper therapies needed to guarantee health, good welfare and fertility in the following lactation. The frequency of diseases (metabolic and infectious) before calving is rare, but several clues suggest that various metabolic and immune variations can begin during the dry period. Interesting preliminary results encourage this perspective and possible candidates are suggested.
Collapse
Affiliation(s)
- Erminio Trevisi
- Istituto di Zootecnica, Facoltà di Scienze Agrarie, Alimentari ed Ambientali, Università Cattolica del Sacro Cuore, 29122 Piacenza, Italy; PRONUTRIGEN-Centro di Ricerca Nutrigenomica e Proteomica, Università Cattolica del Sacro Cuore, 29122 Piacenza, Italy.
| | - Andrea Minuti
- Istituto di Zootecnica, Facoltà di Scienze Agrarie, Alimentari ed Ambientali, Università Cattolica del Sacro Cuore, 29122 Piacenza, Italy; PRONUTRIGEN-Centro di Ricerca Nutrigenomica e Proteomica, Università Cattolica del Sacro Cuore, 29122 Piacenza, Italy
| |
Collapse
|
134
|
Chronic social isolation in adaptation of HPA axis to heterotypic stress. Pharmacol Rep 2017; 69:1213-1223. [PMID: 29128802 DOI: 10.1016/j.pharep.2017.08.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 08/04/2017] [Accepted: 08/23/2017] [Indexed: 11/21/2022]
|
135
|
Chen Y, Xu L, Xie HQH, Xu T, Fu H, Zhang S, Sha R, Xia Y, Zhao B. Identification of differentially expressed genes response to TCDD in rat brain after long-term low-dose exposure. J Environ Sci (China) 2017; 62:92-99. [PMID: 29289296 DOI: 10.1016/j.jes.2017.07.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 07/09/2017] [Accepted: 07/11/2017] [Indexed: 06/07/2023]
Abstract
Several cohort studies have reported that dioxin and dioxin-like polychlorinated biphenyls might impair the nervous system and lead to neurological or neurodegenerative diseases in the elder people, but there is limited research on the involved mechanism. By using microarray analysis, we figured out the differentially expressed genes between brain samples from SD rats after low-dose (0.1μg/(kg▪bw)) dioxin exposure for six months and controls. To investigate the function changes in the course of dioxin exposure, Gene Ontology (GO) annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis were performed on the differentially expressed genes. And the changes of several picked genes have been verified by real-time PCR. A total of 145 up-regulated and 64 down-regulated genes were identified. The metabolic processes, interleukin-1 secretion and production were significantly associated with the differentially expressed genes. And the genes regulated by dioxin also clustered to cholinergic synapse and long-term potentiation. Candidate biomarker genes such as egr1, gad2, gabrb3, abca1, ccr5 and pycard may be toxicological targets for dioxin. Furthermore, synaptic plasticity and neuro-immune system may be two principal affected areas by dioxin.
Collapse
Affiliation(s)
- Yangsheng Chen
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Li Xu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Heidi Q H Xie
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Tuan Xu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hualing Fu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Songyan Zhang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Rui Sha
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yingjie Xia
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bin Zhao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
136
|
Mice lacking interleukin-18 gene display behavioral changes in animal models of psychiatric disorders: Possible involvement of immunological mechanisms. J Neuroimmunol 2017; 314:58-66. [PMID: 29195684 DOI: 10.1016/j.jneuroim.2017.11.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 10/22/2017] [Accepted: 11/09/2017] [Indexed: 11/23/2022]
Abstract
Preclinical and clinical evidence suggests pro-inflammatory cytokines might play an important role in the neurobiology of schizophrenia and stress-related psychiatric disorders. Interleukin-18 (IL-18) is a member of the IL-1 family of cytokines and it is widely expressed in brain regions involved in emotional regulation. Since IL-18 involvement in the neurobiology of mental illnesses, including schizophrenia, remains unknown, this work aimed at investigating the behavior of IL-18 null mice (KO) in different preclinical models: 1. the prepulse inhibition test (PPI), which provides an operational measure of sensorimotor gating and schizophrenic-like phenotypes; 2. amphetamine-induced hyperlocomotion, a model predictive of antipsychotic activity; 3. resident-intruder test, a model predictive of aggressive behavior. Furthermore, the animals were submitted to models used to assess depressive- and anxiety-like behavior. IL-18KO mice showed impaired baseline PPI response, which was attenuated by d-amphetamine at a dose that did not modify PPI response in wild-type (WT) mice, suggesting a hypodopaminergic prefrontal cortex function in those mice. d-Amphetamine, however, induced hyperlocomotion in IL-18KO mice compared to their WT counterparts, suggesting hyperdopaminergic activity in the midbrain. Moreover, IL-18KO mice presented increased basal levels of IL-1β levels in the hippocampus and TNF-α in the prefrontal cortex, suggesting an overcompensation of IL-18 absence by increased levels of other proinflammatory cytokines. Although no alteration was observed in the forced swimming or in the elevated plus maze tests in naïve IL-18KO mice, these mice presented anxiogenic-like behavior after exposure to repeated forced swimming stress. In conclusion, deletion of the IL-18 gene resembled features similar to symptoms observed in schizophrenia (positive and cognitive symptoms, aggressive behavior), in addition to increased susceptibility to stress. The IL-18KO model, therefore, could provide new insights into how changes in brain immunological homeostasis induce behavioral changes related to psychiatric disorders, such as schizophrenia.
Collapse
|
137
|
O'Léime CS, Cryan JF, Nolan YM. Nuclear deterrents: Intrinsic regulators of IL-1β-induced effects on hippocampal neurogenesis. Brain Behav Immun 2017; 66:394-412. [PMID: 28751020 DOI: 10.1016/j.bbi.2017.07.153] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 07/15/2017] [Accepted: 07/23/2017] [Indexed: 12/11/2022] Open
Abstract
Hippocampal neurogenesis, the process by which new neurons are born and develop into the host circuitry, begins during embryonic development and persists throughout adulthood. Over the last decade considerable insights have been made into the role of hippocampal neurogenesis in cognitive function and the cellular mechanisms behind this process. Additionally, an increasing amount of evidence exists on the impact of environmental factors, such as stress and neuroinflammation on hippocampal neurogenesis and subsequent impairments in cognition. Elevated expression of the pro-inflammatory cytokine interleukin-1β (IL-1β) in the hippocampus is established as a significant contributor to the neuronal demise evident in many neurological and psychiatric disorders and is now known to negatively regulate hippocampal neurogenesis. In order to prevent the deleterious effects of IL-1β on neurogenesis it is necessary to identify signalling pathways and regulators of neurogenesis within neural progenitor cells that can interact with IL-1β. Nuclear receptors are ligand regulated transcription factors that are involved in modulating a large number of cellular processes including neurogenesis. In this review we focus on the signalling mechanisms of specific nuclear receptors involved in regulating neurogenesis (glucocorticoid receptors, peroxisome proliferator activated receptors, estrogen receptors, and nuclear receptor subfamily 2 group E member 1 (NR2E1 or TLX)). We propose that these nuclear receptors could be targeted to inhibit neuroinflammatory signalling pathways associated with IL-1β. We discuss their potential to be therapeutic targets for neuroinflammatory disorders affecting hippocampal neurogenesis and associated cognitive function.
Collapse
Affiliation(s)
- Ciarán S O'Léime
- Department of Anatomy and Neuroscience, University College Cork, Ireland
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Ireland; APC Microbiome Institute, University College Cork, Ireland
| | - Yvonne M Nolan
- Department of Anatomy and Neuroscience, University College Cork, Ireland; APC Microbiome Institute, University College Cork, Ireland.
| |
Collapse
|
138
|
Liu A, Wu Y, Li L, Wang Y. The roles of interleukin‐1 and RhoA signaling pathway in rat epilepsy model treated with low‐frequency electrical stimulation. J Cell Biochem 2017; 119:2535-2544. [DOI: 10.1002/jcb.26415] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 09/21/2017] [Indexed: 11/09/2022]
Affiliation(s)
- Ai‐Hua Liu
- Department of NeurologyXuanwu HospitalCapital Medical UniversityBeijingP.R. China
- Beijing Intensive Epilepsy CenterBeijing Key Laboratory of NeuromodulationBeijingP.R. China
| | - Ya‐Ting Wu
- Department of NeurologyXuanwu HospitalCapital Medical UniversityBeijingP.R. China
- Beijing Intensive Epilepsy CenterBeijing Key Laboratory of NeuromodulationBeijingP.R. China
| | - Li‐Ping Li
- Department of NeurologyXuanwu HospitalCapital Medical UniversityBeijingP.R. China
- Beijing Intensive Epilepsy CenterBeijing Key Laboratory of NeuromodulationBeijingP.R. China
| | - Yu‐Ping Wang
- Department of NeurologyXuanwu HospitalCapital Medical UniversityBeijingP.R. China
- Beijing Intensive Epilepsy CenterBeijing Key Laboratory of NeuromodulationBeijingP.R. China
| |
Collapse
|
139
|
Egorov AI, Griffin SM, Converse RR, Styles JN, Sams EA, Wilson A, Jackson LE, Wade TJ. Vegetated land cover near residence is associated with reduced allostatic load and improved biomarkers of neuroendocrine, metabolic and immune functions. ENVIRONMENTAL RESEARCH 2017; 158:508-521. [PMID: 28709033 PMCID: PMC5941947 DOI: 10.1016/j.envres.2017.07.009] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 07/03/2017] [Accepted: 07/04/2017] [Indexed: 05/21/2023]
Abstract
BACKGROUND Greater exposure to urban green spaces has been linked to reduced risks of depression, cardiovascular disease, diabetes and premature death. Alleviation of chronic stress is a hypothesized pathway to improved health. Previous studies linked chronic stress with a biomarker-based composite measure of physiological dysregulation known as allostatic load. OBJECTIVE This study's objective was to assess the relationship between vegetated land cover near residences and allostatic load. METHODS This cross-sectional population-based study involved 206 adult residents of the Durham-Chapel Hill, North Carolina metropolitan area. Exposure was quantified using high-resolution metrics of trees and herbaceous vegetation within 500m of each residence derived from the U.S. Environmental Protection Agency's EnviroAtlas land cover dataset. Eighteen biomarkers of immune, neuroendocrine, and metabolic functions were measured in serum or saliva samples. Allostatic load was defined as a sum of potentially unhealthy biomarker values dichotomized at 10th or 90th percentile of sample distribution. Regression analysis was conducted using generalized additive models with two-dimensional spline smoothing function of geographic coordinates, weighted measures of vegetated land cover allowing decay of effects with distance, and geographic and demographic covariates. RESULTS An inter-quartile range increase in distance-weighted vegetated land cover was associated with 37% (95% Confidence Limits 46%; 27%) reduced allostatic load; significantly reduced adjusted odds of having low level of norepinephrine, dopamine, and dehydroepiandrosterone, and high level of epinephrine, fibrinogen, vascular cell adhesion molecule-1, and interleukin-8 in serum, and α-amylase in saliva; and reduced odds of previously diagnosed depression. CONCLUSIONS The observed effects of vegetated land cover on allostatic load and individual biomarkers are consistent with prevention of depression, cardiovascular disease and premature mortality.
Collapse
Affiliation(s)
- Andrey I Egorov
- National Health and Environmental Effects Research Laboratory, United States Environmental Protection Agency, Research Triangle Park, NC, USA.
| | - Shannon M Griffin
- National Exposure Research Laboratory, United States Environmental Protection Agency, Cincinnati, OH, USA
| | - Reagan R Converse
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jennifer N Styles
- National Health and Environmental Effects Research Laboratory, United States Environmental Protection Agency, Research Triangle Park, NC, USA; National Exposure Research Laboratory, United States Environmental Protection Agency, Cincinnati, OH, USA
| | - Elizabeth A Sams
- National Health and Environmental Effects Research Laboratory, United States Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Anthony Wilson
- Association of Schools and Programs of Public Health fellow at the United States Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Laura E Jackson
- National Health and Environmental Effects Research Laboratory, United States Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Timothy J Wade
- National Health and Environmental Effects Research Laboratory, United States Environmental Protection Agency, Research Triangle Park, NC, USA
| |
Collapse
|
140
|
Li M, Li C, Yu H, Cai X, Shen X, Sun X, Wang J, Zhang Y, Wang C. Lentivirus-mediated interleukin-1β (IL-1β) knock-down in the hippocampus alleviates lipopolysaccharide (LPS)-induced memory deficits and anxiety- and depression-like behaviors in mice. J Neuroinflammation 2017; 14:190. [PMID: 28931410 PMCID: PMC5607621 DOI: 10.1186/s12974-017-0964-9] [Citation(s) in RCA: 159] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 09/14/2017] [Indexed: 12/26/2022] Open
Abstract
Background Recent evidence has suggested that peripheral inflammatory responses induced by lipopolysaccharides (LPS) play an important role in neuropsychiatric dysfunction in rodents. Interleukin-1β (IL-1β), a pro-inflammatory cytokine, has been proposed to be a key mediator in a variety of behavioral dysfunction induced by LPS in mice. Thus, inhibition of IL-1β may have a therapeutic benefit in the treatment of neuropsychiatric disorders. However, the precise underlying mechanism of knock-down of IL-1β in repairing behavioral changes by LPS remains unclear. Methods The mice were treated with either IL-1β shRNA lentivirus or non-silencing shRNA control (NS shRNA) lentivirus by microinjection into the dentate gyrus (DG) regions of the hippocampus. After 7 days of recovery, LPS (1 mg/kg, i.p.) or saline was administered. The behavioral task for memory deficits was conducted in mice by the novel object recognition test (NORT), the anxiety-like behaviors were evaluated by the elevated zero maze (EZM), and the depression-like behaviors were examined by the sucrose preference test (SPT) and the forced swimming test (FST). Furthermore, the levels of malondialdehyde (MDA), superoxide dismutase (SOD), nuclear factor erythroid-derived 2-like 2 (Nrf2), heme oxygenase 1 (HO1), IL-1β, tumor necrosis factor (TNF-α), neuropeptide VGF (non-acronymic), and brain-derived neurotrophic factor (BDNF) were assayed. Results Our results demonstrated that IL-1β knock-down in the hippocampus significantly attenuated the memory deficits and anxiety- and depression-like behaviors induced by LPS in mice. In addition, IL-1β knock-down ameliorated the oxidative and neuroinflammatory responses and abolished the downregulation of VGF and BDNF induced by LPS. Conclusions Collectively, our findings suggest that IL-1β is necessary for the oxidative and neuroinflammatory responses produced by LPS and offers a novel drug target in the IL-1β/oxidative/neuroinflammatory/neurotrophic pathway for treating neuropsychiatric disorders that are closely associated with neuroinflammation, oxidative stress, and the downregulation of VGF and BDNF.
Collapse
Affiliation(s)
- Mengmeng Li
- Ningbo Key Laboratory of Behavioral Neuroscience, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, 315211, People's Republic of China.,Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, 315211, People's Republic of China.,Department of Physiology and Pharmacology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, 315211, People's Republic of China
| | - Chenli Li
- Ningbo Key Laboratory of Behavioral Neuroscience, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, 315211, People's Republic of China.,Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, 315211, People's Republic of China.,Department of Physiology and Pharmacology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, 315211, People's Republic of China
| | - Hanjie Yu
- Ningbo Key Laboratory of Behavioral Neuroscience, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, 315211, People's Republic of China.,Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, 315211, People's Republic of China.,Department of Physiology and Pharmacology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, 315211, People's Republic of China
| | - Xiongxiong Cai
- Ningbo Key Laboratory of Behavioral Neuroscience, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, 315211, People's Republic of China.,Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, 315211, People's Republic of China.,Department of Physiology and Pharmacology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, 315211, People's Republic of China
| | - Xinbei Shen
- Ningbo Key Laboratory of Behavioral Neuroscience, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, 315211, People's Republic of China.,Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, 315211, People's Republic of China.,Department of Physiology and Pharmacology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, 315211, People's Republic of China
| | - Xin Sun
- Ningbo Key Laboratory of Behavioral Neuroscience, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, 315211, People's Republic of China.,Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, 315211, People's Republic of China.,Department of Physiology and Pharmacology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, 315211, People's Republic of China
| | - Jinting Wang
- Ningbo Key Laboratory of Behavioral Neuroscience, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, 315211, People's Republic of China.,Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, 315211, People's Republic of China.,Department of Physiology and Pharmacology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, 315211, People's Republic of China
| | - Yanhua Zhang
- Ningbo Key Laboratory of Behavioral Neuroscience, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, 315211, People's Republic of China.,Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, 315211, People's Republic of China.,Department of Physiology and Pharmacology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, 315211, People's Republic of China
| | - Chuang Wang
- Ningbo Key Laboratory of Behavioral Neuroscience, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, 315211, People's Republic of China. .,Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, 315211, People's Republic of China. .,Department of Physiology and Pharmacology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, 315211, People's Republic of China. .,Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, Ningbo University, Ningbo, 315211, People's Republic of China.
| |
Collapse
|
141
|
Lovelock DF, Deak T. Repeated exposure to two stressors in sequence demonstrates that corticosterone and paraventricular nucleus of the hypothalamus interleukin-1β responses habituate independently. J Neuroendocrinol 2017; 29:10.1111/jne.12514. [PMID: 28803453 PMCID: PMC5617797 DOI: 10.1111/jne.12514] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 08/07/2017] [Accepted: 08/08/2017] [Indexed: 01/09/2023]
Abstract
A wide range of stress-related pathologies such as post-traumatic stress disorder are considered to arise from aberrant or maladaptive forms of stress adaptation. The hypothalamic-pituitary-adrenal (HPA) axis readily adapts to repeated stressor exposure, yet little is known about adaptation in neuroimmune responses to repeated or sequential stress challenges. In Experiment 1, rats were exposed to 10 days of restraint alone (60 minutes daily), forced swim alone (30 minutes daily) or daily sequential exposure to restraint (60 minutes) followed immediately by forced swim (30 minutes), termed sequential stress exposure. Habituation of the corticosterone (CORT) response occurred to restraint by 5 days and swim at 10 days, whereas rats exposed to sequential stress exposure failed to display habituation to the combined challenge. Experiment 2 compared 1 or 5 days of forced swim with sequential stress exposure and examined how each affected expression of several neuroimmune and cellular activation genes in the paraventricular nucleus of the hypothalamus (PVN), prefrontal cortex (PFC) and hippocampus (HPC). Sequential exposure to restraint and swim increased interleukin (IL)-1β in the PVN, an effect that was attenuated after 5 days. Sequential stress exposure also elicited IL-6 and tumour necrosis factor-α responses in the HPC and PFC, respectively, which did not habituate after 5 days. Experiment 3 tested whether prior habituation to restraint (5 days) would alter the IL-1β response evoked by swim exposure imposed immediately after the sixth day of restraint. Surprisingly, a history of repeated exposure to restraint attenuated the PVN IL-1β response after swim in comparison to acutely-exposed subjects despite an equivalent CORT response. Overall, these findings suggest that habituation of neuroimmune responses to stress proceeds: (i) independent of HPA axis habituation; (ii) likely requires more daily sessions of stress to develop; and (iii) IL-1β displays a greater tendency to habituate after repeated stress challenges compared to other stress-reactive cytokines.
Collapse
Affiliation(s)
- Dennis F. Lovelock
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University—SUNY, Binghamton NY 13902-6000
| | - Terrence Deak
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University—SUNY, Binghamton NY 13902-6000
| |
Collapse
|
142
|
Bhakta A, Gavini K, Yang E, Lyman-Henley L, Parameshwaran K. Chronic traumatic stress impairs memory in mice: Potential roles of acetylcholine, neuroinflammation and corticotropin releasing factor expression in the hippocampus. Behav Brain Res 2017; 335:32-40. [DOI: 10.1016/j.bbr.2017.08.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 07/24/2017] [Accepted: 08/05/2017] [Indexed: 12/15/2022]
|
143
|
Domingos LB, Hott SC, Terzian ALB, Resstel LBM. P2X7 purinergic receptors participate in the expression and extinction processes of contextual fear conditioning memory in mice. Neuropharmacology 2017; 128:474-481. [PMID: 28802645 DOI: 10.1016/j.neuropharm.2017.08.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 07/27/2017] [Accepted: 08/08/2017] [Indexed: 12/21/2022]
Abstract
The purinergic system consists of two large receptor families - P2X and P2Y. Both are activated by adenosine triphosphate (ATP), although presenting different functions. These receptors are present in several brain regions, including those involved in emotion and stress-related behaviors. Hence, they seem to participate in fear- and anxiety-related responses. However, few studies have investigated the purinergic system in threatening situations, as observed in contextual fear conditioning (CFC). Therefore, this study investigated the involvement of purinergic receptors in the expression and extinction of aversive memories. C57Bl/6 background mice were submitted to the CFC protocol. Wildtype (WT) mice received i.p. injection of either a nonselective P2 receptor (P2R) antagonist, P178 (10 or 30 mg/kg); a selective P2X7 receptor (P2X7R) antagonist, A438079 (10 mg/kg); a selective P2Y1 receptor (P2Y1R) antagonist, MRS2179 (10 mg/kg); or vehicle 10 min prior to or immediately after the extinction session. Additionally, P2X7R KO mice were tested in the CFC protocol. After P2R antagonist treatment, contextual fear recall increased, while acquisition of extinction was impaired. Similar results were observed with the selective P2X7R antagonist, but not with the selective P2Y1R antagonist. Interestingly, P2X7R KO mice showed increased contextual fear recall, associated with impaired acquisition of extinction, in accordance with pharmacologic P2X7R antagonism. Our results suggest that specific pharmacological or genetic blockade of P2X7R promotes anxiogenic-like effects, along with deficits in extinction learning. Thus, these receptors could present an alternative treatment of stress-related psychiatric disorders.
Collapse
Affiliation(s)
- L B Domingos
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - S C Hott
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - A L B Terzian
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - L B M Resstel
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil.
| |
Collapse
|
144
|
Kaufmann FN, Costa AP, Ghisleni G, Diaz AP, Rodrigues ALS, Peluffo H, Kaster MP. NLRP3 inflammasome-driven pathways in depression: Clinical and preclinical findings. Brain Behav Immun 2017; 64:367-383. [PMID: 28263786 DOI: 10.1016/j.bbi.2017.03.002] [Citation(s) in RCA: 273] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 02/13/2017] [Accepted: 03/01/2017] [Indexed: 12/12/2022] Open
Abstract
Over the past three decades, an intricate interaction between immune activation, release of pro-inflammatory cytokines and changes in brain circuits related to mood and behavior has been described. Despite extensive efforts, questions regarding when inflammation becomes detrimental or how we can target the immune system to develop new therapeutic strategies for the treatment of psychiatric disorders remain unresolved. In this context, novel aspects of the neuroinflammatory process activated in response to stressful challenges have recently been documented in major depressive disorder (MDD). The Nod-like receptor pyrin containing 3 inflammasome (NLRP3) is an intracellular multiprotein complex responsible for a number of innate immune processes associated with infection, inflammation and autoimmunity. Recent data have demonstrated that NLRP3 activation appears to bridge the gap between immune activation and metabolic danger signals or stress exposure, which are key factors in the pathogenesis of psychiatric disorders. In this review, we discuss both preclinical and clinical evidence that links the assembly of the NLRP3 complex and the subsequent proteolysis and release of the pro-inflammatory cytokines interleukin-1β (IL-1β) and interleukin-18 (IL-18) in chronic stress models and patients with MDD. Importantly, we also focus on the therapeutic potential of targeting the NLRP3 inflammasome complex to improve stress resilience and depressive symptoms.
Collapse
Affiliation(s)
- Fernanda N Kaufmann
- Department of Biochemistry, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Ana Paula Costa
- Department of Biochemistry, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Gabriele Ghisleni
- Department of Life and Health Sciences, Catholic University of Pelotas, Rio Grande do Sul, Brazil
| | - Alexandre P Diaz
- Postgraduate Program in Health Sciences, University of Southern Santa Catarina, Santa Catarina, Brazil
| | - Ana Lúcia S Rodrigues
- Department of Biochemistry, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Hugo Peluffo
- Neuroinflammation and Gene Therapy Lab., Institut Pasteur de Montevideo, Uruguay; Dept. Histology and Embryology, Faculty of Medicine, UDELAR, Uruguay
| | - Manuella P Kaster
- Department of Biochemistry, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil.
| |
Collapse
|
145
|
Glenn DE, Acheson DT, Geyer MA, Nievergelt CM, Baker DG, Risbrough VB. Fear learning alterations after traumatic brain injury and their role in development of posttraumatic stress symptoms. Depress Anxiety 2017; 34:723-733. [PMID: 28489272 DOI: 10.1002/da.22642] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 03/20/2017] [Accepted: 04/02/2017] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND It is unknown how traumatic brain injury (TBI) increases risk for posttraumatic stress disorder (PTSD). One potential mechanism is via alteration of fear-learning processes that could affect responses to trauma memories and cues. We utilized a prospective, longitudinal design to determine if TBI is associated with altered fear learning and extinction, and if fear processing mediates effects of TBI on PTSD symptom change. METHODS Eight hundred fifty two active-duty Marines and Navy Corpsmen were assessed before and after deployment. Assessments included TBI history, PTSD symptoms, combat trauma and deployment stress, and a fear-potentiated startle task of fear acquisition and extinction. Startle response and self-reported expectancy and anxiety served as measures of fear conditioning, and PTSD symptoms were measured with the Clinician-Administered PTSD Scale. RESULTS Individuals endorsing "multiple hit" exposure (both deployment TBI and a prior TBI) showed the strongest fear acquisition and highest fear expression compared to groups without multiple hits. Extinction did not differ across groups. Endorsing a deployment TBI was associated with higher anxiety to the fear cue compared to those without deployment TBI. The association of deployment TBI with increased postdeployment PTSD symptoms was mediated by postdeployment fear expression when recent prior-TBI exposure was included as a moderator. TBI associations with increased response to threat cues and PTSD symptoms remained when controlling for deployment trauma and postdeployment PTSD diagnosis. CONCLUSIONS Deployment TBI, and multiple-hit TBI in particular, are associated with increases in conditioned fear learning and expression that may contribute to risk for developing PTSD symptoms.
Collapse
Affiliation(s)
- Daniel E Glenn
- Center of Excellence for Stress and Mental Health, San Diego Veterans Affairs Health Services, CA, USA.,Department of Psychiatry, University of California San Diego, CA, USA
| | - Dean T Acheson
- Center of Excellence for Stress and Mental Health, San Diego Veterans Affairs Health Services, CA, USA.,Department of Psychiatry, University of California San Diego, CA, USA
| | - Mark A Geyer
- Department of Psychiatry, University of California San Diego, CA, USA.,Research Service, VA San Diego Healthcare System, CA, USA
| | - Caroline M Nievergelt
- Center of Excellence for Stress and Mental Health, San Diego Veterans Affairs Health Services, CA, USA.,Department of Psychiatry, University of California San Diego, CA, USA
| | - Dewleen G Baker
- Center of Excellence for Stress and Mental Health, San Diego Veterans Affairs Health Services, CA, USA.,Department of Psychiatry, University of California San Diego, CA, USA
| | - Victoria B Risbrough
- Center of Excellence for Stress and Mental Health, San Diego Veterans Affairs Health Services, CA, USA.,Department of Psychiatry, University of California San Diego, CA, USA
| | -
- Center of Excellence for Stress and Mental Health, San Diego Veterans Affairs Health Services, CA, USA.,Department of Psychiatry, University of California San Diego, CA, USA
| |
Collapse
|
146
|
Hertz L, Chen Y. Glycogenolysis, an Astrocyte-Specific Reaction, is Essential for Both Astrocytic and Neuronal Activities Involved in Learning. Neuroscience 2017; 370:27-36. [PMID: 28668486 DOI: 10.1016/j.neuroscience.2017.06.025] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 06/10/2017] [Accepted: 06/19/2017] [Indexed: 01/26/2023]
Abstract
In brain glycogen, formed from glucose, is degraded (glycogenolysis) in astrocytes but not in neurons. Although most of the degradation follows the same pathway as glucose, its breakdown product, l-lactate, is released from astrocytes in larger amounts than glucose when glycogenolysis is activated by noradrenaline. However, this is not the case when glycogenolysis is activated by high potassium ion (K+) concentrations - possibly because noradrenaline in contrast to high K+ stimulates glycogenolysis by an increase not only in free cytosolic Ca2+ concentration ([Ca2+]i) but also in cyclic AMP (c-AMP), which may increase the expression of the monocarboxylate transporter through which it is released. Several transmitters activate glycogenolysis in astrocytes and do so at different time points after training. This stimulation is essential for memory consolidation because glycogenolysis is necessary for uptake of K+ and stimulates formation of glutamate from glucose, and therefore is needed both for removal of increased extracellular K+ following neuronal excitation (which initially occurs into astrocytes) and for formation of transmitter glutamate and GABA. In addition the released l-lactate has effects on neurons which are essential for learning and for learning-related long-term potentiation (LTP), including induction of the neuronal gene Arc/Arg3.1 and activation of gene cascades mediated by CREB and cofilin. Inhibition of glycogenolysis blocks learning, LTP and all related molecular events, but all changes can be reversed by injection of l-lactate. The effect of extracellular l-lactate is due to both astrocyte-mediated signaling which activates noradrenergic activity on all brain cells and to a minor uptake, possibly into dendritic spines.
Collapse
Affiliation(s)
- Leif Hertz
- Laboratory of Metabolic Brain Diseases, Institute of Metabolic Disease Research and Drug Development, China Medical University, Shenyang, PR China
| | - Ye Chen
- Henry M. Jackson Foundation, Bethesda, MD 20817, USA.
| |
Collapse
|
147
|
Tumer MK, Nursal AF, Tekcan A, Yerliyurt K, Geyko A, Yigit S. The IL-1Ra gene variable number tandem repeat variant is associated with susceptibility to temporomandibular disorders in Turkish population. J Clin Lab Anal 2017; 32. [PMID: 28612927 DOI: 10.1002/jcla.22255] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 04/13/2017] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Temporomandibular joint disorders (TMD) are a group of disorders involving temporomandibular joint and related structures. Interleukine-1 receptor antagonist (IL-1Ra) is an important anti-inflammatory molecule that competes with other interleukin-1 molecules. This study was designed to investigate the possible association of the IL-1Ra VNTR variant with the risk of TMD in the Turkish population. METHODS Peripheral blood samples were collected from 100 patients with TMD (23 males, 77 females) and 110 healthy individuals (35 males, 75 females). Genotyping of IL-1Ra 86 bp VNTR variant was evaluated by gel electrophoresis after polymerase chain reaction (PCR). RESULTS Our results show that there is a statistically significant difference between TMD patients and control group with respect to IL-1Ra genotype distribution and allele frequencies. 1.2, 1.4, and 4.4 genotypes were more common in patients, while 2.2 and 3.3 genotypes were rarer (P<.000). Frequency of alleles 1 and 4 was higher in patient groups (P<.000), whereas alleles 2 and 3 had a lower frequency in patients with TMD (P<.000). CONCLUSIONS This is the first correlation study that evaluates the association between IL-1Ra gene VNTR variant and TMD. The VNTR variant related to IL-1Ra gene showed a strong pattern of association with TMD that may have a potential impact on disease counseling and management. Larger studies with various ethnicities are needed to establish the impact of IL-1Ra VNTR variant on risk of developing TMD.
Collapse
Affiliation(s)
- Mehmet Kemal Tumer
- Faculty of Dentistry, Department of Oral and Maxillofacial Surgery, Gaziosmanpasa University, Tokat, Turkey.,Faculty of Medicine, Department of Medical Biology, Gaziosmanpasa University, Tokat, Turkey
| | - Ayse Feyda Nursal
- Faculty of Medicine, Department of Medical Genetics, Hitit University, Çorum, Turkey
| | - Akin Tekcan
- Faculty of Medicine, Department of Medical Biology, Ahi Evran University, Kırşehir, Turkey
| | - Kaan Yerliyurt
- Faculty of Dentistry, Department of Prosthetic Dentistry, Gaziosmanpasa University, Tokat, Turkey
| | - Anastasia Geyko
- The Laboratory of Evolutionary Genomics, Vavilov Institute of General Genetics, Moscow, Russia
| | - Serbulent Yigit
- Faculty of Medicine, Department of Medical Biology, Gaziosmanpasa University, Tokat, Turkey
| |
Collapse
|
148
|
Urwyler SA, Schuetz P, Ebrahimi F, Donath MY, Christ-Crain M. Interleukin-1 Antagonism Decreases Cortisol Levels in Obese Individuals. J Clin Endocrinol Metab 2017; 102:1712-1718. [PMID: 28324042 DOI: 10.1210/jc.2016-3931] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 02/15/2017] [Indexed: 02/13/2023]
Abstract
CONTEXT Increased cortisol levels in obesity may contribute to the associated metabolic syndrome. In obesity, the activated innate immune system leads to increased interleukin (IL)-1β, which is known to stimulate the release of adrenocorticotropin hormone (ACTH). OBJECTIVES We hypothesized that in obesity IL-1 antagonism would result in downregulation of the hypothalamo-pituitary-adrenal axis, leading to decreased cortisol levels. DESIGN AND PARTICIPANTS In this prospective intervention study, we included 73 patients with obesity (body mass index [BMI] ≥30 kg/m2) and at least one additional feature of the metabolic syndrome. OUTCOME MEASURES The primary end point was change in morning cortisol from baseline to after the administration of the IL-1 receptor antagonist (anakinra/Kineret®, total dose 3 × 100 mg). Secondary end points were effects on salivary cortisol and ACTH. RESULTS Median age was 56 years, 50.7% of patients were female, and median BMI was 36.3 kg/m2. Median morning serum cortisol levels (nmol/L) decreased significantly after IL-1 antagonism [from baseline, 452 to 423; absolute difference, -38.7; 95% confidence interval (CI), -64 to -13.4; P = 0.0019]. Similar effects were found for salivary cortisol levels (-2.8; 95% CI, -4.4 to -1.3; P = 0.0007), ACTH levels (-2.2; 95% CI; -4.2 to -0.1; P = 0.038), systolic blood pressure (-5.2, 95% CI, -8.5 to -1.8; P = 0.0006), and heart rate (-2.9; 95% CI, -4.7 to -1.0; P = 0.0029). CONCLUSION IL-1 antagonism in obese individuals with features of the metabolic syndrome leads to a decrease in serum cortisol, salivary cortisol, and ACTH levels along with a reduction in systolic blood pressure and heart rate.
Collapse
Affiliation(s)
- Sandrine Andrea Urwyler
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, CH-4031 Basel, Switzerland
- Department of Clinical Research, University Hospital Basel, CH-4031 Basel, Switzerland
| | - Philipp Schuetz
- Department of Clinical Research, University Hospital Basel, CH-4031 Basel, Switzerland
- Department of Endocrinology, Medical University Clinic, Kantonsspital Aarau, CH-5001 Aarau, Switzerland
| | - Fahim Ebrahimi
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, CH-4031 Basel, Switzerland
- Department of Clinical Research, University Hospital Basel, CH-4031 Basel, Switzerland
| | - Marc Y Donath
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, CH-4031 Basel, Switzerland
- Department of Clinical Research, University Hospital Basel, CH-4031 Basel, Switzerland
| | - Mirjam Christ-Crain
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, CH-4031 Basel, Switzerland
- Department of Clinical Research, University Hospital Basel, CH-4031 Basel, Switzerland
| |
Collapse
|
149
|
Yan J, Bai J, Gao C, Liang Y, Zhao B, Bian Y. Chronic unpredictable stress abrogates the endotoxin tolerance induced by repeated peripheral LPS challenge via the TLR4 signaling pathway. Neurosci Lett 2017; 645:7-13. [DOI: 10.1016/j.neulet.2017.02.070] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 02/27/2017] [Accepted: 02/27/2017] [Indexed: 02/06/2023]
|
150
|
Su WJ, Zhang Y, Chen Y, Gong H, Lian YJ, Peng W, Liu YZ, Wang YX, You ZL, Feng SJ, Zong Y, Lu GC, Jiang CL. NLRP3 gene knockout blocks NF-κB and MAPK signaling pathway in CUMS-induced depression mouse model. Behav Brain Res 2017; 322:1-8. [PMID: 28093255 DOI: 10.1016/j.bbr.2017.01.018] [Citation(s) in RCA: 162] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 12/20/2016] [Accepted: 01/10/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND Abundant researches indicate that neuroinflammation has important roles in the pathophysiology of depression. Our previous study found that the NLRP3 inflammasome mediated stress-induced depression-like behavior in mice via regulating neuroinflammation. However, it still remains unclear that how the NLRP3 inflammasome influences related inflammatory signaling pathway to contribute to neuroinflammation in depression. METHODS We used wild-type mice and NLRP3 gene knockout mice to explore the role of NLRP3 inflammasome and related inflammatory signaling pathways in chronic unpredictable mild stress (CUMS) induced depression mouse model. RESULTS Both wild-type and NLRP3 knockout stress group mice gained less weight than control group mice after 4 weeks CUMS exposure. The wild-type mice subjected to 4 weeks CUMS displayed depression-like behaviors, including decreased sucrose preference and increased immobility time in the tail suspension test. The NLRP3 knockout stress group mice didn't demonstrate depression-like behaviors. The levels of interleukin-1β protein in serum and hippocampi of CUMS exposed wild-type mice were significantly higher, while the NLRP3 knockout stress group mice didn't show an elevation of interleukin-1β levels. Similarly to early researches, we found that CUMS led to promoted NLRP3 expression in hippocampi. In addition, the hippocampi in CUMS exposed wild-type mice had higher p-JNK and p-p38 protein expression, which indicated activation of the mitogen-activated protein kinases (MAPK) pathway. The knockout of NLRP3 gene inhibited CUMS-induced activation of the MAPK pathway. The nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) protein complex was activated in the hippocampi of wild-type mice after CUMS exposure, while knockout of NLRP3 gene hindered its activation. CONCLUSIONS These data further proved that the NLRP3 inflammasome mediated CUMS-induced depression-like behavior. The NLRP3 inflammasome regulated CUMS-induced MAPK pathway and NF-κB protein complex activation in depression mouse model. Further researches targeting the NLRP3 inflammasome-signaling pathway might be under a promising future in the prevention and treatment of depression.
Collapse
Affiliation(s)
- Wen-Jun Su
- Laboratory of Stress Medicine, Faculty of Psychology and Mental Health, Second Military Medical University, 800 Xiangyin Road, Shanghai, China
| | - Yi Zhang
- Laboratory of Stress Medicine, Faculty of Psychology and Mental Health, Second Military Medical University, 800 Xiangyin Road, Shanghai, China; Department of Psychiatry, Faculty of Psychology and Mental Health, Second Military Medical University, 800 Xiangyin Road, Shanghai, China
| | - Ying Chen
- Department of Pharmacy, The 81st Hospital of PLA, 34 Yanggongjing, Nanjing, China
| | - Hong Gong
- Laboratory of Stress Medicine, Faculty of Psychology and Mental Health, Second Military Medical University, 800 Xiangyin Road, Shanghai, China
| | - Yong-Jie Lian
- Laboratory of Stress Medicine, Faculty of Psychology and Mental Health, Second Military Medical University, 800 Xiangyin Road, Shanghai, China
| | - Wei Peng
- Laboratory of Stress Medicine, Faculty of Psychology and Mental Health, Second Military Medical University, 800 Xiangyin Road, Shanghai, China
| | - Yun-Zi Liu
- Laboratory of Stress Medicine, Faculty of Psychology and Mental Health, Second Military Medical University, 800 Xiangyin Road, Shanghai, China
| | - Yun-Xia Wang
- Laboratory of Stress Medicine, Faculty of Psychology and Mental Health, Second Military Medical University, 800 Xiangyin Road, Shanghai, China
| | - Zi-Li You
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Shi-Jie Feng
- New Drug Safety Evaluation Center, Second Military Medical University, 800 Xiangyin Road, Shanghai, China
| | - Ying Zong
- New Drug Safety Evaluation Center, Second Military Medical University, 800 Xiangyin Road, Shanghai, China
| | - Guo-Cai Lu
- New Drug Safety Evaluation Center, Second Military Medical University, 800 Xiangyin Road, Shanghai, China.
| | - Chun-Lei Jiang
- Laboratory of Stress Medicine, Faculty of Psychology and Mental Health, Second Military Medical University, 800 Xiangyin Road, Shanghai, China.
| |
Collapse
|