201
|
Vasilyeva EF, Kushner SG, Factor MI, Omelchenko MA, Bogdanova ED, Petrakova LN, Brusov OS. The cellular factors of innate immunity in nonpsychotic patients at high risk for schizophrenia. Zh Nevrol Psikhiatr Im S S Korsakova 2016; 116:60-65. [DOI: 10.17116/jnevro201611610160-65] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
202
|
McGuiness B, Gibney SM, Beumer W, Versnel MA, Sillaber I, Harkin A, Drexhage HA. Exaggerated Increases in Microglia Proliferation, Brain Inflammatory Response and Sickness Behaviour upon Lipopolysaccharide Stimulation in Non-Obese Diabetic Mice. Neuroimmunomodulation 2016; 23:137-150. [PMID: 27529430 PMCID: PMC5296925 DOI: 10.1159/000446370] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 04/18/2016] [Indexed: 11/19/2022] Open
Abstract
UNLABELLED The non-obese diabetic (NOD) mouse, an established model for autoimmune diabetes, shows an exaggerated reaction of pancreas macrophages to inflammatory stimuli. NOD mice also display anxiety when immune-stimulated. Chronic mild brain inflammation and a pro-inflammatory microglial activation is critical in psychiatric behaviour. OBJECTIVE To explore brain/microglial activation and behaviour in NOD mice at steady state and after systemic lipopolysaccharide (LPS) injection. METHODS Affymetrix analysis on purified microglia of pre-diabetic NOD mice (8-10 weeks) and control mice (C57BL/6 and CD1 mice, the parental non-autoimmune strain) at steady state and after systemic LPS (100 μg/kg) administration. Quantitative PCR was performed on the hypothalamus for immune activation markers (IL-1β, IFNγ and TNFα) and growth factors (BDNF and PDGF). Behavioural profiling of NOD, CD1, BALB/c and C57BL/6 mice at steady state was conducted and sickness behaviour/anxiety in NOD and CD1 mice was monitored before and after LPS injection. RESULTS Genome analysis revealed cell cycle/cell death and survival aberrancies of NOD microglia, substantiated as higher proliferation on BrdU staining. Inflammation signs were absent. NOD mice had a hyper-reactive response to novel environments with some signs of anxiety. LPS injection induced a higher expression of microglial activation markers, a higher brain pro-inflammatory set point (IFNγ, IDO) and a reduced expression of BDNF and PDGF after immune stimulation in NOD mice. NOD mice displayed exaggerated and prolonged sickness behaviour after LPS administration. CONCLUSION After stimulation with LPS, NOD mice display an increased microglial proliferation and an exaggerated inflammatory brain response with reduced BDNF and PDGF expression and increased sickness behaviour as compared to controls.
Collapse
Affiliation(s)
- Barry McGuiness
- Department of Immunology, Erasmus MC, Rotterdam, The Netherlands
| | - Sinead M. Gibney
- Department of Immunology, Erasmus MC, Rotterdam, The Netherlands
| | - Wouter Beumer
- Department of Immunology, Erasmus MC, Rotterdam, The Netherlands
| | | | | | - Andrew Harkin
- Trinity College Institute of Neuroscience, Trinity College, Dublin, Ireland
| | - Hemmo A. Drexhage
- Department of Immunology, Erasmus MC, Rotterdam, The Netherlands
- *Prof. Hemmo A. Drexhage, Department of Immunology, Na1105, Erasmus MC's Gravendijkwal 230 NL-3015 CE Rotterdam (The Netherlands) E-Mail
| |
Collapse
|
203
|
de Souza DF, Wartchow KM, Lunardi PS, Brolese G, Tortorelli LS, Batassini C, Biasibetti R, Gonçalves CA. Changes in Astroglial Markers in a Maternal Immune Activation Model of Schizophrenia in Wistar Rats are Dependent on Sex. Front Cell Neurosci 2015; 9:489. [PMID: 26733814 PMCID: PMC4689875 DOI: 10.3389/fncel.2015.00489] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 12/03/2015] [Indexed: 01/26/2023] Open
Abstract
Data from epidemiological studies suggest that prenatal exposure to bacterial and viral infection is an important environmental risk factor for schizophrenia. The maternal immune activation (MIA) animal model is used to study how an insult directed at the maternal host can have adverse effects on the fetus, leading to behavioral and neurochemical changes later in life. We evaluated whether the administration of LPS to rat dams during late pregnancy affects astroglial markers (S100B and GFAP) of the offspring in later life. The frontal cortex and hippocampus were compared in male and female offspring on postnatal days (PND) 30 and 60. The S100B protein exhibited an age-dependent pattern of expression, being increased in the frontal cortex and hippocampus of the MIA group at PND 60, while at PND 30, male rats presented increased S100B levels only in the frontal cortex. Considering that S100B secretion is reduced by elevation of glutamate levels, we may hypothesize that this early increment in frontal cortex tissue of males is associated with elevated extracellular levels of glutamate and glutamatergic hypofunction, an alteration commonly associated with SCZ pathology. Moreover, we also found augmented GFAP in the frontal cortex of the LPS group at PND 30, but not in the hippocampus. Taken together data indicate that astroglial changes induced by MIA are dependent on sex and brain region and that these changes could reflect astroglial dysfunction. Such alterations may contribute to our understanding of the abnormal neuronal connectivity and developmental aspects of SCZ and other psychiatric disorders.
Collapse
Affiliation(s)
- Daniela F de Souza
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul Porto Alegre, Brazil
| | - Krista M Wartchow
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul Porto Alegre, Brazil
| | - Paula S Lunardi
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul Porto Alegre, Brazil
| | - Giovana Brolese
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul Porto Alegre, Brazil
| | - Lucas S Tortorelli
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul Porto Alegre, Brazil
| | - Cristiane Batassini
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul Porto Alegre, Brazil
| | - Regina Biasibetti
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul Porto Alegre, Brazil
| | - Carlos-Alberto Gonçalves
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul Porto Alegre, Brazil
| |
Collapse
|
204
|
Elsayed M, Magistretti PJ. A New Outlook on Mental Illnesses: Glial Involvement Beyond the Glue. Front Cell Neurosci 2015; 9:468. [PMID: 26733803 PMCID: PMC4679853 DOI: 10.3389/fncel.2015.00468] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 11/16/2015] [Indexed: 12/27/2022] Open
Abstract
Mental illnesses have long been perceived as the exclusive consequence of abnormalities in neuronal functioning. Until recently, the role of glial cells in the pathophysiology of mental diseases has largely been overlooked. However recently, multiple lines of evidence suggest more diverse and significant functions of glia with behavior-altering effects. The newly ascribed roles of astrocytes, oligodendrocytes and microglia have led to their examination in brain pathology and mental illnesses. Indeed, abnormalities in glial function, structure and density have been observed in postmortem brain studies of subjects diagnosed with mental illnesses. In this review, we discuss the newly identified functions of glia and highlight the findings of glial abnormalities in psychiatric disorders. We discuss these preclinical and clinical findings implicating the involvement of glial cells in mental illnesses with the perspective that these cells may represent a new target for treatment.
Collapse
Affiliation(s)
- Maha Elsayed
- Laboratory of Neuroenergetics and Cellular Dynamics, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne Lausanne, Switzerland
| | - Pierre J Magistretti
- Laboratory of Neuroenergetics and Cellular Dynamics, Brain Mind Institute, Ecole Polytechnique Fédérale de LausanneLausanne, Switzerland; Division of Biological and Environmental Sciences and Engineering, King Abdullah University of Science and TechnologyThuwal, Saudi Arabia; Department of Psychiatry, Center for Psychiatric Neurosciences, University of LausanneLausanne, Switzerland
| |
Collapse
|
205
|
van Buel EM, Bosker FJ, van Drunen J, Strijker J, Douwenga W, Klein HC, Eisel ULM. Electroconvulsive seizures (ECS) do not prevent LPS-induced behavioral alterations and microglial activation. J Neuroinflammation 2015; 12:232. [PMID: 26654099 PMCID: PMC4676811 DOI: 10.1186/s12974-015-0454-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 11/26/2015] [Indexed: 12/19/2022] Open
Abstract
Background Long-term neuroimmune activation is a common finding in major depressive disorder (MDD). Literature suggests a dual effect of electroconvulsive therapy (ECT), a highly effective treatment strategy for MDD, on neuroimmune parameters: while ECT acutely increases inflammatory parameters, such as serum levels of pro-inflammatory cytokines, there is evidence to suggest that repeated ECT sessions eventually result in downregulation of the inflammatory response. We hypothesized that this might be due to ECT-induced attenuation of microglial activity upon inflammatory stimuli in the brain. Methods Adult male C57Bl/6J mice received a series of ten electroconvulsive seizures (ECS) or sham shocks, followed by an intracerebroventricular (i.c.v.) lipopolysaccharide (LPS) or phosphate-buffered saline (PBS) injection. Brains were extracted and immunohistochemically stained for the microglial marker ionized calcium-binding adaptor molecule 1 (Iba1). In addition, a sucrose preference test and an open-field test were performed to quantify behavioral alterations. Results LPS induced a short-term reduction in sucrose preference, which normalized within 3 days. In addition, LPS reduced the distance walked in the open field and induced alterations in grooming and rearing behavior. ECS did not affect any of these parameters. Phenotypical analysis of microglia demonstrated an LPS-induced increase in microglial activity ranging from 84 to 213 % in different hippocampal regions (CA3 213 %; CA1 84 %; dentate gyrus 131 %; and hilus 123 %). ECS-induced alterations in microglial activity were insignificant, ranging from −2.6 to 14.3 % in PBS-injected mice and from −20.2 to 6.6 % in LPS-injected mice. Conclusions We were unable to demonstrate an effect of ECS on LPS-induced microglial activity or behavioral alterations. Electronic supplementary material The online version of this article (doi:10.1186/s12974-015-0454-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- E M van Buel
- Department of Molecular Neurobiology, Groningen Institute of Evolutionary Life Sciences, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, Netherlands. .,Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
| | - F J Bosker
- Department of Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
| | - J van Drunen
- Department of Molecular Neurobiology, Groningen Institute of Evolutionary Life Sciences, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, Netherlands.
| | - J Strijker
- Department of Molecular Neurobiology, Groningen Institute of Evolutionary Life Sciences, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, Netherlands.
| | - W Douwenga
- Department of Molecular Neurobiology, Groningen Institute of Evolutionary Life Sciences, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, Netherlands.
| | - H C Klein
- Department of Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, Netherlands. .,Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
| | - U L M Eisel
- Department of Molecular Neurobiology, Groningen Institute of Evolutionary Life Sciences, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, Netherlands. .,Department of Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
| |
Collapse
|
206
|
Gariup M, Gonzalez A, Lázaro L, Torres F, Serra-Pagès C, Morer A. IL-8 and the innate immunity as biomarkers in acute child and adolescent psychopathology. Psychoneuroendocrinology 2015; 62:233-42. [PMID: 26325024 DOI: 10.1016/j.psyneuen.2015.08.017] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 08/18/2015] [Accepted: 08/19/2015] [Indexed: 11/17/2022]
Abstract
OBJECTIVE The role of inflammation in psychopathology has received great attention over the past decades. Immune system dysfunction and altered cytokine levels have been reported in most psychiatric disorders in adults. Few data are available regarding children and adolescents (C&A), or regarding the relationship between cytokine levels and psychosocial stress. This study investigates the profile of the most described cytokines in a sample of C&A inpatients affected by an acute psychiatric condition requiring hospitalization, in comparison with healthy subjects, as well as possible associations between psychosocial stressors and psychopathology and/or cytokine concentrations. METHODS Patients with a diagnosis of Affective, Anxiety, Adjustment, Psychotic, Obsessive-Compulsive, Tic or Tourette Disorders were consecutively recruited from our clinic between June 2010 and February 2012. Controls were recruited from the same geographic area. All subjects were between 8 and 17 years old. Twelve cytokines are compared: interleukin (IL)-1β, IL-2, IL-4, IL-5, IL-6, IL-8, IL_10, granulocyte-macrophage colony-stimulating factor (GM-CSF), interferon (IFN)-γ, tumor necrosis factor (TNF)-α, IFN-γ-induced protein-10 (IP-10), monocyte chemoattractant protein (MCP)-1. Psychosocial stress was measured through the Stressful Life Events Scale, Child and Parents versions (SLES-C and SLES-P) and the evaluation of the family integrity. RESULTS One hundred and eleven subjects (77C&A inpatients and 34 healthy controls), of which 54 were males (49%), with a median (interquartile range) age of 16 (13.7-17.3) years, were included in this study. IL-1β, IL6, IL8, IP-10, MCP-1 and monocytes were found to be significantly higher in the patient group (p<0.05). Differences were confirmed when adjusting by BMI, age, gender and drug intake at admission for all cytokines except MCP-1. IL-8 and IL-1β were also higher throughout the different diagnostic categories, than in control group (p<0.05). Stress measures were higher in patients. A significant correlation was found between stress measured by the SLES and some inflammatory markers: SLES_C with IL-1β, IL-8, MCP-1, and SLES_P with IL-1β and monocytes absolute and relative counts (Spearman's r between 0.219 and 0.297, p<0.05). Logistic regression identified the following variables as independent predictors of the patient condition, (odds ratio per quartile, p-value): IL8 (1, 0.9, 12.1, 32.0, p=0.044), IP10 (1, 14.1, 2.5, 3.7, p=0.044), monocyte absolute count (1, 1.1, 6.0, 19.4, p=0.030). CONCLUSIONS Results show elevated inflammation markers from the innate immune system across C&A acute psychiatric diagnosis, and suggest a link between psychopathology, inflammation and stress. Inflammatory markers resulted predictors of patient status. These exploratory results are coherent with current psychoneuroimmunology and neurodevelopmental investigations.
Collapse
Affiliation(s)
- Maria Gariup
- Psychiatry Department, Kolding Hospital, Skovvejen, Kolding 6000, Denmark; Medicine Faculty, Barcelona University, C/ Villarroel 170, Barcelona 08036, Spain.
| | - Azucena Gonzalez
- Immunonlogy Dept., Hospital Clínic, C/ Villarroel 170, Barcelona 08036, Spain.
| | - Luisa Lázaro
- Medicine Faculty, Barcelona University, C/ Villarroel 170, Barcelona 08036, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Child and Adolescent Psychiatry, Hospital Clínic, C/ Villarroel 170, Barcelona 08036, Spain; CIBERSAM (Centro de Investigación Biomédica en Red de Salud Mental), Group G04, Hospital Clínic, C/ Villarroel 170, Barcelona 08036, Spain.
| | - Ferran Torres
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Biostatistics Unit, School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Carles Serra-Pagès
- Medicine Faculty, Barcelona University, C/ Villarroel 170, Barcelona 08036, Spain; Immunonlogy Dept., Hospital Clínic, C/ Villarroel 170, Barcelona 08036, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain.
| | - Astrid Morer
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Child and Adolescent Psychiatry, Hospital Clínic, C/ Villarroel 170, Barcelona 08036, Spain; CIBERSAM (Centro de Investigación Biomédica en Red de Salud Mental), Group G04, Hospital Clínic, C/ Villarroel 170, Barcelona 08036, Spain.
| |
Collapse
|
207
|
Demeestere D, Libert C, Vandenbroucke RE. Therapeutic implications of the choroid plexus-cerebrospinal fluid interface in neuropsychiatric disorders. Brain Behav Immun 2015; 50:1-13. [PMID: 26116435 DOI: 10.1016/j.bbi.2015.06.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 05/29/2015] [Accepted: 06/13/2015] [Indexed: 12/31/2022] Open
Abstract
The choroid plexus (CP) comprises an epithelial monolayer that forms an important physical, enzymatic and immunologic barrier, called the blood-cerebrospinal fluid barrier (BCSFB). It is a highly vascularized organ located in the brain ventricles that is key in maintaining brain homeostasis as it produces cerebrospinal fluid (CSF) and has other important secretory functions. Furthermore, the CP-CSF interface plays a putative role in neurogenesis and has been implicated in neuropsychiatric diseases such as the neurodevelopmental disorders schizophrenia and autism. A role for this CNS border was also implicated in sleep disturbances and chronic and/or severe stress, which are risk factors for the development of neuropsychiatric conditions. Understanding the mechanisms by which disturbance of the homeostasis at the CP-CSF interface is involved in these different chronic low-grade inflammatory diseases can give new insights into therapeutic strategies. Hence, this review discusses the different roles that have been suggested so far for the CP in these neuropsychiatric disorders, with special attention to potential therapeutic applications.
Collapse
Affiliation(s)
- Delphine Demeestere
- Inflammation Research Center, VIB, Technologiepark 927, B-9052 Zwijnaarde, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, B-9052 Zwijnaarde, Ghent, Belgium
| | - Claude Libert
- Inflammation Research Center, VIB, Technologiepark 927, B-9052 Zwijnaarde, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, B-9052 Zwijnaarde, Ghent, Belgium
| | - Roosmarijn E Vandenbroucke
- Inflammation Research Center, VIB, Technologiepark 927, B-9052 Zwijnaarde, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, B-9052 Zwijnaarde, Ghent, Belgium.
| |
Collapse
|
208
|
Diversity and plasticity of microglial cells in psychiatric and neurological disorders. Pharmacol Ther 2015; 154:21-35. [PMID: 26129625 DOI: 10.1016/j.pharmthera.2015.06.010] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 06/25/2015] [Indexed: 02/07/2023]
|
209
|
Bergon A, Belzeaux R, Comte M, Pelletier F, Hervé M, Gardiner EJ, Beveridge NJ, Liu B, Carr V, Scott RJ, Kelly B, Cairns MJ, Kumarasinghe N, Schall U, Blin O, Boucraut J, Tooney PA, Fakra E, Ibrahim EC. CX3CR1 is dysregulated in blood and brain from schizophrenia patients. Schizophr Res 2015; 168:434-43. [PMID: 26285829 DOI: 10.1016/j.schres.2015.08.010] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 08/05/2015] [Accepted: 08/06/2015] [Indexed: 12/31/2022]
Abstract
The molecular mechanisms underlying schizophrenia remain largely unknown. Although schizophrenia is a mental disorder, there is increasing evidence to indicate that inflammatory processes driven by diverse environmental factors play a significant role in its development. With gene expression studies having been conducted across a variety of sample types, e.g., blood and postmortem brain, it is possible to investigate convergent signatures that may reveal interactions between the immune and nervous systems in schizophrenia pathophysiology. We conducted two meta-analyses of schizophrenia microarray gene expression data (N=474) and non-psychiatric control (N=485) data from postmortem brain and blood. Then, we assessed whether significantly dysregulated genes in schizophrenia could be shared between blood and brain. To validate our findings, we selected a top gene candidate and analyzed its expression by RT-qPCR in a cohort of schizophrenia subjects stabilized by atypical antipsychotic monotherapy (N=29) and matched controls (N=31). Meta-analyses highlighted inflammation as the major biological process associated with schizophrenia and that the chemokine receptor CX3CR1 was significantly down-regulated in schizophrenia. This differential expression was also confirmed in our validation cohort. Given both the recent data demonstrating selective CX3CR1 expression in subsets of neuroimmune cells, as well as behavioral and neuropathological observations of CX3CR1 deficiency in mouse models, our results of reduced CX3CR1 expression adds further support for a role played by monocyte/microglia in the neurodevelopment of schizophrenia.
Collapse
Affiliation(s)
- Aurélie Bergon
- INSERM, TAGC UMR_S 1090, 13288 Marseille Cedex 09, France; Aix Marseille Université, TAGC UMR_S 1090, 13288 Marseille Cedex 09, France
| | - Raoul Belzeaux
- Aix Marseille Université, CNRS, CRN2M UMR 7286, 13344 Marseille Cedex 15, France; FondaMental, Fondation de Recherche et de Soins en Santé Mentale, 94000 Créteil, France; AP-HM, Hôpital Sainte Marguerite, Pôle de Psychiatrie Universitaire Solaris, 13009 Marseille, France
| | - Magali Comte
- Aix-Marseille Université, CNRS, Institut de Neurosciences de la Timone UMR 7289, 13005 Marseille, France
| | - Florence Pelletier
- Aix Marseille Université, CNRS, CRN2M UMR 7286, 13344 Marseille Cedex 15, France; FondaMental, Fondation de Recherche et de Soins en Santé Mentale, 94000 Créteil, France
| | - Mylène Hervé
- Aix Marseille Université, CNRS, CRN2M UMR 7286, 13344 Marseille Cedex 15, France; FondaMental, Fondation de Recherche et de Soins en Santé Mentale, 94000 Créteil, France
| | - Erin J Gardiner
- School of Biomedical Sciences and Pharmacy and School of Medicine and Public Health, Faculty of Health, The University of Newcastle, University Drive, Callaghan, NSW 2308 Australia; Centre for Translational Neuroscience and Mental Health, The University of Newcastle, Callaghan, NSW 2308 Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia; Schizophrenia Research Institute, Darlinghurst, NSW 2010 Australia
| | - Natalie J Beveridge
- School of Biomedical Sciences and Pharmacy and School of Medicine and Public Health, Faculty of Health, The University of Newcastle, University Drive, Callaghan, NSW 2308 Australia; Centre for Translational Neuroscience and Mental Health, The University of Newcastle, Callaghan, NSW 2308 Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia; Schizophrenia Research Institute, Darlinghurst, NSW 2010 Australia
| | - Bing Liu
- School of Biomedical Sciences and Pharmacy and School of Medicine and Public Health, Faculty of Health, The University of Newcastle, University Drive, Callaghan, NSW 2308 Australia; Centre for Translational Neuroscience and Mental Health, The University of Newcastle, Callaghan, NSW 2308 Australia; Kids Cancer Alliance, Cancer Institute NSW, Sydney, Australia
| | - Vaughan Carr
- Schizophrenia Research Institute, Darlinghurst, NSW 2010 Australia; School of Psychiatry, University of New South Wales, Randwick, NSW 2301, Australia; Department of Psychiatry, Monash University, Clayton, VIC 3168, Australia
| | - Rodney J Scott
- School of Biomedical Sciences and Pharmacy and School of Medicine and Public Health, Faculty of Health, The University of Newcastle, University Drive, Callaghan, NSW 2308 Australia; Centre for Translational Neuroscience and Mental Health, The University of Newcastle, Callaghan, NSW 2308 Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia; Schizophrenia Research Institute, Darlinghurst, NSW 2010 Australia
| | - Brian Kelly
- School of Biomedical Sciences and Pharmacy and School of Medicine and Public Health, Faculty of Health, The University of Newcastle, University Drive, Callaghan, NSW 2308 Australia; Centre for Translational Neuroscience and Mental Health, The University of Newcastle, Callaghan, NSW 2308 Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Murray J Cairns
- School of Biomedical Sciences and Pharmacy and School of Medicine and Public Health, Faculty of Health, The University of Newcastle, University Drive, Callaghan, NSW 2308 Australia; Centre for Translational Neuroscience and Mental Health, The University of Newcastle, Callaghan, NSW 2308 Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia; Schizophrenia Research Institute, Darlinghurst, NSW 2010 Australia
| | - Nishantha Kumarasinghe
- School of Biomedical Sciences and Pharmacy and School of Medicine and Public Health, Faculty of Health, The University of Newcastle, University Drive, Callaghan, NSW 2308 Australia; Centre for Translational Neuroscience and Mental Health, The University of Newcastle, Callaghan, NSW 2308 Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia; Schizophrenia Research Institute, Darlinghurst, NSW 2010 Australia; University of Sri Jayewardenepura, Nugegoda, Sri Lanka; National Institute of Mental Health, Angoda, Sri Lanka
| | - Ulrich Schall
- School of Biomedical Sciences and Pharmacy and School of Medicine and Public Health, Faculty of Health, The University of Newcastle, University Drive, Callaghan, NSW 2308 Australia; Centre for Translational Neuroscience and Mental Health, The University of Newcastle, Callaghan, NSW 2308 Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia; Schizophrenia Research Institute, Darlinghurst, NSW 2010 Australia
| | - Olivier Blin
- CIC-UPCET et Pharmacologie Clinique, Hôpital de la Timone, 13005 Marseille, France
| | - José Boucraut
- Aix Marseille Université, CNRS, CRN2M UMR 7286, 13344 Marseille Cedex 15, France; FondaMental, Fondation de Recherche et de Soins en Santé Mentale, 94000 Créteil, France
| | - Paul A Tooney
- School of Biomedical Sciences and Pharmacy and School of Medicine and Public Health, Faculty of Health, The University of Newcastle, University Drive, Callaghan, NSW 2308 Australia; Centre for Translational Neuroscience and Mental Health, The University of Newcastle, Callaghan, NSW 2308 Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia; Schizophrenia Research Institute, Darlinghurst, NSW 2010 Australia
| | - Eric Fakra
- Aix-Marseille Université, CNRS, Institut de Neurosciences de la Timone UMR 7289, 13005 Marseille, France; CHU de Saint-Etienne, Pôle de Psychiatrie, 42100 Saint-Etienne, France
| | - El Chérif Ibrahim
- Aix Marseille Université, CNRS, CRN2M UMR 7286, 13344 Marseille Cedex 15, France; FondaMental, Fondation de Recherche et de Soins en Santé Mentale, 94000 Créteil, France.
| |
Collapse
|
210
|
Becking K, Haarman BCM, van der Lek RFR, Grosse L, Nolen WA, Claes S, Drexhage HA, Schoevers RA. Inflammatory monocyte gene expression: trait or state marker in bipolar disorder? Int J Bipolar Disord 2015; 3:20. [PMID: 26381439 PMCID: PMC4574035 DOI: 10.1186/s40345-015-0037-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 09/03/2015] [Indexed: 01/14/2023] Open
Abstract
Background This study aimed to examine whether inflammatory gene expression was a trait or a state marker in patients with bipolar disorder (BD). Methods 69 healthy controls (HC), 82 euthymic BD patients and 8 BD patients with a mood episode (7 depressed, 1 manic) were included from the MOODINFLAME study. Six of the eight patients who had a mood episode were also investigated when they were euthymic (6 of the 82 euthymic patients). Of these participants the expression of 35 inflammatory genes was determined in monocytes using quantitative-polymerase chain reaction, of which a total gene expression score was calculated as well as a gene expression score per sub-cluster. Results There were no significant differences in inflammatory monocyte gene expression between healthy controls and euthymic patients. Patients experiencing a mood episode, however, had a significantly higher total gene expression score (10.63 ± 2.58) compared to healthy controls (p = .004) and euthymic patients (p = .009), as well as when compared to their own scores when they were euthymic (p = .02). This applied in particular for the sub-cluster 1 gene expression score, but not for the sub-cluster 2 gene expression score. Conclusions Our study indicates that in BD inflammatory monocyte, gene expression is especially elevated while in a mood episode compared to being euthymic.
Collapse
Affiliation(s)
- K Becking
- ICPE/UCP/Triade (CC.72), Interdisciplinary Center Psychopathology and Emotion Regulation (ICPE), University Medical Center Groningen, University of Groningen, PO Box 30001, 9700 RB, Groningen, The Netherlands. .,Department of Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | - B C M Haarman
- Department of Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands. .,Radiology Morphological Solutions, Berkel en Rodenrijs, The Netherlands.
| | - R F Riemersma van der Lek
- ICPE/UCP/Triade (CC.72), Interdisciplinary Center Psychopathology and Emotion Regulation (ICPE), University Medical Center Groningen, University of Groningen, PO Box 30001, 9700 RB, Groningen, The Netherlands. .,Department of Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | - L Grosse
- Department of Psychiatry, University of Münster, Münster, Germany.
| | - W A Nolen
- Department of Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | - S Claes
- Department of Psychiatry, University of Leuven (KU Leuven), Louvain, Belgium.
| | - H A Drexhage
- Department of Immunology, Erasmus MC, Rotterdam, The Netherlands.
| | - R A Schoevers
- ICPE/UCP/Triade (CC.72), Interdisciplinary Center Psychopathology and Emotion Regulation (ICPE), University Medical Center Groningen, University of Groningen, PO Box 30001, 9700 RB, Groningen, The Netherlands. .,Department of Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
211
|
Cytokines and chemokines as biomarkers of ethanol-induced neuroinflammation and anxiety-related behavior: role of TLR4 and TLR2. Neuropharmacology 2015; 89:352-9. [PMID: 25446779 DOI: 10.1016/j.neuropharm.2014.10.014] [Citation(s) in RCA: 146] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 09/03/2014] [Accepted: 10/14/2014] [Indexed: 12/23/2022]
Abstract
Recent evidence supports the influence of neuroimmune system activation on behavior. We have demonstrated that ethanol activates the innate immune system by stimulating toll-like receptor 4 (TLR4) signaling in glial cells, which triggers the release of inflammatory mediators and causes neuroinflammation. The present study aimed to evaluate whether the ethanol-induced up-regulation of cytokines and chemokines is associated with anxiety-related behavior, 24 h after ethanol removal, and if TLR4 or TLR2 is involved in these effects. We used WT, TLR4-KO and TLR2-KO mice treated with alcohol for 5 months to show that chronic ethanol consumption increases the levels of cytokines (IL-1β, IL-17, TNF-α) and chemokines (MCP-1, MIP-1α, CX3CL1) in the striatum and serum (MCP-1, MIP-1α, CX3CL1) of WT mice. Alcohol deprivation for 24 h induces IFN-γ levels in the striatum and maintains high levels of some cytokines (IL-1β, IL-17) and chemokines (MIP-1α, CX3CL1) in this brain region. The latter events were associated with an increase in anxiogenic-related behavior, as evaluated by the dark and light box and the elevated plus maze tests. Notably, mice lacking TLR4 or TLR2 receptors are largely protected against ethanol-induced cytokine and chemokine release, and behavioral associated effects during alcohol abstinence. These data support the role of TLR4 and TLR2 responses in neuroinflammation and in anxiogenic-related behavior effects during ethanol deprivation, and also provide evidence that chemokines and cytokines can be biomarkers of ethanol-induced neuroimmune response.
Collapse
|
212
|
Adaptive Immunity in Schizophrenia: Functional Implications of T Cells in the Etiology, Course and Treatment. J Neuroimmune Pharmacol 2015; 10:610-9. [PMID: 26162591 DOI: 10.1007/s11481-015-9626-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 07/03/2015] [Indexed: 12/21/2022]
Abstract
Schizophrenia is a severe and highly complex neurodevelopmental disorder with an unknown etiopathology. Recently, immunopathogenesis has emerged as one of the most compelling etiological models of schizophrenia. Over the past few years considerable research has been devoted to the role of innate immune responses in schizophrenia. The findings of such studies have helped to conceptualize schizophrenia as a chronic low-grade inflammatory disorder. Although the contribution of adaptive immune responses has also been emphasized, however, the precise role of T cells in the underlying neurobiological pathways of schizophrenia is yet to be ascertained comprehensively. T cells have the ability to infiltrate brain and mediate neuro-immune cross-talk. Conversely, the central nervous system and the neurotransmitters are capable of regulating the immune system. Neurotransmitter like dopamine, implicated widely in schizophrenia risk and progression can modulate the proliferation, trafficking and functions of T cells. Within brain, T cells activate microglia, induce production of pro-inflammatory cytokines as well as reactive oxygen species and subsequently lead to neuroinflammation. Importantly, such processes contribute to neuronal injury/death and are gradually being implicated as mediators of neuroprogressive changes in schizophrenia. Antipsychotic drugs, commonly used to treat schizophrenia are also known to affect adaptive immune system; interfere with the differentiation and functions of T cells. This understanding suggests a pivotal role of T cells in the etiology, course and treatment of schizophrenia and forms the basis of this review.
Collapse
|
213
|
Theoharides TC, Stewart JM, Hatziagelaki E, Kolaitis G. Brain "fog," inflammation and obesity: key aspects of neuropsychiatric disorders improved by luteolin. Front Neurosci 2015; 9:225. [PMID: 26190965 PMCID: PMC4490655 DOI: 10.3389/fnins.2015.00225] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 06/10/2015] [Indexed: 12/20/2022] Open
Abstract
Brain "fog" is a constellation of symptoms that include reduced cognition, inability to concentrate and multitask, as well as loss of short and long term memory. Brain "fog" characterizes patients with autism spectrum disorders (ASDs), celiac disease, chronic fatigue syndrome, fibromyalgia, mastocytosis, and postural tachycardia syndrome (POTS), as well as "minimal cognitive impairment," an early clinical presentation of Alzheimer's disease (AD), and other neuropsychiatric disorders. Brain "fog" may be due to inflammatory molecules, including adipocytokines and histamine released from mast cells (MCs) further stimulating microglia activation, and causing focal brain inflammation. Recent reviews have described the potential use of natural flavonoids for the treatment of neuropsychiatric and neurodegenerative diseases. The flavone luteolin has numerous useful actions that include: anti-oxidant, anti-inflammatory, microglia inhibition, neuroprotection, and memory increase. A liposomal luteolin formulation in olive fruit extract improved attention in children with ASDs and brain "fog" in mastocytosis patients. Methylated luteolin analogs with increased activity and better bioavailability could be developed into effective treatments for neuropsychiatric disorders and brain "fog."
Collapse
Affiliation(s)
- Theoharis C. Theoharides
- Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Integrative Physiology and Pathobiology, Tufts University School of MedicineBoston, MA, USA
- Departments of Internal Medicine, Tufts University School of Medicine and Tufts Medical CenterBoston, MA, USA
- Psychiatry, Tufts University School of Medicine and Tufts Medical CenterBoston, MA, USA
- Sackler School of Graduate Biomedical Sciences, Tufts University School of MedicineBoston, MA, USA
| | - Julia M. Stewart
- Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Integrative Physiology and Pathobiology, Tufts University School of MedicineBoston, MA, USA
| | - Erifili Hatziagelaki
- Second Department of Internal Medicine, Attikon General Hospital, Athens Medical SchoolAthens, Greece
| | - Gerasimos Kolaitis
- Department of Child Psychiatry, University of Athens Medical School, Aghia Sophia Children's HospitalAthens, Greece
| |
Collapse
|
214
|
Jakobsson J, Bjerke M, Sahebi S, Isgren A, Ekman CJ, Sellgren C, Olsson B, Zetterberg H, Blennow K, Pålsson E, Landén M. Monocyte and microglial activation in patients with mood-stabilized bipolar disorder. J Psychiatry Neurosci 2015; 40:250-8. [PMID: 25768030 PMCID: PMC4478058 DOI: 10.1503/jpn.140183] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Bipolar disorder is associated with medical comorbidities that have been linked to systemic inflammatory mechanisms. There is, however, limited evidence supporting a role of neuroinflammation in bipolar disorder. Here we tested whether microglial activation and associated tissue remodelling processes are related to bipolar disorder by analyzing markers in cerebrospinal fluid (CSF) and serum from patients and healthy controls. METHODS Serum was sampled from euthymic patients with bipolar disorder and healthy controls, and CSF was sampled from a large subset of these individuals. The levels of monocyte chemoattractant protein-1 (MCP-1), YKL-40, soluble cluster of differentiation 14 (sCD14), tissue inhibitor of metalloproteinases-1 (TIMP-1) and tissue inhibitor of metalloproteinases-2 (TIMP-2), were measured, and we adjusted comparisons between patients and controls for confounding factors. RESULTS We obtained serum samples from 221 patients and 112 controls and CSF samples from 125 patients and 87 controls. We found increased CSF levels of MCP-1 and YKL-40 and increased serum levels of sCD14 and YKL-40 in patients compared with controls; these differences remained after controlling for confounding factors, such as age, sex, smoking, blood-CSF barrier function, acute-phase proteins and body mass index. The CSF levels of MCP-1 and YKL-40 correlated with the serum levels, whereas the differences between patients and controls in CSF levels of MCP-1 and YKL-40 were independent of serum levels. LIMITATIONS The cross-sectional study design precludes conclusions about causality. CONCLUSION Our results suggest that both neuroinflammatory and systemic inflammatory processes are involved in the pathophysiology of bipolar disorder. Importantly, markers of immunological processes in the brain were independent of peripheral immunological activity.
Collapse
Affiliation(s)
- Joel Jakobsson
- Correspondence to: Joel Jakobsson, Sahlgrenska University hospital, Blå Stråket 15, floor 3, SE-413 45 Gothenburg, Sweden;
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
215
|
Hope S, Hoseth E, Dieset I, Mørch RH, Aas M, Aukrust P, Djurovic S, Melle I, Ueland T, Agartz I, Ueland T, Westlye LT, Andreassen OA. Inflammatory markers are associated with general cognitive abilities in schizophrenia and bipolar disorder patients and healthy controls. Schizophr Res 2015; 165:188-94. [PMID: 25956633 DOI: 10.1016/j.schres.2015.04.004] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 03/25/2015] [Accepted: 04/07/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND The mechanisms underlying cognitive impairment in schizophrenia and bipolar disorders are largely unknown. Immune abnormalities have been found in both disorders, and inflammatory mediators may play roles in cognitive function. We investigated if inflammatory markers are associated with general cognitive abilities. METHODS Participants with schizophrenia spectrum (N=121) and bipolar spectrum (N=111) disorders and healthy controls (N=241) were included. General intellectual abilities were assessed using the Wechsler Abbreviated Scale of Intelligence (WASI). Serum concentrations of the following immune markers were measured: Soluble tumor necrosis factor receptor 1 (sTNF-R1), interleukin 1 receptor antagonist (IL-1Ra), osteoprotegerin, von Willebrand factor, C-reactive protein, interleukin-6 and CD40 ligand. RESULTS After adjusting for age, sex and diagnostic group, significant negative associations with general cognitive function were found for sTNF-R1 (p=2×10(-5)), IL-1Ra (p=0.002) and sCD40 ligand (p=0.003). Among patients, the associations remained significant (p=0.006, p=0.005 and p=0.02) after adjusting for possible confounders including education, smoking, psychotic and affective symptoms, body mass index, cortisol, medication and time of blood sampling. Subgroup analysis, showed that general cognitive abilities were significantly associated with IL-1Ra and sTNF-R1 in schizophrenia patients, with sCD40L and IL-1Ra in bipolar disorder patients and with sTNF-R1 in healthy controls. CONCLUSION The study shows significant negative associations between inflammatory markers and general cognitive abilities after adjusting for possible confounders. The findings strongly support a role for inflammation in the neurophysiology of cognitive impairment.
Collapse
Affiliation(s)
- Sigrun Hope
- NORMENT, KG Jebsen Centre for Psychosis Research, University of Oslo, Oslo, Norway; Department of Neuro Habilitation, Oslo University Hospital, Ullevål, Oslo, Norway
| | - Eva Hoseth
- NORMENT, KG Jebsen Centre for Psychosis Research, University of Oslo, Oslo, Norway; Division of Mental Health and Addiction, Oslo University Hospital, Ullevål, Oslo, Norway
| | - Ingrid Dieset
- NORMENT, KG Jebsen Centre for Psychosis Research, University of Oslo, Oslo, Norway; Division of Mental Health and Addiction, Oslo University Hospital, Ullevål, Oslo, Norway
| | - Ragni H Mørch
- NORMENT, KG Jebsen Centre for Psychosis Research, University of Oslo, Oslo, Norway; Division of Mental Health and Addiction, Oslo University Hospital, Ullevål, Oslo, Norway
| | - Monica Aas
- NORMENT, KG Jebsen Centre for Psychosis Research, University of Oslo, Oslo, Norway; Division of Mental Health and Addiction, Oslo University Hospital, Ullevål, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway; Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital, Rikshospitalet, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway; KG Jebsen Inflammatory Research Center, University of Oslo, Oslo, Norway
| | - Srdjan Djurovic
- NORMENT, KG Jebsen Centre for Psychosis Research, University of Oslo, Oslo, Norway
| | - Ingrid Melle
- NORMENT, KG Jebsen Centre for Psychosis Research, University of Oslo, Oslo, Norway; Division of Mental Health and Addiction, Oslo University Hospital, Ullevål, Oslo, Norway
| | - Torill Ueland
- NORMENT, KG Jebsen Centre for Psychosis Research, University of Oslo, Oslo, Norway; Department of Psychology, University of Oslo, Oslo, Norway
| | - Ingrid Agartz
- NORMENT, KG Jebsen Centre for Psychosis Research, University of Oslo, Oslo, Norway; Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway; KG Jebsen Inflammatory Research Center, University of Oslo, Oslo, Norway
| | - Lars T Westlye
- NORMENT, KG Jebsen Centre for Psychosis Research, University of Oslo, Oslo, Norway; Division of Mental Health and Addiction, Oslo University Hospital, Ullevål, Oslo, Norway; Department of Psychology, University of Oslo, Oslo, Norway
| | - Ole A Andreassen
- NORMENT, KG Jebsen Centre for Psychosis Research, University of Oslo, Oslo, Norway; Division of Mental Health and Addiction, Oslo University Hospital, Ullevål, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
216
|
Deleidi M, Jäggle M, Rubino G. Immune aging, dysmetabolism, and inflammation in neurological diseases. Front Neurosci 2015; 9:172. [PMID: 26089771 PMCID: PMC4453474 DOI: 10.3389/fnins.2015.00172] [Citation(s) in RCA: 195] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 04/28/2015] [Indexed: 12/17/2022] Open
Abstract
As we age, the immune system undergoes a process of senescence accompanied by the increased production of proinflammatory cytokines, a chronic subclinical condition named as “inflammaging”. Emerging evidence from human and experimental models suggest that immune senescence also affects the central nervous system and promotes neuronal dysfunction, especially within susceptible neuronal populations. In this review we discuss the potential role of immune aging, inflammation and metabolic derangement in neurological diseases. The discovery of novel therapeutic strategies targeting age-linked inflammation may promote healthy brain aging and the treatment of neurodegenerative as well as neuropsychiatric disorders.
Collapse
Affiliation(s)
- Michela Deleidi
- Department of Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Hertie Institute for Clinical Brain Research, University of Tübingen Tübingen, Germany
| | - Madeline Jäggle
- Department of Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Hertie Institute for Clinical Brain Research, University of Tübingen Tübingen, Germany
| | - Graziella Rubino
- Department of Internal Medicine II, Center for Medical Research, University of Tübingen Tübingen, Germany
| |
Collapse
|
217
|
Amoruso A, Bardelli C, Cattaneo CI, Fresu LG, Manzetti E, Brunelleschi S. Neurokinin (NK)-1 receptor expression in monocytes from bipolar disorder patients: a pilot study. J Affect Disord 2015; 178:188-92. [PMID: 25841180 DOI: 10.1016/j.jad.2015.03.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 03/05/2015] [Accepted: 03/05/2015] [Indexed: 11/28/2022]
Abstract
BACKGROUND Neurokinin 1 receptors (NK-1R) have been involved in several psychiatric disorders including major depression, but less is known for bipolar disorder (BD). METHOD We compared NK-1R expression and Substance P (SP) ability to induce NF-κB activation in monocytes from BD patients and healthy donors (HD), also looking for the effects of tobacco smoke. After informed written consent, 20 euthymic BD patients, either bipolar type 1 (BDI) or type 2 (BDII), and 14 age-matched healthy donors (HD) were enrolled. NK-1R expression in monocytes was evaluated by Western blot and expressed as the ratio between NK-1R and Na(+)/K(+)-ATPase protein expressions. NF-κB activation was assessed by measuring the nuclear content of the p50 subunit (ELISA kit). RESULTS NK-1R expression was significantly reduced (P<0.001) in monocytes from BD patients as compared to HD, with no major differences between BDI and BDII patients. Tobacco smoke enhanced NK-1R expression in HD, but not in BD patients. Un-stimulated monocytes from BD patients presented a constitutively higher (P<0.05) content of nuclear p50 subunit as compared to HD. SP and an NK-1R agonist induced NF-κB activation, with a higher effect in HD: this effect was receptor-mediated as it was abrogated by an NK-1R antagonist. LIMITATIONS As a pilot study enrolling 20 BD patients, an obvious limitation is the sample size. CONCLUSIONS Our results show the existence of a relevant alteration in NK-1R expression in BD patients and further suggest SP involvement in BD, so improving our understanding of the underlying mechanisms of this disease.
Collapse
Affiliation(s)
- Angela Amoruso
- Department of Health Sciences, School of Medicine, University "A. Avogadro", Via Solaroli, 17, 28100 Novara, Italy
| | - Claudio Bardelli
- Department of Health Sciences, School of Medicine, University "A. Avogadro", Via Solaroli, 17, 28100 Novara, Italy
| | - Carlo Ignazio Cattaneo
- Department of Mental Health, ASL NO, Centre of Mental Health, Viale Zoppis, 8, 28021 Borgomanero, Novara, Italy
| | - Luigia Grazia Fresu
- Department of Health Sciences, School of Medicine, University "A. Avogadro", Via Solaroli, 17, 28100 Novara, Italy
| | - Elena Manzetti
- Department of Mental Health, ASL NO, Centre of Mental Health, Viale Zoppis, 8, 28021 Borgomanero, Novara, Italy
| | - Sandra Brunelleschi
- Department of Health Sciences, School of Medicine, University "A. Avogadro", Via Solaroli, 17, 28100 Novara, Italy; Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), School of Medicine, Novara, Italy.
| |
Collapse
|
218
|
Réus GZ, Fries GR, Stertz L, Badawy M, Passos IC, Barichello T, Kapczinski F, Quevedo J. The role of inflammation and microglial activation in the pathophysiology of psychiatric disorders. Neuroscience 2015; 300:141-54. [PMID: 25981208 DOI: 10.1016/j.neuroscience.2015.05.018] [Citation(s) in RCA: 450] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 04/22/2015] [Accepted: 05/07/2015] [Indexed: 12/30/2022]
Abstract
Psychiatric disorders, including major depressive disorder (MDD), bipolar disorder (BD) and schizophrenia, affect a significant percentage of the world population. These disorders are associated with educational difficulties, decreased productivity and reduced quality of life, but their underlying pathophysiological mechanisms are not fully elucidated. Recently, studies have suggested that psychiatric disorders could be considered as inflammatory disorders, even though the exact mechanisms underlying this association are not known. An increase in inflammatory response and oxidative stress may lead to inflammation, which in turn can stimulate microglia in the brain. Microglial activation is roused by the M1 phenotype, which is associated with an increase in interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α). On the contrary, M2 phenotype is associated with a release of anti-inflammatory cytokines. Thus, it is possible that the inflammatory response from microglial activation can contribute to brain pathology, as well as influence treatment responses. This review will highlight the role of inflammation in the pathophysiology of psychiatric disorders, such as MDD, BD, schizophrenia, and autism. More specifically, the role of microglial activation and associated molecular cascades will also be discussed as a means by which these neuroinflammatory mechanisms take place, when appropriate.
Collapse
Affiliation(s)
- G Z Réus
- Center for Translational Psychiatry, Department of Psychiatry and Behavioral Sciences, The University of Texas Medical School at Houston, Houston, TX, USA; Laboratório de Neurociências, Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil.
| | - G R Fries
- Center for Translational Psychiatry, Department of Psychiatry and Behavioral Sciences, The University of Texas Medical School at Houston, Houston, TX, USA; Molecular Psychiatry Unit and National Science and Technology Institute for Translational Medicine (INCT-TM), Hospital de Clínicas de Porto Alegre (HCPA), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - L Stertz
- Center for Translational Psychiatry, Department of Psychiatry and Behavioral Sciences, The University of Texas Medical School at Houston, Houston, TX, USA; Molecular Psychiatry Unit and National Science and Technology Institute for Translational Medicine (INCT-TM), Hospital de Clínicas de Porto Alegre (HCPA), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - M Badawy
- Center for Translational Psychiatry, Department of Psychiatry and Behavioral Sciences, The University of Texas Medical School at Houston, Houston, TX, USA
| | - I C Passos
- Center for Translational Psychiatry, Department of Psychiatry and Behavioral Sciences, The University of Texas Medical School at Houston, Houston, TX, USA; Molecular Psychiatry Unit and National Science and Technology Institute for Translational Medicine (INCT-TM), Hospital de Clínicas de Porto Alegre (HCPA), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - T Barichello
- Center for Translational Psychiatry, Department of Psychiatry and Behavioral Sciences, The University of Texas Medical School at Houston, Houston, TX, USA; Laboratório de Microbiologia Experimental, Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - F Kapczinski
- Center for Translational Psychiatry, Department of Psychiatry and Behavioral Sciences, The University of Texas Medical School at Houston, Houston, TX, USA; Molecular Psychiatry Unit and National Science and Technology Institute for Translational Medicine (INCT-TM), Hospital de Clínicas de Porto Alegre (HCPA), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - J Quevedo
- Center for Translational Psychiatry, Department of Psychiatry and Behavioral Sciences, The University of Texas Medical School at Houston, Houston, TX, USA; Laboratório de Neurociências, Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| |
Collapse
|
219
|
Franklin TC, Wohleb ES, Duman RS. The Role of Immune Cells in the Brain during Physiological and Pathological Conditions. Psychiatr Ann 2015. [DOI: 10.3928/00485713-20150501-05] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
220
|
Zheng X, Zhang X, Wang G, Hao H. Treat the brain and treat the periphery: toward a holistic approach to major depressive disorder. Drug Discov Today 2015; 20:562-8. [PMID: 25849660 DOI: 10.1016/j.drudis.2015.03.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 03/07/2015] [Accepted: 03/27/2015] [Indexed: 12/17/2022]
Abstract
The limited medication for major depressive disorder (MDD) against an ever-rising disease burden presents an urgent need for therapeutic innovations. During recent years, studies looking at the systems regulation of mental health and disease have shown a remarkably powerful control of MDD by systemic signals. Meanwhile, the identification of a host of targets outside the brain opens the way to treat MDD by targeting systemic signals. We examine these emerging findings and consider the implications for current thinking regarding MDD pathogenesis and treatment. We highlight the opportunities and challenges of a periphery-targeting strategy and propose its incorporation into a holistic approach.
Collapse
Affiliation(s)
- Xiao Zheng
- Department of Pharmacy, Nanjing University of Chinese Medicine Affiliated Hospital, Nanjing 210029, China.
| | - Xueli Zhang
- Department of Pharmacy, Southeast University Affiliated Zhong Da Hospital, Nanjing 210009, China
| | - Guangji Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Haiping Hao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
221
|
Ziebell JM, Adelson PD, Lifshitz J. Microglia: dismantling and rebuilding circuits after acute neurological injury. Metab Brain Dis 2015; 30:393-400. [PMID: 24733573 PMCID: PMC4198517 DOI: 10.1007/s11011-014-9539-y] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 03/26/2014] [Indexed: 01/06/2023]
Abstract
The brain is comprised of neurons and its support system including astrocytes, glial cells and microglia, thereby forming neurovascular units. Neurons require support from glial cells to establish and maintain functional circuits, but microglia are often overlooked. Microglia function as the immune cell of the central nervous system, acting to monitor the microenvironment for changes in signaling, pathogens and injury. More recently, other functional roles for microglia within the healthy brain have been identified, including regulating synapse formation, elimination and function. This review aims to highlight and discuss these alternate microglial roles in the healthy and in contrast, diseased brain with a focus on two acute neurological diseases, traumatic brain injury and epilepsy. In these conditions, microglial roles in synaptic stripping and stabilization as part of neuronal:glial interactions may position them as mediators of the transition between injury-induced circuit dismantling and subsequent reorganization. Increased understanding of microglia roles could identify therapeutic targets to mitigate the consequences of neurological disease.
Collapse
Affiliation(s)
- Jenna M Ziebell
- Department of Child Health, University of Arizona College of Medicine, Phoenix, AZ, USA,
| | | | | |
Collapse
|
222
|
Cattaneo A, Macchi F, Plazzotta G, Veronica B, Bocchio-Chiavetto L, Riva MA, Pariante CM. Inflammation and neuronal plasticity: a link between childhood trauma and depression pathogenesis. Front Cell Neurosci 2015; 9:40. [PMID: 25873859 PMCID: PMC4379909 DOI: 10.3389/fncel.2015.00040] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 01/27/2015] [Indexed: 12/13/2022] Open
Abstract
During the past two decades, there has been increasing interest in understanding and characterizing the role of inflammation in major depressive disorder (MDD). Indeed, several are the evidences linking alterations in the inflammatory system to Major Depression, including the presence of elevated levels of pro-inflammatory cytokines, together with other mediators of inflammation. However, it is still not clear whether inflammation represents a cause or whether other factors related to depression result in these immunological effects. Regardless, exposure to early life stressful events, which represent a vulnerability factor for the development of psychiatric disorders, act through the modulation of inflammatory responses, but also of neuroplastic mechanisms over the entire life span. Indeed, early life stressful events can cause, possibly through epigenetic changes that persist over time, up to adulthood. Such alterations may concur to increase the vulnerability to develop psychopathologies. In this review we will discuss the role of inflammation and neuronal plasticity as relevant processes underlying depression development. Moreover, we will discuss the role of epigenetics in inducing alterations in inflammation-immune systems as well as dysfunction in neuronal plasticity, thus contributing to the long-lasting negative effects of stressful life events early in life and the consequent enhanced risk for depression. Finally we will provide an overview on the potential role of inflammatory system to aid diagnosis, predict treatment response, enhance treatment matching, and prevent the onset or relapse of Major Depression.
Collapse
Affiliation(s)
- Annamaria Cattaneo
- Stress, Psychiatry and Immunology Laboratory, Department of Psychological Medicine, Institute of Psychiatry, King's College London London, UK ; IRCCS Centro S Giovanni di Dio, Fatebenefratelli Brescia, Italy
| | - Flavia Macchi
- Department of Pharmacological and Biomolecular Sciences, University of Milan Milan, Italy
| | - Giona Plazzotta
- IRCCS Centro S Giovanni di Dio, Fatebenefratelli Brescia, Italy
| | - Begni Veronica
- Department of Pharmacological and Biomolecular Sciences, University of Milan Milan, Italy
| | - Luisella Bocchio-Chiavetto
- IRCCS Centro S Giovanni di Dio, Fatebenefratelli Brescia, Italy ; Faculty of Psychology, eCampus University Novedrate (Como), Italy
| | - Marco Andrea Riva
- Department of Pharmacological and Biomolecular Sciences, University of Milan Milan, Italy
| | - Carmine Maria Pariante
- Stress, Psychiatry and Immunology Laboratory, Department of Psychological Medicine, Institute of Psychiatry, King's College London London, UK
| |
Collapse
|
223
|
Ma L, Kulesskaya N, Võikar V, Tian L. Differential expression of brain immune genes and schizophrenia-related behavior in C57BL/6N and DBA/2J female mice. Psychiatry Res 2015; 226:211-6. [PMID: 25661533 DOI: 10.1016/j.psychres.2015.01.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 12/18/2014] [Accepted: 01/01/2015] [Indexed: 12/21/2022]
Abstract
Mounting evidence suggests the association of immune genes with complex neuropsychiatric diseases, such as schizophrenia. However, immune gene expression in the brain and their involvement in schizophrenia-related behavior in animal models have not been well studied so far. We analyzed the social (resident-intruder) and sensorimotor gating (pre-pulse inhibition (PPI) of acoustic startle) behaviors, and expression profiles of several brain immune genes in adult C57BL/6N and DBA/2J female mice. Compared to C57BL/6N mice, DBA/2J mice exhibited less social interaction in the resident-intruder test and reduced pre-pulse inhibition. The mRNA levels of Il1b and Il6 genes were significantly higher in the cortex and hypothalamus, while the mRNA level of C1qb was lower in the cortex, hippocampus and hypothalamus of DBA/2J mice compared to C57BL/6N mice. Furthermore, Tnfsf13b was up-regulated in the cortex and hippocampus, and so did Cd47 in the hippocampus, while Cx3cl1 was down-regulated in the cortex of DBA/2J mice. Our study demonstrates the differential expression of several immune genes in C57BL/6N and DBA/2J strains and more importantly provides clues on their potential importance in regulating schizophrenia-related endophenotypes in animal models.
Collapse
Affiliation(s)
- Li Ma
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| | | | - Vootele Võikar
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Li Tian
- Neuroscience Center, University of Helsinki, Helsinki, Finland; Psychiatry Research Center, Beijing Huilongguan Hospital, Beijing, China.
| |
Collapse
|
224
|
Steiner J, Schiltz K, Bernstein HG, Bogerts B. Antineuronal antibodies against neurotransmitter receptors and synaptic proteins in schizophrenia: current knowledge and clinical implications. CNS Drugs 2015; 29:197-206. [PMID: 25724386 DOI: 10.1007/s40263-015-0233-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
When Eugen Bleuler coined the term 'schizophrenia' he believed that various causes of illness may underlie this disease. Currently, neurodevelopmental abnormalities and consecutive impairments in dopaminergic and glutamatergic neurotransmission are considered as major causes of schizophrenia. However, there are various indications for involvement of immune processes, at least in subgroups of patients. Circulating antineuronal antibodies provide a promising link between the well-described disturbances in neurotransmission and the immune hypothesis of schizophrenia. This review summarizes important studies that have examined the role of glutamate, dopamine, acetylcholine and serotonin receptor autoantibodies, and other antineuronal antibodies against synaptic proteins in the serum of patients diagnosed with schizophrenia. Currently, it is not known whether the presence of antineuronal antibodies in blood should be considered as a causal or disease-modulating factor in schizophrenia. Due to emerging evidence regarding the important role of the blood-brain barrier, combined testing of serum and cerebrospinal fluid is likely to be more appropriate to answer this question than pure serum analyses. We suggest implementation of such testing in first-onset and treatment-resistant patients as part of the diagnostic process. In addition, future clinical trials should evaluate if immunotherapy (e.g. cortisone pulse therapy, intravenous immunoglobulins, plasmapheresis, rituximab, or cyclophosphamide) is helpful in cases with a neuroinflammatory component.
Collapse
Affiliation(s)
- Johann Steiner
- Department of Psychiatry, University of Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany,
| | | | | | | |
Collapse
|
225
|
Zheng CL, Wilmot B, Walter NA, Oberbeck D, Kawane S, Searles RP, McWeeney SK, Hitzemann R. Splicing landscape of the eight collaborative cross founder strains. BMC Genomics 2015; 16:52. [PMID: 25652416 PMCID: PMC4320832 DOI: 10.1186/s12864-015-1267-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 01/22/2015] [Indexed: 12/20/2022] Open
Abstract
Background The Collaborative Cross (CC) is a large panel of genetically diverse recombinant inbred mouse strains specifically designed to provide a systems genetics resource for the study of complex traits. In part, the utility of the CC stems from the extensive genome-wide annotations of founder strain sequence and structural variation. Still missing, however, are transcriptome-specific annotations of the CC founder strains that could further enhance the utility of this resource. Results We provide a comprehensive survey of the splicing landscape of the 8 CC founder strains by leveraging the high level of alternative splicing within the brain. Using deep transcriptome sequencing, we found that a majority of the splicing landscape is conserved among the 8 strains, with ~65% of junctions being shared by at least 2 strains. We, however, found a large number of potential strain-specific splicing events as well, with an average of ~3000 and ~500 with ≥3 and ≥10 sequence read coverage, respectively, within each strain. To better understand strain-specific splicing within the CC founder strains, we defined criteria for and identified high-confidence strain-specific splicing events. These splicing events were defined as exon-exon junctions 1) found within only one strain, 2) with a read coverage ≥10, and 3) defined by a canonical splice site. With these criteria, a total of 1509 high-confidence strain-specific splicing events were identified, with the majority found within two of the wild-derived strains, CAST and PWK. Strikingly, the overwhelming majority, 94%, of these strain-specific splicing events are not yet annotated. Strain-specific splicing was also located within genomic regions recently reported to be over- and under-represented within CC populations. Conclusions Phenotypic characterization of CC populations is increasing; thus these results will not only aid in further elucidating the transcriptomic architecture of the individual CC founder strains, but they will also help in guiding the utilization of the CC populations in the study of complex traits. This report is also the first to establish guidelines in defining and identifying strain-specific splicing across different mouse strains. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1267-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Christina L Zheng
- Department of Medical Informatics and Clinical Epidemiology, Division of Bioinformatics and Computational Biology, Oregon Health & Science University, Portland, Oregon, USA. .,Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA.
| | - Beth Wilmot
- Department of Medical Informatics and Clinical Epidemiology, Division of Bioinformatics and Computational Biology, Oregon Health & Science University, Portland, Oregon, USA. .,Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA. .,Oregon Clinical and Translational Research Institute, Oregon Health & Science University, Portland, Oregon, USA.
| | - Nicole Ar Walter
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, USA. .,Portland Alcohol Research Center, Oregon Health & Science University, Portland, Oregon, USA.
| | - Denesa Oberbeck
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, USA.
| | - Sunita Kawane
- Oregon Clinical and Translational Research Institute, Oregon Health & Science University, Portland, Oregon, USA.
| | - Robert P Searles
- Integrated Genomics Laboratory, Oregon Health & Science University, Portland, Oregon, USA.
| | - Shannon K McWeeney
- Department of Medical Informatics and Clinical Epidemiology, Division of Bioinformatics and Computational Biology, Oregon Health & Science University, Portland, Oregon, USA. .,Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA. .,Oregon Clinical and Translational Research Institute, Oregon Health & Science University, Portland, Oregon, USA. .,Department of Public Health and Preventative Medicine, Division of Biostatistics, Oregon Health & Science University, Portland, Oregon, USA.
| | - Robert Hitzemann
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, USA. .,Veterans Affairs Research Service, Portland, OR, USA.
| |
Collapse
|
226
|
Mesman E, Hillegers MH, Ambree O, Arolt V, Nolen WA, Drexhage HA. Monocyte activation, brain-derived neurotrophic factor (BDNF), and S100B in bipolar offspring: a follow-up study from adolescence into adulthood. Bipolar Disord 2015; 17:39-49. [PMID: 25039314 DOI: 10.1111/bdi.12231] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Accepted: 04/29/2014] [Indexed: 11/29/2022]
Abstract
OBJECTIVES There is increasing evidence that both immune and neurochemical alterations are involved in the pathogenesis of bipolar disorder; however, their precise role remains unclear. In this study, we aimed to evaluate neuro-immune changes in a prospective study on children of patients with bipolar disorder. METHODS Bipolar offspring, from the prospective Dutch bipolar offspring study (n = 140), were evaluated cross-sectionally within a longitudinal context at adolescence, young adulthood, and adulthood. We examined the expression of 44 inflammation-related genes in monocytes, the cytokines pentraxin 3 (PTX3), chemokine ligand 2 (CCL2), and interleukin-1β (IL-1β), and brain-derived neurotrophic factor (BDNF) and S100 calcium binding protein B (S100B) in the serum of bipolar offspring and healthy controls. RESULTS During adolescence, bipolar offspring showed increased inflammatory gene expression in monocytes, high serum PTX3 levels, but normal CCL2 levels. BDNF levels were decreased, while S100B levels were normal. During young adulthood, monocyte activation remained, although to a lesser degree. Serum PTX3 levels remained high, and signs of monocyte migration became apparent through increased CCL2 levels. BDNF and S100B levels were not measured. At adulthood, circulating monocytes had lost their activation state, but CCL2 levels remained increased. Both BDNF and S100B were now increased. Abnormalities were independent of psychopathology state at all stages. CONCLUSIONS This study suggests an aberrant neuro-immune state in bipolar offspring, which followed a dynamic course from adolescence into adulthood and was present irrespective of lifetime or future mood disorders. We therefore assumed that the aberrant neuro-immune state reflects a general state of vulnerability for mood disorders rather than being of direct predictive value.
Collapse
Affiliation(s)
- Esther Mesman
- Brain Center Rudolf Magnus, Department of Psychiatry, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | | | | | |
Collapse
|
227
|
Qin J, Yang X, Zhang RX, Luo YX, Li JL, Hou J, Zhang C, Li YJ, Shi J, Lu L, Wang JX, Zhu WL. Monocyte mediated brain targeting delivery of macromolecular drug for the therapy of depression. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2015; 11:391-400. [DOI: 10.1016/j.nano.2014.09.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2014] [Revised: 05/30/2014] [Accepted: 09/22/2014] [Indexed: 12/25/2022]
|
228
|
Grosse L, Carvalho LA, Wijkhuijs AJM, Bellingrath S, Ruland T, Ambrée O, Alferink J, Ehring T, Drexhage HA, Arolt V. Clinical characteristics of inflammation-associated depression: Monocyte gene expression is age-related in major depressive disorder. Brain Behav Immun 2015; 44:48-56. [PMID: 25150007 DOI: 10.1016/j.bbi.2014.08.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 08/10/2014] [Accepted: 08/12/2014] [Indexed: 11/25/2022] Open
Abstract
Increased inflammatory activation might only be present in a subgroup of depressed individuals in which immune processes are especially relevant to disease development. We aimed to analyze demographic, depression, and trauma characteristics of major depressive disorder (MDD) patients with regard to inflammatory monocyte gene expression. Fifty-six naturalistically treated MDD patients (32 ± 12 years) and 57 healthy controls (HC; 31 ± 11 years) were analyzed by the Inventory of Depressive Symptomatology (IDS) and by the Childhood Trauma Questionnaire (CTQ). We determined the expression of 38 inflammatory and immune activation genes including the glucocorticoid receptor (GR)α and GRβ genes in purified CD14(+) monocytes using quantitative-polymerase chain reaction (RT-qPCR). Monocyte gene expression was age-dependent, particularly in MDD patients. Increased monocyte gene expression and decreased GRα/β ratio were only present in MDD patients aged ⩾ 28 years. Post hoc analyses of monocyte immune activation in patients <28 years showed two subgroups: a subgroup with a severe course of depression (recurrent type, onset <15 years) - additionally characterized by panic/arousal symptoms and childhood trauma - that had a monocyte gene expression similar to HC, and a second subgroup with a milder course of the disorder (73% first episode depression, onset ⩾15 years) - additionally characterized by the absence of panic symptoms - that exhibited a strongly reduced inflammatory monocyte activation compared to HC. In conclusion, monocyte immune activation was not uniformly raised in MDD patients but was increased only in patients of 28 years and older.
Collapse
Affiliation(s)
- Laura Grosse
- Department of Psychiatry and Psychotherapy, University of Münster, Germany.
| | - Livia A Carvalho
- Department of Epidemiology and Public Health, University College London, United Kingdom
| | | | | | - Tillmann Ruland
- Department of Psychiatry and Psychotherapy, University of Münster, Germany
| | - Oliver Ambrée
- Department of Psychiatry and Psychotherapy, University of Münster, Germany
| | - Judith Alferink
- Department of Psychiatry and Psychotherapy, University of Münster, Germany; Cells-in-Motion Cluster of Excellence (EXC 1003-CiM), University of Münster, Germany
| | - Thomas Ehring
- Department of Psychology, University of Münster, Germany
| | - Hemmo A Drexhage
- Department of Immunology, Erasmus MC, Rotterdam, The Netherlands
| | - Volker Arolt
- Department of Psychiatry and Psychotherapy, University of Münster, Germany
| |
Collapse
|
229
|
Wohleb ES, McKim DB, Sheridan JF, Godbout JP. Monocyte trafficking to the brain with stress and inflammation: a novel axis of immune-to-brain communication that influences mood and behavior. Front Neurosci 2015; 8:447. [PMID: 25653581 PMCID: PMC4300916 DOI: 10.3389/fnins.2014.00447] [Citation(s) in RCA: 240] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 12/19/2014] [Indexed: 12/13/2022] Open
Abstract
HIGHLIGHTSPsychological stress activates neuroendocrine pathways that alter immune responses.Stress-induced alterations in microglia phenotype and monocyte priming leads to aberrant peripheral and central inflammation.Elevated pro-inflammatory cytokine levels caused by microglia activation and recruitment of monocytes to the brain contribute to development and persistent anxiety-like behavior.Mechanisms that mediate interactions between microglia, endothelial cells, and macrophages and how these contribute to changes in behavior are discussed.Sensitization of microglia and re-distribution of primed monocytes are implicated in re-establishment of anxiety-like behavior. Psychological stress causes physiological, immunological, and behavioral alterations in humans and rodents that can be maladaptive and negatively affect quality of life. Several lines of evidence indicate that psychological stress disrupts key functional interactions between the immune system and brain that ultimately affects mood and behavior. For example, activation of microglia, the resident innate immune cells of the brain, has been implicated as a key regulator of mood and behavior in the context of prolonged exposure to psychological stress. Emerging evidence implicates a novel neuroimmune circuit involving microglia activation and sympathetic outflow to the peripheral immune system that further reinforces stress-related behaviors by facilitating the recruitment of inflammatory monocytes to the brain. Evidence from various rodent models, including repeated social defeat (RSD), revealed that trafficking of monocytes to the brain promoted the establishment of anxiety-like behaviors following prolonged stress exposure. In addition, new evidence implicates monocyte trafficking from the spleen to the brain as key regulator of recurring anxiety following exposure to prolonged stress. The purpose of this review is to discuss mechanisms that cause stress-induced monocyte re-distribution in the brain and how dynamic interactions between microglia, endothelial cells, and brain macrophages lead to maladaptive behavioral responses.
Collapse
Affiliation(s)
- Eric S Wohleb
- Department of Psychiatry, Yale University School of Medicine New Haven, CT, USA
| | - Daniel B McKim
- Division of Biosciences, The Ohio State University College of Dentistry Columbus, OH, USA ; Department of Neuroscience, The Ohio State University College of Medicine Columbus, OH, USA
| | - John F Sheridan
- Division of Biosciences, The Ohio State University College of Dentistry Columbus, OH, USA ; Institute for Behavioral Medicine Research, The Ohio State University College of Medicine Columbus, OH, USA ; Center for Brain and Spinal Cord Repair, The Ohio State University College of Medicine Columbus, OH, USA
| | - Jonathan P Godbout
- Department of Neuroscience, The Ohio State University College of Medicine Columbus, OH, USA ; Institute for Behavioral Medicine Research, The Ohio State University College of Medicine Columbus, OH, USA ; Center for Brain and Spinal Cord Repair, The Ohio State University College of Medicine Columbus, OH, USA
| |
Collapse
|
230
|
Glial cells as key players in schizophrenia pathology: recent insights and concepts of therapy. Schizophr Res 2015; 161:4-18. [PMID: 24948484 DOI: 10.1016/j.schres.2014.03.035] [Citation(s) in RCA: 147] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 02/27/2014] [Accepted: 03/01/2014] [Indexed: 02/07/2023]
Abstract
The past decade has witnessed an explosion of knowledge on the impact of glia for the neurobiological foundation of schizophrenia. A plethora of studies have shown structural and functional abnormalities in all three types of glial cells. There is convincing evidence of reduced numbers of oligodendrocytes, impaired cell maturation and altered gene expression of myelin/oligodendrocyte-related genes that may in part explain white matter abnormalities and disturbed inter- and intra-hemispheric connectivity, which are characteristic signs of schizophrenia. Earlier reports of astrogliosis could not be confirmed by later studies, although the expression of a variety of astrocyte-related genes is abnormal in psychosis. Since astrocytes play a key role in the synaptic metabolism of glutamate, GABA, monoamines and purines, astrocyte dysfunction may contribute to certain aspects of disturbed neurotransmission in schizophrenia. Finally, increased densities of microglial cells and aberrant expression of microglia-related surface markers in schizophrenia suggest that immunological/inflammatory factors are of considerable relevance for the pathophysiology of psychosis. This review describes current evidence for the multifaceted role of glial cells in schizophrenia and discusses efforts to develop glia-directed therapies for the treatment of the disease.
Collapse
|
231
|
Vargas-Caraveo A, Pérez-Ishiwara DG, Martínez-Martínez A. Chronic Psychological Distress as an Inducer of Microglial Activation and Leukocyte Recruitment into the Area Postrema. Neuroimmunomodulation 2015; 22:311-21. [PMID: 25765708 DOI: 10.1159/000369350] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 10/27/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Chronic psychological distress can cause neuroinflammation, but the involvement of leukocytes in this inflammatory response remains unclear. The area postrema (AP) is considered a neural-immune interface because it lacks a blood-brain barrier and a site for leukocyte recruitment in neuroinflammatory conditions induced by immunological insults, but its role in chronic psychological distress has not been explored. OBJECTIVE To determine leukocyte recruitment to the AP after chronic psychological distress. METHODS Rats were exposed to cat odor for 5 consecutive days to induce distress, and, on the 6th day, their brains were dissected to perform immunohistofluorescence studies of the AP. Immune cells were identified and quantified with CD45 and CD11b markers. The distribution of neurons and immune cells was determined using TrkA and CD45 markers, respectively. RESULTS Distress induced a significant increase in CD45(+) and CD11b(+) cells in the AP. Three immunophenotypes were determined in the control and distress groups: CD45(+)/CD11b(-), CD45(+)/CD11b(+) and CD45(-)/CD11b(+). CD expression, morphology and fluorescence intensity enabled the identification of different immune cell types: starting from longitudinal ramified microglia (mainly in the control group) to amoeboid microglia, monocytes and lymphocytes (mostly in the distressed group). TrkA and CD45 expression in the AP revealed the proximity between soma neurons and leukocytes. Interestingly, some CD45(+) cells expressed TrkA, with increased expression in the distressed group. CONCLUSIONS The identification of microglial activation, leukocyte recruitment and the close proximity between neurons and leukocytes in the AP after chronic psychological distress exposure suggests the AP as a site for distress-induced immune responses and engraftment of leukocytes infiltrating the CNS.
Collapse
Affiliation(s)
- Alejandra Vargas-Caraveo
- Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, and Centro de Investigación en Biotecnologia Aplicada del IPN, México, México
| | | | | |
Collapse
|
232
|
Wędrychowicz A, Zając A, Pilecki M, Kościelniak B, Tomasik PJ. Peptides from adipose tissue in mental disorders. World J Psychiatry 2014; 4:103-111. [PMID: 25540725 PMCID: PMC4274582 DOI: 10.5498/wjp.v4.i4.103] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Revised: 11/14/2014] [Accepted: 12/10/2014] [Indexed: 02/05/2023] Open
Abstract
Adipose tissue is a dynamic endocrine organ that is essential to regulation of metabolism in humans. A new approach to mental disorders led to research on involvement of adipokines in the etiology of mental disorders and mood states and their impact on the health status of psychiatric patients, as well as the effects of treatment for mental health disorders on plasma levels of adipokines. There is evidence that disturbances in adipokine secretion are important in the pathogenesis, clinical presentation and outcome of mental disorders. Admittedly leptin and adiponectin are involved in pathophysiology of depression. A lot of disturbances in secretion and plasma levels of adipokines are observed in eating disorders with a significant impact on the symptoms and course of a disease. It is still a question whether observed dysregulation of adipokines secretion are primary or secondary. Moreover findings in this area are somewhat inconsistent, owing to differences in patient age, sex, socioeconomic status, smoking habits, level of physical activity, eating pathology, general health or medication. This was the rationale for our detailed investigation into the role of the endocrine functions of adipose tissue in mental disorders. It seems that we are continually at the beginning of understanding of the relation between adipose tissue and mental disorders.
Collapse
|
233
|
Nakagawa Y, Chiba K. Role of microglial m1/m2 polarization in relapse and remission of psychiatric disorders and diseases. Pharmaceuticals (Basel) 2014; 7:1028-48. [PMID: 25429645 PMCID: PMC4276905 DOI: 10.3390/ph7121028] [Citation(s) in RCA: 152] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 11/14/2014] [Accepted: 11/14/2014] [Indexed: 12/27/2022] Open
Abstract
Psychiatric disorders such as schizophrenia and major depressive disorder were thought to be caused by neurotransmitter abnormalities. Patients with these disorders often experience relapse and remission; however the underlying molecular mechanisms of relapse and remission still remain unclear. Recent advanced immunological analyses have revealed that M1/M2 polarization of macrophages plays an important role in controlling the balance between promotion and suppression in inflammation. Microglial cells share certain characteristics with macrophages and contribute to immune-surveillance in the central nervous system (CNS). In this review, we summarize immunoregulatory functions of microglia and discuss a possible role of microglial M1/M2 polarization in relapse and remission of psychiatric disorders and diseases. M1 polarized microglia can produce pro-inflammatory cytokines, reactive oxygen species, and nitric oxide, suggesting that these molecules contribute to dysfunction of neural network in the CNS. Alternatively, M2 polarized microglia express cytokines and receptors that are implicated in inhibiting inflammation and restoring homeostasis. Based on these aspects, we propose a possibility that M1 and M2 microglia are related to relapse and remission, respectively in psychiatric disorders and diseases. Consequently, a target molecule skewing M2 polarization of microglia may provide beneficial therapies for these disorders and diseases in the CNS.
Collapse
Affiliation(s)
- Yutaka Nakagawa
- Research Strategy and Planning Department, Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama 227-0033, Japan.
| | - Kenji Chiba
- Advanced Medical Research Laboratories, Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama 227-0033, Japan.
| |
Collapse
|
234
|
Sellgren C, Frisell T, Lichtenstein P, Landèn M, Askling J. The association between schizophrenia and rheumatoid arthritis: a nationwide population-based Swedish study on intraindividual and familial risks. Schizophr Bull 2014; 40:1552-9. [PMID: 24714379 PMCID: PMC4193721 DOI: 10.1093/schbul/sbu054] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Numerous studies have reported a reduced risk of rheumatoid arthritis (RA) in schizophrenia. The mechanisms are unknown, but recent genome-wide association studies of schizophrenia have shown strong associations with markers spanning the major histocompatibility complex region, indicating a possible role for adaptive immunity also in schizophrenia. In this population-based cohort study, we assess the associations between RA and schizophrenia and the extent to which any observed associations are specific to RA/schizophrenia. We then extend the assessments per RA subtype and to risks in first-degree relatives. The study population included every individual identified in the Swedish Population Register born in Sweden between 1932 and 1989. The risk for RA in schizophrenia was significantly decreased (hazard ratio [HR] = 0.69, 95% CI = 0.59-0.80), but similar reductions were noted for osteoarthritis (a noninflammatory joint disorder) and ankylosing spondylitis (a non-RA inflammatory disorder). Comparable associations were seen in schizoaffective subjects while no significant associations were observed in bipolar disorder. Overall, first-degree relatives of schizophrenia patients were not at reduced risk of RA, but the risk for seronegative RA was significantly decreased in children and siblings of schizophrenia probands (HR = 0.13, 95% CI = 0.02-0.95 and HR = 0.67, 95% CI = 049-0.92, respectively). In conclusion, our intraindividual analyses suggest that differential misclassification bias is an important factor for the observed inverse association and emphasize the need of optimized care-provision for nonpsychiatric symptoms in schizophrenia patients. Our familial analyses indicted the possibility of an inverse coinheritance of schizophrenia and seronegative RA.
Collapse
Affiliation(s)
- Carl Sellgren
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden;
| | - Thomas Frisell
- Clinical Epidemiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Paul Lichtenstein
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Mikael Landèn
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden;,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Johan Askling
- Clinical Epidemiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden;,Department of Rheumatology, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
235
|
Missault S, Van den Eynde K, Vanden Berghe W, Fransen E, Weeren A, Timmermans JP, Kumar-Singh S, Dedeurwaerdere S. The risk for behavioural deficits is determined by the maternal immune response to prenatal immune challenge in a neurodevelopmental model. Brain Behav Immun 2014; 42:138-46. [PMID: 24973728 DOI: 10.1016/j.bbi.2014.06.013] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 06/06/2014] [Accepted: 06/16/2014] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Schizophrenia is a highly disabling psychiatric disorder with a proposed neurodevelopmental basis. One mechanism through which genetic and environmental risk factors might act is by triggering persistent brain inflammation, as evidenced by long-lasting neuro-immunological disturbances in patients. Our goal was to investigate whether microglia activation is a neurobiological correlate to the altered behaviour in the maternal immune activation (MIA) model, a well-validated animal model with relevance to schizophrenia. A recent observation in the MIA model is the differential maternal body weight response to the immune stimulus, correlated with a different behavioural outcome in the offspring. Although it is generally assumed that the differences in maternal weight response reflect differences in cytokine response, this has not been investigated so far. Our aim was to investigate whether (i) the maternal weight response to MIA reflects differences in the maternal cytokine response, (ii) the differential behavioural phenotype of the offspring extends to depressive symptoms such as anhedonia and (iii) there are changes in chronic microglia activation dependent on the behavioural phenotype. METHODS Based on a dose-response study, MIA was induced in pregnant rats by injecting 4mg/kg Poly I:C at gestational day 15. Serum samples were collected to assess the amount of TNF-α in the maternal blood following MIA. MIA offspring were divided into weight loss (WL; n=14) and weight gain (WG; n=10) groups, depending on the maternal body weight response to Poly I:C. Adult offspring were behaviourally phenotyped for prepulse inhibition, locomotor activity with and without amphetamine and MK-801 challenge, and sucrose preference. Finally, microglia activation was scored on CD11b- and Iba1-immunohistochemically stained sections. RESULTS Pregnant dams that lost weight following MIA showed increased levels of TNF-α compared to controls, unlike dams that gained weight following MIA. Poly I:C WL offspring showed the most severe behavioural outcome. Poly I:C WG offspring, on the other hand, did not show clear behavioural deficits. Most interestingly a reduced sucrose preference indicative of anhedonia was found in Poly I:C WL but not Poly I:C WG offspring compared to controls. Finally, there were no significant differences in microglia activation scores between any of the investigated groups. CONCLUSIONS The individual maternal immune response to MIA is an important determinant of the behavioural outcome in offspring, including negative symptoms such as anhedonia. We failed to find any significant difference in the level of microglia activation between Poly I:C WL, Poly I:C WG and control offspring.
Collapse
Affiliation(s)
- S Missault
- Experimental Laboratory of Translational Neuroscience and Otolaryngology, Faculty of Medicine and Health Sciences, University of Antwerp, Campus Drie Eiken, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - K Van den Eynde
- Experimental Laboratory of Translational Neuroscience and Otolaryngology, Faculty of Medicine and Health Sciences, University of Antwerp, Campus Drie Eiken, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - W Vanden Berghe
- Laboratory of Protein Science, Proteomics & Epigenetic Signaling, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Campus Drie Eiken, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - E Fransen
- StatUA, University of Antwerp, Campus Drie Eiken, Prins Boudewijnlaan 43, 2650 Edegem, Belgium
| | - A Weeren
- StatUA, University of Antwerp, City Campus, Prinsstraat 13, 2000 Antwerpen, Belgium
| | - J P Timmermans
- Laboratory of Cell Biology & Histology, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
| | - S Kumar-Singh
- Laboratory of Cell Biology & Histology, Faculty of Medicine and Health Sciences, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
| | - S Dedeurwaerdere
- Experimental Laboratory of Translational Neuroscience and Otolaryngology, Faculty of Medicine and Health Sciences, University of Antwerp, Campus Drie Eiken, Universiteitsplein 1, 2610 Wilrijk, Belgium.
| |
Collapse
|
236
|
Roman A, Kreiner G, Nalepa I. Macrophages and depression - a misalliance or well-arranged marriage? Pharmacol Rep 2014; 65:1663-72. [PMID: 24553015 DOI: 10.1016/s1734-1140(13)71528-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Revised: 08/30/2013] [Indexed: 12/29/2022]
Abstract
Depression is a severe medical condition with multiple manifestations and diverse, largely unknown etiologies. The immune system, particularly macrophages, plays an important role in the pathology of the illness. Macrophages represent a heterogeneous population of immune cells that is dispersed throughout the body. The central nervous system is populated by several types of macrophages, including microglia, perivascular cells, meningeal and choroid plexus macrophages and pericytes. These cells occupy different brain compartments and have various functions. Under basal conditions, brain macrophages support the proper function of neural cells, organize and preserve the neuronal network and maintain homeostasis. As cells of the innate immune system, they recognize and react to any disturbances in homeostasis, eliminating pathogens or damaged cells, terminating inflammation and proceeding to initiate tissue reconstruction. Disturbances in these processes result in diverse pathologies. In particular, tissue stress or malfunction, both in the brain and in the periphery, produce sustained inflammatory states, which may cause depression. Excessive release of proinflammatory mediators is responsible for alterations of neurotransmitter systems and the occurrence of depressive symptoms. Almost all antidepressive drugs target monoamine or serotonin neurotransmission and also have anti-inflammatory or immunosuppressive properties. In addition, non-pharmacological treatments, such as electroconvulsive shock, can also exert anti-inflammatory effects. Recent studies have shown that antidepressive therapies can affect the functional properties of peripheral and brain macrophages and skew them toward the anti-inflammatory M2 phenotype. Because macrophages can affect outcome of inflammatory diseases, alleviate sickness behavior and improve cognitive function, it is possible that the effects of antidepressive treatments may be, at least in part, mediated by changes in macrophage activity.
Collapse
Affiliation(s)
- Adam Roman
- Department of Brain Biochemistry, Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, PL 31-343 Kraków, Poland.
| | | | | |
Collapse
|
237
|
Pyrroloquinoline quinone (PQQ) inhibits lipopolysaccharide induced inflammation in part via downregulated NF-κB and p38/JNK activation in microglial and attenuates microglia activation in lipopolysaccharide treatment mice. PLoS One 2014; 9:e109502. [PMID: 25314304 PMCID: PMC4196908 DOI: 10.1371/journal.pone.0109502] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 09/11/2014] [Indexed: 01/25/2023] Open
Abstract
Therapeutic strategies designed to inhibit the activation of microglia may lead to significant advancement in the treatment of most neurodegenerative diseases. Pyrroloquinoline quinone (PQQ) is a naturally occurring redox cofactor that acts as an essential nutrient, antioxidant, and has been reported to exert potent immunosuppressive effects. In the present study, the anti-inflammatory effects of PQQ was investigated in LPS treated primary microglia cells. Our observations showed that pretreatment with PQQ significantly inhibited the production of NO and PGE2 and suppressed the expression of pro-inflammatory mediators such as iNOS, COX-2, TNF-a, IL-1b, IL-6, MCP-1 and MIP-1a in LPS treated primary microglia cells. The nuclear translocation of NF-κB and the phosphorylation level of p65, p38 and JNK MAP kinase pathways were also inhibited by PQQ in LPS stimulated primary microglia cells. Further a systemic LPS treatment acute inflammation murine brain model was used to study the suppressive effects of PQQ against neuroinflammation in vivo. Mice treated with PQQ demonstrated marked attenuation of neuroinflammation based on Western blotting and immunohistochemistry analysis of Iba1-against antibody in the brain tissue. Indicated that PQQ protected primary cortical neurons against microglia-mediated neurotoxicity. These results collectively suggested that PQQ might be a promising therapeutic agent for alleviating the progress of neurodegenerative diseases associated with microglia activation.
Collapse
|
238
|
Kern S, Skoog I, Börjesson-Hanson A, Blennow K, Zetterberg H, Ostling S, Kern J, Gudmundsson P, Marlow T, Rosengren L, Waern M. Higher CSF interleukin-6 and CSF interleukin-8 in current depression in older women. Results from a population-based sample. Brain Behav Immun 2014; 41:55-8. [PMID: 24858658 DOI: 10.1016/j.bbi.2014.05.006] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 05/12/2014] [Accepted: 05/13/2014] [Indexed: 10/25/2022] Open
Abstract
OBJECTIVE The literature regarding cerebrospinal fluid (CSF) cytokines in geriatric depression is sparse. The aim of this study was to examine associations between CSF interleukin-6 (IL-6), interleukin-8 (IL-8) and depression in a population-based sample of older women who were followed for 17 years. METHODS 86 dementia-free women aged 70-84 years who participated in the Prospective Population Study of Women in Gothenburg, Sweden took part in a lumbar puncture in 1992-3. CSF IL-6 and CSF IL-8 were measured. Psychiatric symptoms were rated with the Comprehensive Psychopathological Rating Scale at baseline and at three subsequent face-to-face examinations. Depression (major or minor) was diagnosed in accordance with DSM-IV/DSM-IV research criteria. RESULTS At baseline, women with ongoing major (n=10) or minor depression (n=9) had higher levels of CSF IL-6 (p=0.008) and CSF IL-8 (p=0.007) compared with those without depression (n=67). Higher CSF IL-8 was related to higher MADRS score (p=0.003). New cases of depression were observed in 9 women during follow-ups. No associations between CSF cytokine levels and future depression could be shown in women without depression at baseline. CONCLUSION Higher levels of CSF IL-6 and IL-8 were associated with current depression in this population-based sample. CSF IL-6 and CSF IL-8 may play a role in depression in late life.
Collapse
Affiliation(s)
- Silke Kern
- Neuropsychiatric Epidemiology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Wallinsgatan 6, 43141 Mölndal, Sweden; Clinical Neurochemistry Laboratory, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Sahlgrenska University Hospital, 43180 Mölndal, Sweden.
| | - Ingmar Skoog
- Neuropsychiatric Epidemiology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Wallinsgatan 6, 43141 Mölndal, Sweden.
| | - Anne Börjesson-Hanson
- Neuropsychiatric Epidemiology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Wallinsgatan 6, 43141 Mölndal, Sweden.
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Sahlgrenska University Hospital, 43180 Mölndal, Sweden.
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Sahlgrenska University Hospital, 43180 Mölndal, Sweden; UCL Institute of Neurology, Queen Square, London WC1N 3BG, United Kingdom.
| | - Svante Ostling
- Neuropsychiatric Epidemiology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Wallinsgatan 6, 43141 Mölndal, Sweden.
| | - Jürgen Kern
- Neuropsychiatric Epidemiology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Wallinsgatan 6, 43141 Mölndal, Sweden.
| | - Pia Gudmundsson
- Neuropsychiatric Epidemiology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Wallinsgatan 6, 43141 Mölndal, Sweden.
| | - Thomas Marlow
- Neuropsychiatric Epidemiology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Wallinsgatan 6, 43141 Mölndal, Sweden.
| | - Lars Rosengren
- Department of Neurology, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden.
| | - Margda Waern
- Neuropsychiatric Epidemiology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Wallinsgatan 6, 43141 Mölndal, Sweden.
| |
Collapse
|
239
|
Kang SS, Kurti A, Fair DA, Fryer JD. Dietary intervention rescues maternal obesity induced behavior deficits and neuroinflammation in offspring. J Neuroinflammation 2014; 11:156. [PMID: 25212412 PMCID: PMC4172780 DOI: 10.1186/s12974-014-0156-9] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 08/21/2014] [Indexed: 12/27/2022] Open
Abstract
Obesity induces a low-grade inflammatory state and has been associated with behavioral and cognitive alterations. Importantly, maternal environmental insults can adversely impact subsequent offspring behavior and have been linked with neurodevelopmental disorders such as autism spectrum disorder (ASD) and attention deficit hyperactivity disorder (AHDH). It is unknown if maternal obesity significantly alters offspring sociability, a key ASD feature, and if altering maternal diet will provide an efficacious intervention paradigm for behavioral deficits. Here we investigated the impact of maternal high fat diet (HFD) and maternal dietary intervention during lactation on offspring behavior and brain inflammation in mice. We found that maternal HFD increased anxiety and decreased sociability in female offspring. Additionally, female offspring from HFD-fed dams also exhibited increased brain IL-1β and TNFα and microglial activation. Importantly, maternal dietary intervention during lactation was sufficient to alleviate social deficits and brain inflammation. Maternal obesity during gestation alone was sufficient to increase hyperactivity in male offspring, a phenotype that was not ameliorated by dietary intervention. These data suggest that maternal HFD acts as a prenatal/perinatal insult that significantly impacts offspring behavior and inflammation and that dietary intervention during lactation may be an easily translatable, efficacious intervention to offset some of these manifestations.
Collapse
|
240
|
Hung YN, Yang SY, Huang MC, Lung FW, Lin SK, Chen KY, Kuo CJ, Chen YY. Cancer incidence in people with affective disorder: nationwide cohort study in Taiwan, 1997-2010. Br J Psychiatry 2014; 205:183-8. [PMID: 24970771 DOI: 10.1192/bjp.bp.114.144741] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND Cancer is a serious public health problem worldwide, and its relationship with affective disorders is not clear. Aims To investigate alcohol- and tobacco-related cancer risk among patients with affective disorders in a large Taiwanese cohort. METHOD Records of newly admitted patients with affective disorders from January 1997 through December 2002 were retrieved from the Psychiatric Inpatient Medical Claims database in Taiwan. Cancers were stratified by site and grouped into tobacco- or alcohol-related cancers. Standardised incidence ratios (SIRs) were calculated to compare the risk of cancer between those with affective disorders and the general population. RESULTS Some 10 207 patients with bipolar disorder and 9826 with major depression were included. The risk of cancer was higher in patients with major depression (SIR = 2.01, 95% CI 1.85-2.19) than in those with bipolar disorder (SIR 1.39, 95% CI 1.26-1.53). The elevated cancer risk among individuals ever admitted to hospital for affective disorders was more pronounced in tobacco- and/or alcohol-related cancers. CONCLUSIONS Elevated cancer risk was found in patients who had received in-patient care for affective disorders. They require holistic approaches to lifestyle behaviours and associated cancer risks.
Collapse
Affiliation(s)
- Yen-Ni Hung
- Yen-Ni Hung, PhD, School of Gerontology Health Management and Master Program in Long-Term Care, College of Nursing, Taipei Medical University, Department of Nursing, School of Nursing, National Yang-Ming University and Department of Education and Research, Taipei City Hospital, Taipei; Shu-Yu Yang, PhD, Taipei City Psychiatric Center, Taipei City Hospital, Taipei and Graduate Institute of Clinical Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan; Ming-Chyi Huang, MD, PhD, Taipei City Psychiatric Center, Taipei City Hospital and Department of Psychiatry, School of Medicine, Taipei Medical University, Taipei; For-Wey Lung, MD, PhD, Taipei City Psychiatric Center, Taipei City Hospital, Taipei; Shih-Ku Lin, MD,Taipei City Psychiatric Center, Taipei City Hospital and Department of Psychiatry, School of Medicine, Taipei Medical University, Taipei; Kuan-Yu Chen, MD, PhD, Taipei City Psychiatric Center, Taipei City Hospital, Taipei; Chian-Jue Kuo, MD, PhD, Taipei City Psychiatric Center, Taipei City Hospital and Department of Psychiatry, School of Medicine, Taipei Medical University and Psychiatric Research Center, Taipei Medical University Hospital, Taipei, Taiwan; Ying-Yeh Chen, MD, ScD, Taipei City Psychiatric Center, Taipei City Hospital and Institute of Public Health and Department of Public Health, National Yang-Ming University, Taipei, Taiwan
| | - Shu-Yu Yang
- Yen-Ni Hung, PhD, School of Gerontology Health Management and Master Program in Long-Term Care, College of Nursing, Taipei Medical University, Department of Nursing, School of Nursing, National Yang-Ming University and Department of Education and Research, Taipei City Hospital, Taipei; Shu-Yu Yang, PhD, Taipei City Psychiatric Center, Taipei City Hospital, Taipei and Graduate Institute of Clinical Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan; Ming-Chyi Huang, MD, PhD, Taipei City Psychiatric Center, Taipei City Hospital and Department of Psychiatry, School of Medicine, Taipei Medical University, Taipei; For-Wey Lung, MD, PhD, Taipei City Psychiatric Center, Taipei City Hospital, Taipei; Shih-Ku Lin, MD,Taipei City Psychiatric Center, Taipei City Hospital and Department of Psychiatry, School of Medicine, Taipei Medical University, Taipei; Kuan-Yu Chen, MD, PhD, Taipei City Psychiatric Center, Taipei City Hospital, Taipei; Chian-Jue Kuo, MD, PhD, Taipei City Psychiatric Center, Taipei City Hospital and Department of Psychiatry, School of Medicine, Taipei Medical University and Psychiatric Research Center, Taipei Medical University Hospital, Taipei, Taiwan; Ying-Yeh Chen, MD, ScD, Taipei City Psychiatric Center, Taipei City Hospital and Institute of Public Health and Department of Public Health, National Yang-Ming University, Taipei, Taiwan
| | - Ming-Chyi Huang
- Yen-Ni Hung, PhD, School of Gerontology Health Management and Master Program in Long-Term Care, College of Nursing, Taipei Medical University, Department of Nursing, School of Nursing, National Yang-Ming University and Department of Education and Research, Taipei City Hospital, Taipei; Shu-Yu Yang, PhD, Taipei City Psychiatric Center, Taipei City Hospital, Taipei and Graduate Institute of Clinical Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan; Ming-Chyi Huang, MD, PhD, Taipei City Psychiatric Center, Taipei City Hospital and Department of Psychiatry, School of Medicine, Taipei Medical University, Taipei; For-Wey Lung, MD, PhD, Taipei City Psychiatric Center, Taipei City Hospital, Taipei; Shih-Ku Lin, MD,Taipei City Psychiatric Center, Taipei City Hospital and Department of Psychiatry, School of Medicine, Taipei Medical University, Taipei; Kuan-Yu Chen, MD, PhD, Taipei City Psychiatric Center, Taipei City Hospital, Taipei; Chian-Jue Kuo, MD, PhD, Taipei City Psychiatric Center, Taipei City Hospital and Department of Psychiatry, School of Medicine, Taipei Medical University and Psychiatric Research Center, Taipei Medical University Hospital, Taipei, Taiwan; Ying-Yeh Chen, MD, ScD, Taipei City Psychiatric Center, Taipei City Hospital and Institute of Public Health and Department of Public Health, National Yang-Ming University, Taipei, Taiwan
| | - For-Wey Lung
- Yen-Ni Hung, PhD, School of Gerontology Health Management and Master Program in Long-Term Care, College of Nursing, Taipei Medical University, Department of Nursing, School of Nursing, National Yang-Ming University and Department of Education and Research, Taipei City Hospital, Taipei; Shu-Yu Yang, PhD, Taipei City Psychiatric Center, Taipei City Hospital, Taipei and Graduate Institute of Clinical Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan; Ming-Chyi Huang, MD, PhD, Taipei City Psychiatric Center, Taipei City Hospital and Department of Psychiatry, School of Medicine, Taipei Medical University, Taipei; For-Wey Lung, MD, PhD, Taipei City Psychiatric Center, Taipei City Hospital, Taipei; Shih-Ku Lin, MD,Taipei City Psychiatric Center, Taipei City Hospital and Department of Psychiatry, School of Medicine, Taipei Medical University, Taipei; Kuan-Yu Chen, MD, PhD, Taipei City Psychiatric Center, Taipei City Hospital, Taipei; Chian-Jue Kuo, MD, PhD, Taipei City Psychiatric Center, Taipei City Hospital and Department of Psychiatry, School of Medicine, Taipei Medical University and Psychiatric Research Center, Taipei Medical University Hospital, Taipei, Taiwan; Ying-Yeh Chen, MD, ScD, Taipei City Psychiatric Center, Taipei City Hospital and Institute of Public Health and Department of Public Health, National Yang-Ming University, Taipei, Taiwan
| | - Shih-Ku Lin
- Yen-Ni Hung, PhD, School of Gerontology Health Management and Master Program in Long-Term Care, College of Nursing, Taipei Medical University, Department of Nursing, School of Nursing, National Yang-Ming University and Department of Education and Research, Taipei City Hospital, Taipei; Shu-Yu Yang, PhD, Taipei City Psychiatric Center, Taipei City Hospital, Taipei and Graduate Institute of Clinical Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan; Ming-Chyi Huang, MD, PhD, Taipei City Psychiatric Center, Taipei City Hospital and Department of Psychiatry, School of Medicine, Taipei Medical University, Taipei; For-Wey Lung, MD, PhD, Taipei City Psychiatric Center, Taipei City Hospital, Taipei; Shih-Ku Lin, MD,Taipei City Psychiatric Center, Taipei City Hospital and Department of Psychiatry, School of Medicine, Taipei Medical University, Taipei; Kuan-Yu Chen, MD, PhD, Taipei City Psychiatric Center, Taipei City Hospital, Taipei; Chian-Jue Kuo, MD, PhD, Taipei City Psychiatric Center, Taipei City Hospital and Department of Psychiatry, School of Medicine, Taipei Medical University and Psychiatric Research Center, Taipei Medical University Hospital, Taipei, Taiwan; Ying-Yeh Chen, MD, ScD, Taipei City Psychiatric Center, Taipei City Hospital and Institute of Public Health and Department of Public Health, National Yang-Ming University, Taipei, Taiwan
| | - Kuan-Yu Chen
- Yen-Ni Hung, PhD, School of Gerontology Health Management and Master Program in Long-Term Care, College of Nursing, Taipei Medical University, Department of Nursing, School of Nursing, National Yang-Ming University and Department of Education and Research, Taipei City Hospital, Taipei; Shu-Yu Yang, PhD, Taipei City Psychiatric Center, Taipei City Hospital, Taipei and Graduate Institute of Clinical Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan; Ming-Chyi Huang, MD, PhD, Taipei City Psychiatric Center, Taipei City Hospital and Department of Psychiatry, School of Medicine, Taipei Medical University, Taipei; For-Wey Lung, MD, PhD, Taipei City Psychiatric Center, Taipei City Hospital, Taipei; Shih-Ku Lin, MD,Taipei City Psychiatric Center, Taipei City Hospital and Department of Psychiatry, School of Medicine, Taipei Medical University, Taipei; Kuan-Yu Chen, MD, PhD, Taipei City Psychiatric Center, Taipei City Hospital, Taipei; Chian-Jue Kuo, MD, PhD, Taipei City Psychiatric Center, Taipei City Hospital and Department of Psychiatry, School of Medicine, Taipei Medical University and Psychiatric Research Center, Taipei Medical University Hospital, Taipei, Taiwan; Ying-Yeh Chen, MD, ScD, Taipei City Psychiatric Center, Taipei City Hospital and Institute of Public Health and Department of Public Health, National Yang-Ming University, Taipei, Taiwan
| | - Chian-Jue Kuo
- Yen-Ni Hung, PhD, School of Gerontology Health Management and Master Program in Long-Term Care, College of Nursing, Taipei Medical University, Department of Nursing, School of Nursing, National Yang-Ming University and Department of Education and Research, Taipei City Hospital, Taipei; Shu-Yu Yang, PhD, Taipei City Psychiatric Center, Taipei City Hospital, Taipei and Graduate Institute of Clinical Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan; Ming-Chyi Huang, MD, PhD, Taipei City Psychiatric Center, Taipei City Hospital and Department of Psychiatry, School of Medicine, Taipei Medical University, Taipei; For-Wey Lung, MD, PhD, Taipei City Psychiatric Center, Taipei City Hospital, Taipei; Shih-Ku Lin, MD,Taipei City Psychiatric Center, Taipei City Hospital and Department of Psychiatry, School of Medicine, Taipei Medical University, Taipei; Kuan-Yu Chen, MD, PhD, Taipei City Psychiatric Center, Taipei City Hospital, Taipei; Chian-Jue Kuo, MD, PhD, Taipei City Psychiatric Center, Taipei City Hospital and Department of Psychiatry, School of Medicine, Taipei Medical University and Psychiatric Research Center, Taipei Medical University Hospital, Taipei, Taiwan; Ying-Yeh Chen, MD, ScD, Taipei City Psychiatric Center, Taipei City Hospital and Institute of Public Health and Department of Public Health, National Yang-Ming University, Taipei, Taiwan
| | - Ying-Yeh Chen
- Yen-Ni Hung, PhD, School of Gerontology Health Management and Master Program in Long-Term Care, College of Nursing, Taipei Medical University, Department of Nursing, School of Nursing, National Yang-Ming University and Department of Education and Research, Taipei City Hospital, Taipei; Shu-Yu Yang, PhD, Taipei City Psychiatric Center, Taipei City Hospital, Taipei and Graduate Institute of Clinical Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan; Ming-Chyi Huang, MD, PhD, Taipei City Psychiatric Center, Taipei City Hospital and Department of Psychiatry, School of Medicine, Taipei Medical University, Taipei; For-Wey Lung, MD, PhD, Taipei City Psychiatric Center, Taipei City Hospital, Taipei; Shih-Ku Lin, MD,Taipei City Psychiatric Center, Taipei City Hospital and Department of Psychiatry, School of Medicine, Taipei Medical University, Taipei; Kuan-Yu Chen, MD, PhD, Taipei City Psychiatric Center, Taipei City Hospital, Taipei; Chian-Jue Kuo, MD, PhD, Taipei City Psychiatric Center, Taipei City Hospital and Department of Psychiatry, School of Medicine, Taipei Medical University and Psychiatric Research Center, Taipei Medical University Hospital, Taipei, Taiwan; Ying-Yeh Chen, MD, ScD, Taipei City Psychiatric Center, Taipei City Hospital and Institute of Public Health and Department of Public Health, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
241
|
Refining and integrating schizophrenia pathophysiology – Relevance of the allostatic load concept. Neurosci Biobehav Rev 2014; 45:183-201. [DOI: 10.1016/j.neubiorev.2014.06.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 04/02/2014] [Accepted: 06/09/2014] [Indexed: 12/20/2022]
|
242
|
Haarman BCMB, Riemersma-Van der Lek RF, de Groot JC, Ruhé HGE, Klein HC, Zandstra TE, Burger H, Schoevers RA, de Vries EFJ, Drexhage HA, Nolen WA, Doorduin J. Neuroinflammation in bipolar disorder - A [(11)C]-(R)-PK11195 positron emission tomography study. Brain Behav Immun 2014; 40:219-25. [PMID: 24703991 DOI: 10.1016/j.bbi.2014.03.016] [Citation(s) in RCA: 147] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 03/11/2014] [Accepted: 03/23/2014] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND The "monocyte-T-cell theory of mood disorders" regards neuroinflammation, i.e. marked activation of microglia, as a driving force in bipolar disorder. Microglia activation can be visualized in vivo using [(11)C]-(R)-PK11195 PET. Indirect evidence suggests the hippocampus as a potential focus of neuroinflammation in bipolar disorder. We aim to determine if there is increased [(11)C]-(R)-PK11195 binding to activated microglia in the hippocampus of patients with bipolar I disorder when compared to healthy controls. MATERIAL AND METHODS Fourteen patients with bipolar I disorder and eleven healthy controls were included in the analyses. Dynamic 60-min PET scans were acquired after the injection of [(11)C]-(R)-PK11195. All subjects underwent psychiatric interviews as well as an MRI scan, which was used for anatomic co-registration in the data analysis. The data from the PET scans was analyzed with a two-tissue-compartment model to calculate the binding potential, using the metabolite-corrected plasma and blood curve as input. RESULTS A significantly increased [(11)C]-(R)-PK11195 binding potential, which is indicative of neuroinflammation, was found in the right hippocampus of the patients when compared to the healthy controls (1.66 (CI 1.45-1.91) versus 1.33 (CI 1.16-1.53); p=0.033, respectively). Although the same trend was observed in the left hippocampus, this difference was not statistically significant. CONCLUSION This study is the first to demonstrate the presence of focal neuroinflammation in the right hippocampus in bipolar I disorder.
Collapse
Affiliation(s)
| | - Rixt F Riemersma-Van der Lek
- University of Groningen, University Medical Center Groningen, Department of Psychiatry, Groningen, The Netherlands
| | - Jan Cees de Groot
- University of Groningen, University Medical Center Groningen, Department of Radiology, Groningen, The Netherlands
| | - Henricus G Eric Ruhé
- University of Groningen, University Medical Center Groningen, Department of Psychiatry, Groningen, The Netherlands
| | - Hans C Klein
- University of Groningen, University Medical Center Groningen, Department of Psychiatry, Groningen, The Netherlands; University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, Groningen, The Netherlands
| | - Tjitske E Zandstra
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, Groningen, The Netherlands
| | - Huibert Burger
- University of Groningen, University Medical Center Groningen, Department of Psychiatry, Groningen, The Netherlands; University of Groningen, University Medical Center Groningen, Department of General Practice, Groningen, The Netherlands
| | - Robert A Schoevers
- University of Groningen, University Medical Center Groningen, Department of Psychiatry, Groningen, The Netherlands
| | - Erik F J de Vries
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, Groningen, The Netherlands
| | - Hemmo A Drexhage
- Erasmus MC, Department of Immunology, Rotterdam, The Netherlands
| | - Willem A Nolen
- University of Groningen, University Medical Center Groningen, Department of Psychiatry, Groningen, The Netherlands
| | - Janine Doorduin
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, Groningen, The Netherlands
| |
Collapse
|
243
|
Re-establishment of anxiety in stress-sensitized mice is caused by monocyte trafficking from the spleen to the brain. Biol Psychiatry 2014; 75:970-81. [PMID: 24439304 PMCID: PMC4084643 DOI: 10.1016/j.biopsych.2013.11.029] [Citation(s) in RCA: 231] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Revised: 10/31/2013] [Accepted: 11/18/2013] [Indexed: 11/22/2022]
Abstract
BACKGROUND Persistent anxiety-like symptoms may have an inflammatory-related pathophysiology. Our previous work using repeated social defeat (RSD) in mice showed that recruitment of peripheral myeloid cells to the brain is required for the development of anxiety. Here, we aimed to determine if 1) RSD promotes prolonged anxiety through redistribution of myeloid cells and 2) prior exposure to RSD sensitizes the neuroimmune axis to secondary subthreshold stress. METHODS Mice were subjected to RSD and several immune and behavioral parameters were determined .5, 8, or 24 days later. In follow-up studies, control and RSD mice were subjected to subthreshold stress at 24 days. RESULTS Repeated social defeat-induced macrophage recruitment to the brain corresponded with development and maintenance of anxiety-like behavior 8 days after RSD, but neither remained at 24 days. Nonetheless, social avoidance and an elevated neuroinflammatory profile were maintained at 24 days. Subthreshold social defeat in RSD-sensitized mice increased peripheral macrophage trafficking to the brain that promoted re-establishment of anxiety. Moreover, subthreshold social defeat increased social avoidance in RSD-sensitized mice compared with naïve mice. Stress-induced monocyte trafficking was linked to redistribution of myeloid progenitor cells in the spleen. Splenectomy before subthreshold stress attenuated macrophage recruitment to the brain and prevented anxiety-like behavior in RSD-sensitized mice. CONCLUSIONS These data indicate that monocyte trafficking from the spleen to the brain contributes re-establishment of anxiety in stress-sensitized mice. These findings show that neuroinflammatory mechanisms promote mood disturbances following stress-sensitization and outline novel neuroimmune interactions that underlie recurring anxiety disorders such as posttraumatic stress disorder.
Collapse
|
244
|
Inhibition of stress-induced hepatic tryptophan 2,3-dioxygenase exhibits antidepressant activity in an animal model of depressive behaviour. Int J Neuropsychopharmacol 2014; 17:917-28. [PMID: 24472498 DOI: 10.1017/s1461145713001673] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The role of hepatic tryptophan 2,3 dioxygenase (TDO) was assessed in the provocation of stress-induced depression-related behaviour in the rat. TDO drives tryptophan metabolism via the kynurenine pathway (KP) and leads to the production of neuroactive metabolites including kynurenine. A single 2 h period of restraint stress in adult male Sprague-Dawley rats provoked an increase in circulating concentrations of the glucocorticoid corticosterone and induction of hepatic TDO expression and activity. Repeated exposure to stress (10 d of 2 h restraint each day) provoked an increase in immobility in the forced swimming test (FST) indicative of depression-related behaviour. Immobility was accompanied by an increase in the circulating corticosterone concentrations, expression and activity of hepatic TDO and increase in the expression of TDO in the cerebral cortex. Increased TDO activity was associated with raised circulating kynurenine concentrations and a reduction in circulating tryptophan concentrations indicative of KP activation. Co-treatment with the TDO inhibitor allopurinol (20 mg/kg, i.p.), attenuated the chronic stress-related increase in immobility in the FST and the accompanying increase in circulating kynurenine concentrations. These findings indicate that stress-induced corticosterone and consequent activation of hepatic TDO, tryptophan metabolism and production of kynurenine provoke a depression-related behavioural phenotype. Inhibition of stress-related hepatic TDO activity promotes antidepressant activity. TDO may therefore represent a promising target for the treatment of depression associated with stress-related disorders in which there is evidence for KP activation.
Collapse
|
245
|
O'Connor TG, Moynihan JA, Caserta MT. Annual research review: The neuroinflammation hypothesis for stress and psychopathology in children--developmental psychoneuroimmunology. J Child Psychol Psychiatry 2014; 55:615-31. [PMID: 24372371 PMCID: PMC4029900 DOI: 10.1111/jcpp.12187] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/06/2013] [Indexed: 12/14/2022]
Abstract
Experimental animal and adult human data suggest that stress exposure is associated with alterations in immune system function that may underlie increased susceptibility to disease and behavioral disorders. The implications of these data for child psychology and psychiatry are not yet clear. The current review seeks to distil and translate the relevant animal and adult human work to children to advance a developmental model of psychoneuroimmunology. In addition to reviewing key specific findings, we consider biological/conceptual models and technical aspects of psychoneuroimmunology work in pediatric populations, and outline the rationales and advantages of integrating hypotheses concerning neuroinflammation in developmental studies of psychopathology.
Collapse
|
246
|
Luo G, Wang S, Li Z, Wei R, Zhang L, Liu H, Wang C, Niu R, Wang J. Maternal bisphenol a diet induces anxiety-like behavior in female juvenile with neuroimmune activation. Toxicol Sci 2014; 140:364-73. [PMID: 24824810 DOI: 10.1093/toxsci/kfu085] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Maternal Bisphenol A (BPA) diet triggers anxiety in rodents, but the underlying mechanism is still unclear. Accumulating epidemiological and experimental data have demonstrated that the anxiety is associated with aberrant neuroimmune response. In this study, we found that maternal BPA diet (MBD) exacerbated anxiety-like behavior in female juvenile mice, and the molecular evidence further showed that this behavioral phenotype was connected to the neuroimmune activation, such as elevated tumor necrosis factor alpha (TNF-α) and interleukin (IL)-6 levels in prefrontal cortex (PFC) rather than in peripheral blood, which indicated that neuroimmune response might be ascribed to neuroglial activation because activated neuroglia cells could secrete proinflammatory cytokines. Subsequently, we found that ionized calcium-binding adapter molecule (Iba)-1 as a selective marker for microglia and glial fibrillary acidic protein as a specific marker for astrocyte were significantly increased at transcriptional and translational levels, which confirmed the neuroglial activation in this model. Therefore, we conclude that MBD induces excessive anxiety-like behavior in female juvenile with elevated TNF-α and IL-6 levels, as well as activated microglia and astrocyte in PFC. Herein caution must be taken to prevent potential risks from MBD becuase exacerbated anxiety-like behavior in female juvenile by MBD may be a critical contribution for subsequent growth or mental disorders.
Collapse
Affiliation(s)
- Guangying Luo
- Shanxi Key Laboratory of Environmental Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, PR China
| | - Shaolin Wang
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, Virginia 22911
| | - Zhigang Li
- Shanxi Key Laboratory of Environmental Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, PR China Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China
| | - Ruifen Wei
- Shanxi Key Laboratory of Environmental Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, PR China
| | - Lianjie Zhang
- Shanxi Key Laboratory of Environmental Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, PR China
| | - Huanhuan Liu
- Shanxi Key Laboratory of Environmental Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, PR China
| | - Chong Wang
- Shanxi Key Laboratory of Environmental Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, PR China
| | - Ruiyan Niu
- Shanxi Key Laboratory of Environmental Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, PR China
| | - Jundong Wang
- Shanxi Key Laboratory of Environmental Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, PR China
| |
Collapse
|
247
|
McGuire JL, Hammond JH, Yates SD, Chen D, Haroutunian V, Meador-Woodruff JH, McCullumsmith RE. Altered serine/threonine kinase activity in schizophrenia. Brain Res 2014; 1568:42-54. [PMID: 24780530 DOI: 10.1016/j.brainres.2014.04.029] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Revised: 04/19/2014] [Accepted: 04/21/2014] [Indexed: 10/25/2022]
Abstract
Converging evidence implicates alterations in multiple signaling pathways in the etiology of schizophrenia. Previously, these studies were limited to the analysis of one or a few phosphoproteins at a time. Here, we use a novel kinase array platform to simultaneously investigate the convergence of multiple signaling cascades implicated in schizophrenia. This technology uses consensus peptide substrates to assess activity levels of a large number (>100) of serine/threonine protein kinases. 19 peptide substrates were differentially phosphorylated (>15% change) in the frontal cortex in schizophrenia. These peptide substrates were examined using Ingenuity Pathway Analysis to group them according to the functions and to identify processes most likely affected in schizophrenia. Pathway analysis placed 14 of the 19 peptides into cellular homeostatic pathways, 10 into pathways governing cytoskeletal organization, and 8 into pathways governing ion homeostasis. These data are the first to simultaneously investigate comprehensive changes in signaling cascades in a severe psychiatric disorder. The examination of kinase activity in signaling pathways may facilitate the identification of novel substrates for drug discovery and the development of safer and more effective pharmacological treatment for schizophrenia.
Collapse
Affiliation(s)
- Jennifer L McGuire
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH, USA.
| | - John H Hammond
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Stefani D Yates
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Dongquan Chen
- Division of Preventative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Vahram Haroutunian
- Department of Psychiatry, Mount Sinai School of Medicine, New York, NY, USA.
| | - James H Meador-Woodruff
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Robert E McCullumsmith
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
248
|
Ma M, Ren Q, Zhang JC, Hashimoto K. Effects of Brilliant Blue G on Serum Tumor Necrosis Factor-α Levels and Depression-like Behavior in Mice after Lipopolysaccharide Administration. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE 2014; 12:31-6. [PMID: 24851118 PMCID: PMC4022763 DOI: 10.9758/cpn.2014.12.1.31] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 06/03/2013] [Accepted: 06/12/2013] [Indexed: 01/07/2023]
Abstract
OBJECTIVE Accumulating evidence suggests that inflammation plays a role in the pathophysiology of major depression. The adenosine triphosphate (ATP)-sensitive P2X7 receptor (P2X7R) plays a crucial role in microglial activation caused by inflammation. The dye brilliant blue G (BBG) is a P2X7R antagonist. This study examined whether BBG shows antidepressant effects in an inflammation-induced model of depression. METHODS We examined the effects of BBG (12.5, 25, or 50 mg/kg) on serum tumor necrosis factor-α (TNF-α) levels after administering the bacterial endotoxin lipopolysaccharide (LPS; 0.5 mg/kg) and the effects of BBG (50 mg/kg) on depression-like behavior in the tail-suspension test (TST) and forced swimming test (FST). RESULTS Pretreatment with BBG (12.5, 25, or 50 mg/kg) significantly blocked the increase in serum TNF-α levels after a single dose of LPS (0.5 mg/kg). Furthermore, BBG (50 mg/kg) significantly attenuated the increase in immobility time in the TST and FST after LPS (0.5 mg/kg) administration. CONCLUSION The results suggest that BBG has anti-inflammatory and antidepressant effects in mice after LPS administration. Therefore, P2X7R antagonists are potential therapeutic drugs for inflammation-related major depression.
Collapse
Affiliation(s)
- Min Ma
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Qian Ren
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Ji-Chun Zhang
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| |
Collapse
|
249
|
Knockdown of interleukin-1 receptor type-1 on endothelial cells attenuated stress-induced neuroinflammation and prevented anxiety-like behavior. J Neurosci 2014; 34:2583-91. [PMID: 24523548 DOI: 10.1523/jneurosci.3723-13.2014] [Citation(s) in RCA: 159] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Interleukin-1β (IL-1β) is an inflammatory cytokine that plays a prominent role in stress-induced behavioral changes. In a model of repeated social defeat (RSD), elevated IL-1β expression in the brain was associated with recruitment of primed macrophages that were necessary for development of anxiety-like behavior. Moreover, microglia activation and anxiety-like behavior associated with RSD did not occur in IL-1 receptor type-1 knock-out (IL-1R1(KO)) mice. Therefore, the objective of this study was to examine the role of IL-1 signaling in RSD-induced macrophage trafficking to the brain and anxiety-like behavior. Initial studies revealed that RSD did not increase circulating myeloid cells in IL-1R1(KO) mice, resulting in limited macrophage trafficking to the brain. In addition, IL-1R1(KO) bone marrow-chimera mice showed that IL-1R1 expression was essential for macrophage trafficking into the brain. To differentiate cellular mediators of stress-induced IL-1 signaling, endothelial-specific IL-1R1 knock-down (eIL-1R1kd) mice were used. Both wild-type (WT) and eIL-1R1kd mice had increased circulating monocytes, recruitment of macrophages to the brain, and altered microglia activation after RSD. Nonetheless, RSD-induced expression of IL-1β, TNF-α, and IL-6 mRNA in brain CD11b(+) cells was attenuated in eIL-1R1kd mice compared with WT. Moreover, anxiety-like behavior did not develop in eIL-1R1kd mice. Collectively, these findings demonstrated that there was limited RSD-induced priming of myeloid cells in IL-1R1(KO) mice and disrupted propagation of neuroinflammatory signals in the brain of eIL-1R1kd mice. Furthermore, these data showed that transduction of IL-1 signaling by endothelial cells potentiates stress-induced neuroinflammation and promotes anxiety-like behavior.
Collapse
|
250
|
Qin J, Zhang RX, Li JL, Wang JX, Hou J, Yang X, Zhu WL, Shi J, Lu L. cRGD mediated liposomes enhanced antidepressant-like effects of edaravone in rats. Eur J Pharm Sci 2014; 58:63-71. [PMID: 24704101 DOI: 10.1016/j.ejps.2014.03.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2014] [Revised: 03/13/2014] [Accepted: 03/25/2014] [Indexed: 11/28/2022]
Abstract
The delayed onset of therapeutic outcomes is a major drawback of the current antidepressants. The blood-brain barrier is the most important bottleneck impeding drug transport into the brain. Therefore, development of novel antidepressant medications with rapid onset and sustained activity is urgent. RGD liposomes showed an excellent effect of brain-targeting drug delivery and increased the entering rate to the brain. In the present study, we prepared cyclic RGD liposomes loaded with edaravone (cRGD-ERLs) and evaluated the potential antidepressant-like effects of this drug delivery system in rats. The results showed single injection of cRGD-ERLs produced significant antidepressant-like effects in both forced swim and novelty suppressed feeding test. Moreover, acute cRGD-ERLs increased the expression of c-fos in the medial prefrontal cortex, suggesting that cRGD-ERLs could activate the neuronal function. Furthermore, cRGD-ERLs reversed the increase of lipopolysaccharides (LPS)-induced plasma cytokine IL-1β and IL-6, suggesting that normalization of cytokine level might be involved in the behavioral response of cRGD-ERLs. Finally, cRGD-ERLs prevented the increase of immobility induced by LPS in the forced swim test. Overall, the current data revealed a novel brain-target drug delivery system, which can be used to improve the therapeutic outcomes of antidepressants by increase of crossing rate to the brain.
Collapse
Affiliation(s)
- Jing Qin
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Ruo-Xi Zhang
- National Institute on Drug Dependence, Peking University, Beijing 100191, China
| | - Jia-Li Li
- National Institute on Drug Dependence, Peking University, Beijing 100191, China
| | - Jian-Xin Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Jia Hou
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Xu Yang
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Wei-Li Zhu
- National Institute on Drug Dependence, Peking University, Beijing 100191, China.
| | - Jie Shi
- National Institute on Drug Dependence, Peking University, Beijing 100191, China
| | - Lin Lu
- National Institute on Drug Dependence, Peking University, Beijing 100191, China
| |
Collapse
|