51
|
Herman FJ, Simkovic S, Pasinetti GM. Neuroimmune nexus of depression and dementia: Shared mechanisms and therapeutic targets. Br J Pharmacol 2019; 176:3558-3584. [PMID: 30632147 DOI: 10.1111/bph.14569] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 11/26/2018] [Accepted: 12/04/2018] [Indexed: 12/12/2022] Open
Abstract
Dysfunctional immune activity is a physiological component of both Alzheimer's disease (AD) and major depressive disorder (MDD). The extent to which altered immune activity influences the development of their respective cognitive symptoms and neuropathologies remains under investigation. It is evident, however, that immune activity affects neuronal function and circuit integrity. In both disorders, alterations are present in similar immune networks and neuroendocrine signalling pathways, immune responses persist in overlapping neuroanatomical locations, and morphological and structural irregularities are noted in similar domains. Epidemiological studies have also linked the two disorders, and their genetic and environmental risk factors intersect along immune-activating pathways and can be synonymous with one another. While each of these disorders individually contains a large degree of heterogeneity, their shared immunological components may link distinct phenotypes within each disorder. This review will therefore highlight the shared immune pathways of AD and MDD, their overlapping neuroanatomical features, and previously applied, as well as novel, approaches to pharmacologically manipulate immune pathways, in each neurological condition. LINKED ARTICLES: This article is part of a themed section on Therapeutics for Dementia and Alzheimer's Disease: New Directions for Precision Medicine. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.18/issuetoc.
Collapse
Affiliation(s)
- Francis J Herman
- Department of Neurology, Mount Sinai School of Medicine, New York City, New York, USA
| | - Sherry Simkovic
- Department of Neurology, Mount Sinai School of Medicine, New York City, New York, USA
| | - Giulio M Pasinetti
- Department of Neurology, Mount Sinai School of Medicine, New York City, New York, USA.,Geriatrics Research. Education, and Clinical Center, JJ Peters VA Medical Center, Bronx, New York, USA
| |
Collapse
|
52
|
Ghosh S, Choudhury S, Mukherjee S, Gupta P, Chowdhury O, Baral R, Chattopadhyay S. Fluoxetine triggers selective apoptosis in inflammation‐induced proliferating (Ki‐67
high
) thymocytes. Immunol Cell Biol 2019; 97:470-484. [DOI: 10.1111/imcb.12227] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 11/08/2017] [Accepted: 12/19/2018] [Indexed: 12/27/2022]
Affiliation(s)
- Sayan Ghosh
- Department of Physiology University of Calcutta UCSTA 92, Acharya Prafulla Chandra Road 700009 Kolkata India
| | - Sreetama Choudhury
- Department of Physiology University of Calcutta UCSTA 92, Acharya Prafulla Chandra Road 700009 Kolkata India
| | - Sudeshna Mukherjee
- Department of Physiology University of Calcutta UCSTA 92, Acharya Prafulla Chandra Road 700009 Kolkata India
| | - Payal Gupta
- Department of Physiology University of Calcutta UCSTA 92, Acharya Prafulla Chandra Road 700009 Kolkata India
| | - Olivia Chowdhury
- Department of Physiology University of Calcutta UCSTA 92, Acharya Prafulla Chandra Road 700009 Kolkata India
| | - Rathindranath Baral
- Chittaranjan National Cancer Institute 37, S.P. Mukherjee Road 700026 Kolkata India
| | - Sreya Chattopadhyay
- Department of Physiology University of Calcutta UCSTA 92, Acharya Prafulla Chandra Road 700009 Kolkata India
- Centre for Research in Nanoscience and Nanotechnology University of Calcutta JD‐2, Salt Lake, Sector III 700098 Kolkata India
| |
Collapse
|
53
|
Wu H, Denna TH, Storkersen JN, Gerriets VA. Beyond a neurotransmitter: The role of serotonin in inflammation and immunity. Pharmacol Res 2019; 140:100-114. [DOI: 10.1016/j.phrs.2018.06.015] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 06/01/2018] [Accepted: 06/17/2018] [Indexed: 12/16/2022]
|
54
|
Rheumatoid arthritis and depression: an inflammatory perspective. Lancet Psychiatry 2019; 6:164-173. [PMID: 30366684 DOI: 10.1016/s2215-0366(18)30255-4] [Citation(s) in RCA: 248] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 06/11/2018] [Accepted: 06/20/2018] [Indexed: 12/13/2022]
Abstract
The coexistence of immune-mediated inflammatory diseases with depression has long been recognised. Data that illustrate the intimate associations between peripheral and brain immune responses raise the possibility of shared pathophysiological mechanisms. These associations include the negative effects of proinflammatory cytokines on monoaminergic neurotransmission, neurotrophic factors, and measures of synaptic plasticity. The evidence supporting this association is accumulating and includes findings from clinical trials of immunomodulatory therapy, indicating that these interventions can provide benefits to mental health independent of improvements in physical disease scores. In this Review, we assess this evidence in relation to rheumatoid arthritis and depression, with a focus on innate immune and molecular responses to inflammation, and discuss the challenges of assessing causation in this population, acknowledging the difficulty of assessing the confounding and contributory effects of pain and fatigue. We also discuss how future clinical and preclinical research might improve diagnosis of depression in people with rheumatoid arthritis and shed light on mechanisms that could be substrates for therapeutic interventions.
Collapse
|
55
|
Laumet G, Edralin JD, Chiang ACA, Dantzer R, Heijnen CJ, Kavelaars A. Resolution of inflammation-induced depression requires T lymphocytes and endogenous brain interleukin-10 signaling. Neuropsychopharmacology 2018; 43:2597-2605. [PMID: 30054585 PMCID: PMC6224384 DOI: 10.1038/s41386-018-0154-1] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 06/18/2018] [Accepted: 07/09/2018] [Indexed: 01/13/2023]
Abstract
In humans, depression is often associated with low-grade inflammation, activation of the tryptophan/kynurenine pathway, and mild lymphopenia. Preclinical research confirms that inflammation induces depression-like behavior through activation of the tryptophan/kynurenine pathway. However, the mechanisms governing recovery from depression are unknown. Understanding the pathways leading to resolution of depression will likely lead to identification of novel targets for treatment. We investigated the contribution of T lymphocytes to the resolution of lipopolysaccharide-induced depression-like behavior. Duration of depression-like behavior was markedly prolonged in mice without mature T or B lymphocytes (Rag1-/- mice). This prolonged depression-like behavior was associated with persistent upregulation of the tryptophan-metabolizing enzyme indoleamine-2,3-dioxygenase (Ido)1 in the prefrontal cortex (PFC). Reconstitution of Rag1-/- mice with T lymphocytes normalized resolution of depression-like behavior and expression of Ido1 in the PFC. During resolution of inflammation-induced depression-like behavior, T lymphocytes accumulated in the meninges and were required for induction of interleukin (IL)-10 in the meninges and the PFC. Inhibition of IL-10 signaling by nasal administration of neutralizing anti-IL-10 antibody to WT mice led to persistent upregulation of Ido1 in the PFC and prolonged depression-like behavior. Conversely, nasal administration of recombinant IL-10 in Rag1-/- mice normalized Ido1 expression and resolution of depression-like behavior. In conclusion, the present data show for the first time that resolution of inflammation-induced depression is an active process requiring T lymphocytes acting via an IL-10-dependent pathway to decrease Ido1 expression in the brain. We propose that targeting the T lymphocyte/IL-10 resolution pathway could represent a novel approach to promote recovery from major depressive disorder.
Collapse
Affiliation(s)
- Geoffroy Laumet
- 0000 0001 2291 4776grid.240145.6Laboratory of Neuroimmunology, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, 1515 Holcombe Blvd., Houston, TX 77030 USA
| | - Jules Daniel Edralin
- 0000 0001 2291 4776grid.240145.6Laboratory of Neuroimmunology, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, 1515 Holcombe Blvd., Houston, TX 77030 USA
| | - Angie Chi-An Chiang
- 0000 0001 2291 4776grid.240145.6Laboratory of Neuroimmunology, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, 1515 Holcombe Blvd., Houston, TX 77030 USA
| | - Robert Dantzer
- 0000 0001 2291 4776grid.240145.6Laboratory of Neuroimmunology, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, 1515 Holcombe Blvd., Houston, TX 77030 USA
| | - Cobi J. Heijnen
- 0000 0001 2291 4776grid.240145.6Laboratory of Neuroimmunology, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, 1515 Holcombe Blvd., Houston, TX 77030 USA
| | - Annemieke Kavelaars
- Laboratory of Neuroimmunology, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, 1515 Holcombe Blvd., Houston, TX, 77030, USA.
| |
Collapse
|
56
|
Elevated Levels of Serum IL-17A in Community-Dwelling Women with Higher Depressive Symptoms. Behav Sci (Basel) 2018; 8:bs8110102. [PMID: 30400354 PMCID: PMC6262626 DOI: 10.3390/bs8110102] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 10/31/2018] [Accepted: 10/31/2018] [Indexed: 12/26/2022] Open
Abstract
Recent studies indicate that patients with depression have increased concentrations of serum pro-inflammatory cytokines. However, studies of IL-17 and studies on community-dwellers are few. The purpose of this study was to investigate serum cytokine levels, especially IL-17A, among subjects with high and low depressive symptoms of a general population. The participants comprised 20 female community-dwellers aged 40 years or older who contributed to a Shika study in Ishikawa, Japan. Ten participants who showed higher and ten who showed lower depressive symptoms among 208 females assessed by the Japanese version of the Centre for Epidemiologic Studies Depression Scale (CES-D) were selected for this study. Serum samples were analyzed for TNF-alpha, IL-6, IL-10, IL-12, and IL-17A using a multiplex Luminex analysis. For the comparison between the high and low depressive groups statistically, linear regression analyses were applied. The serum level of IL-17A was significantly higher among the high depressive participants (p < 0.05) even after controlling possible confounders, whereas there were no differences in TNF-alpha, IL-6, IL-10, or IL-12 between the high and low depressive groups. Our findings supported an association between serum IL-17A levels and depressive symptoms. Peripheral IL-17A immune response may be a preventive and treatment target for depression.
Collapse
|
57
|
Dazzan P, Fusté M, Davies W. Do Defective Immune System-Mediated Myelination Processes Increase Postpartum Psychosis Risk? Trends Mol Med 2018; 24:942-949. [PMID: 30348609 PMCID: PMC6224363 DOI: 10.1016/j.molmed.2018.09.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 09/12/2018] [Accepted: 09/14/2018] [Indexed: 12/17/2022]
Abstract
Postpartum (or puerperal) psychosis (PP) is a rare, severe psychiatric disorder that affects women shortly after childbirth; risk is particularly high in individuals with a history of bipolar disorder or PP, but the underlying pathophysiology remains poorly understood. Emerging evidence suggests that immune system (dys)function plays an important role in disorder onset. On the basis of new findings from clinical and animal model studies, we hypothesise that the abundance and/or activity of regulatory T cells, and the efficacy of consequent (re)myelination processes in the brain mediated by CCN proteins, is perturbed in PP; this pathway may be modulated by risk and protective/treatment factors for the disorder, and identifying abnormalities within it could signpost novel predictive biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Paola Dazzan
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK; National Institute for Health Research (NIHR) Mental Health Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London, London, UK
| | - Montserrat Fusté
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | - William Davies
- Medical Research Council Centre for Neuropsychiatric Genetics and Genomics and Neuroscience and Mental Health Research Institute, Schools of Medicine and Psychology, Cardiff University, Cardiff, UK.
| |
Collapse
|
58
|
Kelly DL, Li X, Kilday C, Feldman S, Clark S, Liu F, Buchanan RW, Tonelli LH. Increased circulating regulatory T cells in medicated people with schizophrenia. Psychiatry Res 2018; 269:517-523. [PMID: 30195746 PMCID: PMC6207456 DOI: 10.1016/j.psychres.2018.09.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 08/01/2018] [Accepted: 09/03/2018] [Indexed: 01/01/2023]
Abstract
Immunological abnormalities are increasingly reported in people with schizophrenia, but no clear functional biomarkers associated with genetic correlates of the disease have been found. Regulatory T cells (Tregs) are key immunoregulatory cells involved in the control of inflammatory processes and their functions are directly related to the human leucocyte antigen (HLA) gene, which has been implicated in schizophrenia genetic studies. However, there is a lack of studies reporting Treg status in people with schizophrenia. In the current study, the proportion of circulating Tregs was examined using flow cytometry in 26 medicated participants with schizophrenia and 17 healthy controls. Psychiatric symptoms and cognitive function were evaluated using the Scale for the Assessment of Negative Symptoms, the Brief Psychiatric Rating Scale, and the MATRICS Consensus Cognitive Battery. The proportion of Tregs was found to be significantly greater in the schizophrenia group compared to healthy controls. No differences were observed in total lymphocyte counts or CD3+ and CD4+ T cells, confirming a specific effect for Tregs. Elevated Tregs in schizophrenia correlated with fewer negative symptoms, a core domain of the illness. These results suggest that Tregs may contribute to improved negative symptoms in schizophrenia, possibly by counteracting on-going inflammatory processes.
Collapse
Affiliation(s)
- Deanna L Kelly
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Xin Li
- Laboratory of Behavioral Neuroimmunology, Department of Psychiatry, University of Maryland School of Medicine, 685 West Baltimore Street, MSTF Building Room 934 E, Baltimore 21201, MD, USA
| | - Catherine Kilday
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Stephanie Feldman
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Sarah Clark
- Laboratory of Behavioral Neuroimmunology, Department of Psychiatry, University of Maryland School of Medicine, 685 West Baltimore Street, MSTF Building Room 934 E, Baltimore 21201, MD, USA
| | - Fang Liu
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Robert W Buchanan
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Leonardo H Tonelli
- Laboratory of Behavioral Neuroimmunology, Department of Psychiatry, University of Maryland School of Medicine, 685 West Baltimore Street, MSTF Building Room 934 E, Baltimore 21201, MD, USA.
| |
Collapse
|
59
|
Clark SM, Song C, Li X, Keegan AD, Tonelli LH. CD8 + T cells promote cytokine responses to stress. Cytokine 2018; 113:256-264. [PMID: 30033139 DOI: 10.1016/j.cyto.2018.07.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 06/19/2018] [Accepted: 07/12/2018] [Indexed: 11/27/2022]
Abstract
Psychological stress is known to have profound effects on immune function and to promote inflammatory conditions. Elevated circulating levels of cytokines associated with stress are known to increase the risk to several diseases, but little is known about this mechanism. This study assessed the role of T cells on cytokine levels after exposure to stress in the learned helplessness paradigm. Adoptive transfer of CD4+ T cells into Rag2-/- mice did not change cytokine levels to stress while CD8+ T cells resulted in an increase in TNF-α, IL-6 and IFN-γ in stressed Rag2-/- mice. Moreover, depletion of CD8+ T cells in WT mice abolished these cytokine responses to stress. Corticosterone and behavioral stress responsiveness was impaired in Rag2-/- mice reconstituted with CD8+ T cells. Notably, depletion of these cells in WT mice had no effect on behavior or corticosterone levels. Exposure to stress did not change the expression of canonical markers of T cell activation including CD62L and CD44 or modified intracellular cytokine content, suggesting that they are not the main producers of circulating cytokines in response to stress. These results show that CD8+ T cells promote TNF-α, IL-6 and IFN-γ responses to stress, possibly by stimulating non-lymphoid cells.
Collapse
Affiliation(s)
- Sarah M Clark
- Laboratory of Behavioral Neuroimmunology, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA; Research and Development Service, Department of Veterans Affairs, VA Maryland Health Care System, Baltimore, MD, USA
| | - Chang Song
- Laboratory of Behavioral Neuroimmunology, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Xin Li
- Laboratory of Behavioral Neuroimmunology, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Achsah D Keegan
- Center for Vascular and Inflammatory Diseases, Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA; Research and Development Service, Department of Veterans Affairs, VA Maryland Health Care System, Baltimore, MD, USA
| | - Leonardo H Tonelli
- Laboratory of Behavioral Neuroimmunology, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA; Research and Development Service, Department of Veterans Affairs, VA Maryland Health Care System, Baltimore, MD, USA.
| |
Collapse
|
60
|
Sacramento PM, Monteiro C, Dias ASO, Kasahara TM, Ferreira TB, Hygino J, Wing AC, Andrade RM, Rueda F, Sales MC, Vasconcelos CC, Bento CAM. Serotonin decreases the production of Th1/Th17 cytokines and elevates the frequency of regulatory CD4 + T-cell subsets in multiple sclerosis patients. Eur J Immunol 2018; 48:1376-1388. [PMID: 29719048 DOI: 10.1002/eji.201847525] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 03/28/2018] [Accepted: 04/26/2018] [Indexed: 12/11/2022]
Abstract
Excessive levels of proinflammatory cytokines in the CNS are associated with reduced serotonin (5-HT) synthesis, a neurotransmitter with diverse immune effects. In this study, we evaluated the ability of exogenous 5-HT to modulate the T-cell behavior of patients with MS, a demyelinating autoimmune disease mediated by Th1 and Th17 cytokines. Here, 5-HT attenuated, in vitro, T-cell proliferation and Th1 and Th17 cytokines production in cell cultures from MS patients. Additionally, 5-HT reduced IFN-γ and IL-17 release by CD8+ T cells. By contrast, 5-HT increased IL-10 production by CD4+ T cells from MS patients. A more accurate analysis of these IL-10-secreting CD4+ T cells revealed that 5-HT favors the expansion of FoxP3+ CD39+ regulatory T cells (Tregs) and type 1 regulatory T cells. Notably, this neurotransmitter also elevated the frequency of Treg17 cells, a novel regulatory T-cell subset. The effect of 5-HT in upregulating CD39+ Treg and Treg17 cells was inversely correlated with the number of active brain lesions. Finally, in addition to directly reducing cytokine production by purified Th1 and Th17 cells, 5-HT enhanced in vitro Treg function. In summary, our data suggest that serotonin may play a protective role in the pathogenesis of MS.
Collapse
Affiliation(s)
- Priscila M Sacramento
- Post-graduate Program in Microbiology, Department of General Medicine Department, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.,Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Clarice Monteiro
- Post-graduate Program in Microbiology, Department of General Medicine Department, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.,Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Aleida S O Dias
- Post-graduate Program in Microbiology, Department of General Medicine Department, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.,Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Taissa M Kasahara
- Post-graduate Program in Microbiology, Department of General Medicine Department, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.,Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Thaís B Ferreira
- Post-graduate Program in Microbiology, Department of General Medicine Department, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.,Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Joana Hygino
- Post-graduate Program in Molecular and Cellular Biology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Cristina Wing
- Post-graduate Program in Neurology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Regis M Andrade
- Department of General Medicine Department, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernanda Rueda
- Clinical of Diagnosis by Image, Barra da Tijuca Unity, Rio de Janeiro, Brazil
| | - Marisa C Sales
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Cleonice A M Bento
- Post-graduate Program in Microbiology, Department of General Medicine Department, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.,Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.,Post-graduate Program in Molecular and Cellular Biology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
61
|
Ellul P, Mariotti-Ferrandiz E, Leboyer M, Klatzmann D. Regulatory T Cells As Supporters of Psychoimmune Resilience: Toward Immunotherapy of Major Depressive Disorder. Front Neurol 2018; 9:167. [PMID: 29615964 PMCID: PMC5869201 DOI: 10.3389/fneur.2018.00167] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 03/06/2018] [Indexed: 12/28/2022] Open
Abstract
There is growing evidence that inflammation plays a role in major depressive disorder (MDD). As the main role of regulatory T cells (Tregs) is to control inflammation, this might denote a Treg insufficiency in MDD. However, neither a qualitative nor a quantitative defect of Tregs has been ascertained and no causality direction between inflammation and depression has been established. Here, after reviewing the evidence supporting a relation between Treg insufficiency and MDD, we conclude that a novel therapeutic approach based on Treg stimulation could be valuable in at least the subset of patients with inflammatory MDD. Low-dose interleukin-2 appears to be a good candidate as it is not only a safe stimulator of Tregs in humans but also an inhibitor of pro-inflammatory Th17 lymphocytes. Here, we discuss that a thorough immune investigation as well as immunotherapy will be heuristic for deciphering the pathophysiology of MDD.
Collapse
Affiliation(s)
- Pierre Ellul
- Sorbonne Université, Assistance Publique - Hôpitaux de Paris (AP-HP), Robert Debré Hospital, Department of Child and Adolescent Psychiatry, Paris, France
| | - Encarnita Mariotti-Ferrandiz
- Sorbonne Université, INSERM, Immunology-Immunopathology-Immunotherapy (i3), AP-HP, Hôpital Pitié-Salpêtrière, Biotherapy (CIC-BTi) and Inflammation-Immunopathology-Biotherapy Department (i2B), Paris, France
| | - Marion Leboyer
- Team 15, INSERM U955 Institut Mondor de Recherche Biomédicale (IMRB), Créteil, France.,Faculté de Médecine, Université Paris-Est Créteil Val de Marne (UPEC), DHU PePSY, Pôle de Psychiatrie et d'addictologie, Hôpitaux Universitaires Mondor, Assistance Publique - Hôpitaux de Paris (AP-HP), Créteil, France.,Fondation FondaMental, Créteil, France
| | - David Klatzmann
- Sorbonne Université, INSERM, Immunology-Immunopathology-Immunotherapy (i3), AP-HP, Hôpital Pitié-Salpêtrière, Biotherapy (CIC-BTi) and Inflammation-Immunopathology-Biotherapy Department (i2B), Paris, France
| |
Collapse
|
62
|
Zhang X, Rocha-Ferreira E, Li T, Vontell R, Jabin D, Hua S, Zhou K, Nazmi A, Albertsson AM, Sobotka K, Ek J, Thornton C, Hagberg H, Mallard C, Leavenworth JW, Zhu C, Wang X. γδT cells but not αβT cells contribute to sepsis-induced white matter injury and motor abnormalities in mice. J Neuroinflammation 2017; 14:255. [PMID: 29262837 PMCID: PMC5738716 DOI: 10.1186/s12974-017-1029-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 12/08/2017] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Infection and sepsis are associated with brain white matter injury in preterm infants and the subsequent development of cerebral palsy. METHODS In the present study, we used a neonatal mouse sepsis-induced white matter injury model to determine the contribution of different T cell subsets (αβT cells and γδT cells) to white matter injury and consequent behavioral changes. C57BL/6J wild-type (WT), T cell receptor (TCR) δ-deficient (Tcrd -/-, lacking γδT cells), and TCRα-deficient (Tcra -/-, lacking αβT cells) mice were administered with lipopolysaccharide (LPS) at postnatal day (PND) 2. Brain myelination was examined at PNDs 12, 26, and 60. Motor function and anxiety-like behavior were evaluated at PND 26 or 30 using DigiGait analysis and an elevated plus maze. RESULTS White matter development was normal in Tcrd -/- and Tcrα -/- compared to WT mice. LPS exposure induced reductions in white matter tissue volume in WT and Tcrα -/- mice, but not in the Tcrd -/- mice, compared with the saline-treated groups. Neither LPS administration nor the T cell deficiency affected anxiety behavior in these mice as determined with the elevated plus maze. DigiGait analysis revealed motor function deficiency after LPS-induced sepsis in both WT and Tcrα -/- mice, but no such effect was observed in Tcrd -/- mice. CONCLUSIONS Our results suggest that γδT cells but not αβT cells contribute to sepsis-induced white matter injury and subsequent motor function abnormalities in early life. Modulating the activity of γδT cells in the early stages of preterm white matter injury might represent a novel therapeutic strategy for the treatment of perinatal brain injury.
Collapse
Affiliation(s)
- Xiaoli Zhang
- Henan Key Laboratory of Child Brain Injury, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.,Perinatal Center, Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30, Gothenburg, Sweden
| | - Eridan Rocha-Ferreira
- Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Tao Li
- Henan Key Laboratory of Child Brain Injury, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.,Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 436, 405 30, Gothenburg, Sweden
| | - Regina Vontell
- Department of Perinatal Imaging and Health, Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, SE1 7EH, UK
| | - Darakhshan Jabin
- Perinatal Center, Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30, Gothenburg, Sweden
| | - Sha Hua
- Perinatal Center, Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30, Gothenburg, Sweden.,Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University Medical School, Luwan Branch, Shanghai, China
| | - Kai Zhou
- Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Women's and Children's Health, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden
| | - Arshed Nazmi
- Perinatal Center, Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30, Gothenburg, Sweden
| | - Anna-Maj Albertsson
- Perinatal Center, Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30, Gothenburg, Sweden
| | - Kristina Sobotka
- Perinatal Center, Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30, Gothenburg, Sweden
| | - Joakim Ek
- Perinatal Center, Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30, Gothenburg, Sweden
| | - Claire Thornton
- Department of Perinatal Imaging and Health, Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, SE1 7EH, UK
| | - Henrik Hagberg
- Perinatal Center, Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30, Gothenburg, Sweden.,Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Perinatal Imaging and Health, Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, SE1 7EH, UK
| | - Carina Mallard
- Perinatal Center, Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30, Gothenburg, Sweden
| | - Jianmei W Leavenworth
- Department of Neurosurgery, The University of Alabama at Birmingham, Birmingham, AL, 35233, USA.,Department of Microbiology, The University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Changlian Zhu
- Henan Key Laboratory of Child Brain Injury, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China. .,Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 436, 405 30, Gothenburg, Sweden.
| | - Xiaoyang Wang
- Perinatal Center, Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30, Gothenburg, Sweden. .,Department of Pediatrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
63
|
Pharmacologic activation of cholinergic alpha7 nicotinic receptors mitigates depressive-like behavior in a mouse model of chronic stress. J Neuroinflammation 2017; 14:234. [PMID: 29197398 PMCID: PMC5712092 DOI: 10.1186/s12974-017-1007-2] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 11/20/2017] [Indexed: 02/07/2023] Open
Abstract
Background It has been shown that chronic stress-induced depression is associated with exaggerated inflammatory response in the brain. Alpha7 nicotinic acetylcholine receptors (α7nAChRs) regulate the cholinergic anti-inflammatory pathway, but the role of cholinergic signaling and α7nAChR in chronic stress has not yet been examined. Methods In this study, we used a well-documented model of depression in which mice were exposed to 6 h of restraint stress for 21 consecutive days. Components of cholinergic signaling and TLR4 signaling were analyzed in the hippocampus. The main targets of neuroinflammation and neuronal damage were also evaluated after a series of tests for depression-like behavior. Results Chronic restraint stress (CRS) induced alterations in components of central cholinergic signaling in hippocampus, including increases in choline acetyltransferase protein expression and decreases in nuclear STAT3 signaling. CRS also increased TLR4 signaling activity, interleukin-1β, and tumor necrosis factor-α expression, microglial activation, and neuronal morphologic changes. Cholinergic stimulation with the α7nAChR agonist DMXBA significantly alleviated CRS-induced depressive-like behavior, neuroinflammation, and neuronal damage, but these effects were abolished by the selective α7nAChR antagonist α-bungarotoxin. Furthermore, activation of α7nAChRs restored the central cholinergic signaling function, inhibited TLR4-mediated inflammatory signaling and microglial activity, and increased the number of regulatory T cells in the hippocampus. Conclusions These findings provide evidence that α7nAChR activation mitigates CRS-induced neuroinflammation and cell death, suggesting that α7nAChRs could be a new therapeutic target for the prevention and treatment of depression. Electronic supplementary material The online version of this article (10.1186/s12974-017-1007-2) contains supplementary material, which is available to authorized users.
Collapse
|
64
|
Saraykar S, Cao B, Barroso LS, Pereira KS, Bertola L, Nicolau M, Ferreira JD, Dias NS, Vieira EL, Teixeira AL, Silva APM, Diniz BS. Plasma IL-17A levels in patients with late-life depression. ACTA ACUST UNITED AC 2017; 40:212-215. [PMID: 29069253 PMCID: PMC6900762 DOI: 10.1590/1516-4446-2017-2299] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 06/05/2017] [Indexed: 02/07/2023]
Abstract
Objective: A consistent body of research has confirmed that patients with major depressive disorder (MDD) have increased concentrations of pro-inflammatory cytokines, including IL-6, TNF-α, IL-1β, the soluble IL-2 receptor, and C-reactive protein, compared to controls; however, there is limited information on IL-17A in MDD. Moreover, information about IL-17A in older populations, i.e., patients with late-life depression (LLD), is conspicuously missing from the literature. The purpose of this study was to investigate the role of IL-17A in LLD. Methods: A convenience sample of 129 individuals, 74 with LLD and 55 non-depressed controls, were enrolled in this study. The Mann-Whitney U test was used to compare plasma IL-17A levels between LLD and controls subjects, and Spearman’s rank order correlation was used to investigate correlation of these levels with clinical, neuropsychological, and cognitive assessments. Results: Plasma IL-17A levels were not statistically different between LLD patients and controls (p = 0.94). Among all subjects (LLD + control), plasma IL-17A did not correlate significantly with depressive symptoms (rho = -0.009, p = 0.92) but a significant correlation was observed with cognitive assessments (rho = 0.22, p = 0.01). Conclusion: Our findings do not support an association between plasma IL-17A levels and LLD. Nevertheless, IL-17A may be associated with cognitive impairment in LLD patients. If this finding is confirmed in future longitudinal studies, modulation of the T-helper 17 cell (Th17) immune response may be a treatment target for cognitive impairment in this population.
Collapse
Affiliation(s)
- Smita Saraykar
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Bo Cao
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Lucelia S Barroso
- Programa de Pós-Graduação em Medicina Molecular, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Kelly S Pereira
- Psychiatry Department, Instituto de Previdência dos Servidores do Estado de Minas Gerais (IPSEMG), Belo Horizonte, MG, Brazil
| | - Laiss Bertola
- Programa de Pós-Graduação em Medicina Molecular, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Mariana Nicolau
- Programa de Pós-Graduação em Medicina Molecular, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Jessica D Ferreira
- Programa de Pós-Graduação em Medicina Molecular, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Natalia S Dias
- Programa de Pós-Graduação em Medicina Molecular, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Erica L Vieira
- Divisão de Neurociências, Laboratório Interdisciplinar de Investigação Médica, Faculdade de Medicina, UFMG, Belo Horizonte, MG, Brazil
| | - Antonio L Teixeira
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA.,Divisão de Neurociências, Laboratório Interdisciplinar de Investigação Médica, Faculdade de Medicina, UFMG, Belo Horizonte, MG, Brazil
| | - Ana Paula M Silva
- Programa de Pós-Graduação em Medicina Molecular, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Breno S Diniz
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA.,Programa de Pós-Graduação em Medicina Molecular, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| |
Collapse
|
65
|
Herkenham M, Kigar SL. Contributions of the adaptive immune system to mood regulation: Mechanisms and pathways of neuroimmune interactions. Prog Neuropsychopharmacol Biol Psychiatry 2017; 79:49-57. [PMID: 27613155 PMCID: PMC5339070 DOI: 10.1016/j.pnpbp.2016.09.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 07/22/2016] [Accepted: 09/05/2016] [Indexed: 12/20/2022]
Abstract
Clinical and basic studies of functional interactions between adaptive immunity, affective states, and brain function are reviewed, and the neural, humoral, and cellular routes of bidirectional communication between the brain and the adaptive immune system are evaluated. In clinical studies of depressed populations, lymphocytes-the principal cells of the adaptive immune system-exhibit altered T cell subtype ratios and CD4+ helper T cell polarization profiles. In basic studies using psychological stress to model depression, T cell profiles are altered as well, consistent with stress effects conveyed by the hypothalamic-pituitary-adrenal axis and sympathetic nervous system. Lymphocytes in turn have effects on behavior and CNS structure and function. CD4+ T cells in particular appear to modify affective behavior and rates of hippocampal dentate gyrus neurogenesis. These observations force the question of how such actions are carried out. CNS effects may occur via cellular and molecular mechanisms whereby effector memory T cells and the cytokine profiles they produce in the blood interact with the blood-brain barrier in ways that remain to be clarified. Understanding the mechanisms by which T cells polarize and interact with the brain to alter mood states is key to advances in the field, and may permit development of therapies that target cells in the periphery, thus bypassing problems associated with bioavailability of drugs within the brain.
Collapse
Affiliation(s)
- Miles Herkenham
- Section on Functional Neuroanatomy, Intramural Research Program, National Institute of Mental Health, NIH, Bethesda, MD, USA.
| | - Stacey L Kigar
- Section on Functional Neuroanatomy, Intramural Research Program, National Institute of Mental Health, NIH, Bethesda, MD, USA
| |
Collapse
|
66
|
Park HS, Han A, Yeo HL, Park MJ, You MJ, Choi HJ, Hong CW, Lee SH, Kim SH, Kim B, Kwon MS. Chronic high dose of captopril induces depressive-like behaviors in mice: possible mechanism of regulatory T cell in depression. Oncotarget 2017; 8:72528-72543. [PMID: 29069807 PMCID: PMC5641150 DOI: 10.18632/oncotarget.19879] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 07/06/2017] [Indexed: 01/05/2023] Open
Abstract
Major depression has various types of symptoms and disease courses with inconsistent response to monoamine-related antidepressants. Thus, monoamine theory may not be the only pathophysiologic pathway relevant to depression. Recently, it has been suggested that regulatory T cell (Treg) is associated with depression. Based on our previous study that showed decreased regulatory T cell (Treg) population following chronic high-dose captopril (CHC, 40 mg/kg/day * 21 days) administration, we examined whether CHC alone can induce depressive-like behaviors in mice even without stressful stimuli. In this study, we found that CHC induced depressive-like behaviors in tail suspension test (TST) and forced swimming test (FST) without systemic illness, while it did not induce anhedonic behavior, anxiety-like behaviors, or sociality-related behavior. The depressive-like behaviors were rescued by either CHC washout or antidepressant. CHC caused reduction in foxp3 and gata3 mRNA expression in the lymph nodes with elevation in plasma IL-1β and IL-6. Interestingly, CHC increased serum angiotensin II level. In the hippocampus, CHC increased TNF-α and IL-6 mRNA expression with microglia activation while reduced glucocorticoid receptor expression. However, CHC did not affect to hippocampal kynurenine pathway, serotonin level, hypothalamic corticotropin-releasing hormone mRNA level, or serum corticosterone level. Consequently, we propose that CHC may induce a specific form of depressive-like behaviors via Treg reduction and microglial activation.
Collapse
Affiliation(s)
- Hyun-Sun Park
- Department of Pharmacology, School of Medicine, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Arum Han
- Department of Pharmacology, School of Medicine, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Hye-Lim Yeo
- Department of Pharmacology, School of Medicine, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea.,Cell Therapy Center and Department of Neurology, College of Medicine, Hanyang University, Haengdang-dong, Seoul, Republic of Korea
| | - Min-Jung Park
- Department of Pharmacology, School of Medicine, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Min-Jung You
- Department of Pharmacology, School of Medicine, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Hyun Jin Choi
- College of Pharmacy, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Chang-Won Hong
- Department of Physiology, School of Medicine, Kyungpook National University, Daegu, Gyeongbuk, Republic of Korea
| | - Sang-Hyuk Lee
- Department of Psychiatry, CHA Bundang Medical Center, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Seung Hyun Kim
- Cell Therapy Center and Department of Neurology, College of Medicine, Hanyang University, Haengdang-dong, Seoul, Republic of Korea
| | - Borah Kim
- Department of Psychiatry, CHA Bundang Medical Center, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Min-Soo Kwon
- Department of Pharmacology, School of Medicine, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea
| |
Collapse
|
67
|
Blocking of carnitine palmitoyl transferase 1 potently reduces stress-induced depression in rat highlighting a pivotal role of lipid metabolism. Sci Rep 2017; 7:2158. [PMID: 28526869 PMCID: PMC5438386 DOI: 10.1038/s41598-017-02343-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 04/10/2017] [Indexed: 11/12/2022] Open
Abstract
Major depressive disorder is a complex and common mental disease, for which the pathology has not been elucidated. The purpose of this study is to provide knowledge about the importance of mitochondrial dysfunction, dysregulated lipid metabolism and inflammation. Mitochondrial carnitine palmitoyl transferase 1a (CPT1a) is a key molecule involved in lipid metabolism and mutations in CPT1a causing reduced function is hypothesized to have a protective role in the development of depression. Moreover, CPT1a is found to be upregulated in suicide patients with history of depression. Therefore, we hypothesized that inhibition of CPT1a activity can be developed as an innovative treatment strategy for depression. Stress exposure combined with different pharmacological treatment regimens; Etomoxir, CPT1 blocker, and Escitalopram, a favoured antidepressant drug, was applied in state-of-the-art chronic mild stress model. Etomoxir treatment induced statistical significant reduction of anhedonic behavior compared to vehicle treatment (p < 0.0001) and reversed depression-like phenotype in 90% of the rats (p = 0.0007), whereas Escitalopram only proved 57% efficacy. Moreover, Etomoxir revealed downregulation of interferon-γ, interleukin-17α and tumor necrosis factor-α. This indicate that alteration in metabolism is pivotal in the pathogenesis of depression, since CPT1 blockage is highly efficient in treating anhedonia and inflammation, thereby opening up for a novel class of antidepressant medication.
Collapse
|
68
|
Therapeutic Implications of Brain-Immune Interactions: Treatment in Translation. Neuropsychopharmacology 2017; 42:334-359. [PMID: 27555382 PMCID: PMC5143492 DOI: 10.1038/npp.2016.167] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 07/22/2016] [Accepted: 08/17/2016] [Indexed: 02/06/2023]
Abstract
A wealth of data has been amassed that details a complex, yet accessible, series of pathways by which the immune system, notably inflammation, can influence the brain and behavior. These data have opened the window to a diverse array of novel targets whose potential efficacy is tied to specific neurotransmitters and neurocircuits as well as specific behaviors. What is clear is that the impact of inflammation on the brain cuts across psychiatric disorders and engages dopaminergic and glutamatergic pathways that regulate motivation and motor activity as well as the sensitivity to threat. Given the ability to identify patient populations with increased inflammation, the precision of interventions can be further tuned, in conjunction with the ability to establish target engagement in the brain through the use of multiple neuroimaging strategies. After a brief overview of the mechanisms by which inflammation affects the brain and behavior, this review examines the extant literature on the efficacy of anti-inflammatory treatments, while forging guidelines for future intelligent clinical trial design. An examination of the most promising therapeutic strategies is also provided, along with some of the most exciting clinical trials that are currently being planned or underway.
Collapse
|
69
|
Song C, Nicholson JD, Clark SM, Li X, Keegan AD, Tonelli LH. Expansion of brain T cells in homeostatic conditions in lymphopenic Rag2(-/-) mice. Brain Behav Immun 2016; 57:161-172. [PMID: 27013354 PMCID: PMC5010944 DOI: 10.1016/j.bbi.2016.03.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 03/16/2016] [Accepted: 03/19/2016] [Indexed: 10/22/2022] Open
Abstract
The concept of the brain as an immune privileged organ is rapidly evolving in light of new findings outlining the sophisticated relationship between the central nervous and the immune systems. The role of T cells in brain development and function, as well as modulation of behavior has been demonstrated by an increasing number of studies. Moreover, recent studies have redefined the existence of a brain lymphatic system and the presence of T cells in specific brain structures, such as the meninges and choroid plexus. Nevertheless, much information is needed to further the understanding of brain T cells and their relationship with the central nervous system under non-inflammatory conditions. In the present study we employed the Rag2(-/-) mouse model of lymphocyte deficiency and reconstitution by adoptive transfer to study the temporal and anatomical expansion of T cells in the brain under homeostatic conditions. Lymphopenic Rag2(-/-) mice were reconstituted with 10 million lymphoid cells and studied at one, two and four weeks after transfer. Moreover, lymphoid cells and purified CD4(+) and CD8(+) T cells from transgenic GFP expressing mice were used to define the neuroanatomical localization of transferred cells. T cell numbers were very low in the brain of reconstituted mice up to one week after transfer and significantly increased by 2weeks, reaching wild type values at 4weeks after transfer. CD4(+) T cells were the most abundant lymphocyte subtype found in the brain followed by CD8(+) T cells and lastly B cells. Furthermore, proliferation studies showed that CD4(+) T cells expand more rapidly than CD8(+) T cells. Lymphoid cells localize abundantly in meningeal structures, choroid plexus, and circumventricular organs. Lymphocytes were also found in vascular and perivascular spaces and in the brain parenchyma across several regions of the brain, in particular in structures rich in white matter content. These results provide proof of concept that the brain meningeal system, as well as vascular and perivascular spaces, are homing sites of lymphocytes and suggest the possibility of a brain specific T cell subtype.
Collapse
Affiliation(s)
- Chang Song
- Laboratory of Behavioral Neuroimmunology, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD
| | - James D. Nicholson
- Laboratory of Behavioral Neuroimmunology, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD
| | - Sarah M. Clark
- Laboratory of Behavioral Neuroimmunology, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD,Research and Development Service, Department of Veterans Affairs, VA Maryland Health Care System, Baltimore, MD
| | - Xin Li
- Laboratory of Behavioral Neuroimmunology, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD
| | - Achsah D Keegan
- Research and Development Service, Department of Veterans Affairs, VA Maryland Health Care System, Baltimore, MD,Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD
| | - Leonardo H. Tonelli
- Laboratory of Behavioral Neuroimmunology, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD,Research and Development Service, Department of Veterans Affairs, VA Maryland Health Care System, Baltimore, MD,Corresponding author: Leonardo H. Tonelli, PhD, 685 West Baltimore Street, MSTF Building Room 934 C, Baltimore, Maryland 21201, , Telephone: 410-706-2325
| |
Collapse
|
70
|
Prelog M, Hilligardt D, Schmidt CA, Przybylski GK, Leierer J, Almanzar G, El Hajj N, Lesch KP, Arolt V, Zwanzger P, Haaf T, Domschke K. Hypermethylation of FOXP3 Promoter and Premature Aging of the Immune System in Female Patients with Panic Disorder? PLoS One 2016; 11:e0157930. [PMID: 27362416 PMCID: PMC4928917 DOI: 10.1371/journal.pone.0157930] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Accepted: 06/07/2016] [Indexed: 02/02/2023] Open
Abstract
Immunological abnormalities associated with pathological conditions, such as higher infection rates, inflammatory diseases, cancer or cardiovascular events are common in patients with panic disorder. In the present study, T cell receptor excision circles (TRECs), Forkhead-Box-Protein P3 gene (FOXP3) methylation of regulatory T cells (Tregs) and relative telomere lengths (RTLs) were investigated in a total and subsamples of 131 patients with panic disorder as compared to 131 age- and sex-matched healthy controls in order to test for a potential dysfunction and premature aging of the immune system in anxiety disorders. Significantly lower TRECs (p = 0.004) as well as significant hypermethylation of the FOXP3 promoter region (p = 0.005) were observed in female (but not in male) patients with panic disorder as compared to healthy controls. No difference in relative telomere length was discerned between patients and controls, but significantly shorter telomeres in females, smokers and older persons within the patient group. The presently observed reduced TRECs in panic disorder patients and FOXP3 hypermethylation in female patients with panic disorder potentially reflect impaired thymus and immunosuppressive Treg function, which might partly account for the known increased morbidity and mortality of anxiety disorders conferred by e.g. cancer and cardiovascular disorders.
Collapse
Affiliation(s)
- Martina Prelog
- Department of Pediatrics, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Deborah Hilligardt
- Department of Pediatrics, University Hospital Wuerzburg, Wuerzburg, Germany
| | | | - Grzegorz K. Przybylski
- Clinic for Internal Medicine C, University of Greifswald, Greifswald, Germany
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | - Johannes Leierer
- Department of Internal Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - Giovanni Almanzar
- Department of Pediatrics, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Nady El Hajj
- Institute of Human Genetics, University of Wuerzburg, Wuerzburg, Germany
| | - Klaus-Peter Lesch
- Molecular Psychiatry, Department of Psychiatry, Psychosomatics and Psychotherapy, University of Wuerzburg, Wuerzburg, Germany
| | - Volker Arolt
- Department of Psychiatry and Psychotherapy, University of Muenster, Muenster, Germany
| | - Peter Zwanzger
- Department of Psychiatry and Psychotherapy, University of Muenster, Muenster, Germany
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-Universtät Munich, Munich, Germany
- kbo-Inn-Salzach-Klinikum, Wasserburg am Inn, Germany
| | - Thomas Haaf
- Institute of Human Genetics, University of Wuerzburg, Wuerzburg, Germany
| | - Katharina Domschke
- Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
71
|
Miller AH, Raison CL. The role of inflammation in depression: from evolutionary imperative to modern treatment target. Nat Rev Immunol 2016; 16:22-34. [PMID: 26711676 DOI: 10.1038/nri.2015.5] [Citation(s) in RCA: 2355] [Impact Index Per Article: 261.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Crosstalk between inflammatory pathways and neurocircuits in the brain can lead to behavioural responses, such as avoidance and alarm, that are likely to have provided early humans with an evolutionary advantage in their interactions with pathogens and predators. However, in modern times, such interactions between inflammation and the brain appear to drive the development of depression and may contribute to non-responsiveness to current antidepressant therapies. Recent data have elucidated the mechanisms by which the innate and adaptive immune systems interact with neurotransmitters and neurocircuits to influence the risk for depression. Here, we detail our current understanding of these pathways and discuss the therapeutic potential of targeting the immune system to treat depression.
Collapse
Affiliation(s)
- Andrew H Miller
- Emory University School of Medicine, Winship Cancer Institute, Atlanta, 30322 Georgia, USA
| | - Charles L Raison
- School of Human Ecology, University of Wisconsin-Madison, Madison, 53706 Wisconsin, USA
| |
Collapse
|
72
|
Immunization with a heat-killed preparation of the environmental bacterium Mycobacterium vaccae promotes stress resilience in mice. Proc Natl Acad Sci U S A 2016; 113:E3130-9. [PMID: 27185913 DOI: 10.1073/pnas.1600324113] [Citation(s) in RCA: 162] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The prevalence of inflammatory diseases is increasing in modern urban societies. Inflammation increases risk of stress-related pathology; consequently, immunoregulatory or antiinflammatory approaches may protect against negative stress-related outcomes. We show that stress disrupts the homeostatic relationship between the microbiota and the host, resulting in exaggerated inflammation. Repeated immunization with a heat-killed preparation of Mycobacterium vaccae, an immunoregulatory environmental microorganism, reduced subordinate, flight, and avoiding behavioral responses to a dominant aggressor in a murine model of chronic psychosocial stress when tested 1-2 wk following the final immunization. Furthermore, immunization with M. vaccae prevented stress-induced spontaneous colitis and, in stressed mice, induced anxiolytic or fear-reducing effects as measured on the elevated plus-maze, despite stress-induced gut microbiota changes characteristic of gut infection and colitis. Immunization with M. vaccae also prevented stress-induced aggravation of colitis in a model of inflammatory bowel disease. Depletion of regulatory T cells negated protective effects of immunization with M. vaccae on stress-induced colitis and anxiety-like or fear behaviors. These data provide a framework for developing microbiome- and immunoregulation-based strategies for prevention of stress-related pathologies.
Collapse
|
73
|
Clark SM, Soroka JA, Song C, Li X, Tonelli LH. CD4(+) T cells confer anxiolytic and antidepressant-like effects, but enhance fear memory processes in Rag2(-/-) mice. Stress 2016; 19:303-11. [PMID: 27295202 PMCID: PMC4960826 DOI: 10.1080/10253890.2016.1191466] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Accumulating evidence supports a role of T cells in behavioral stress responsiveness. Our laboratory previously reported that lymphocyte deficient Rag2(-/-) mice on a BALB/c background display resilience to maladaptive stress responses when compared with immune competent mice in the predator odor exposure (POE) paradigm, while exhibiting similar behavior in a cued fear-conditioning (FC) paradigm. In the present study, Rag2(-/-) mice on a C57BL/6 background were assessed in the same behavioral paradigms, as well as additional tests of anxiety and depressive-like behavior. Furthermore, the effects of naïve CD4(+ ) T cells were evaluated by adoptive transfer of functional cells from nonstressed, wild-type donors to Rag2(-/-) mice. Consistent with our prior results, Rag2(-/-) mice displayed an attenuated startle response after POE. Nevertheless, reconstitution of Rag2(-/-) mice with CD4(+ ) T cells did not modify startle reactivity. Additionally, in contrast with our previous findings, Rag2(-/-) mice showed attenuated fear responses in the FC paradigm compared to wild-type mice and reconstitution with CD4(+ ) T cells promoted fear learning and memory. Notably, reconstitution with CD4(+ ) T cells had anxiolytic and antidepressant-like effects in Rag2(-/-) mice that had not been previously stressed, but had no effect after POE. Taken together, our results support a role of CD4(+ ) T cells in emotionality, but also indicate that they may promote fear responses by enhancing learning and memory processes.
Collapse
Affiliation(s)
- Sarah M. Clark
- Laboratory of Behavioral Neuroimmunology, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD
- Research and Development Service, Department of Veterans Affairs, VA Maryland Health Care System, Baltimore, MD
- Corresponding author: Sarah M. Clark, PhD, 685 West Baltimore Street, MSTF 934-E, Baltimore, MD 21201, , Telephone: 410-706-2325
| | - Jennifer A Soroka
- Laboratory of Behavioral Neuroimmunology, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD
| | - Chang Song
- Laboratory of Behavioral Neuroimmunology, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD
| | - Xin Li
- Laboratory of Behavioral Neuroimmunology, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD
| | - Leonardo H Tonelli
- Laboratory of Behavioral Neuroimmunology, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD
- Research and Development Service, Department of Veterans Affairs, VA Maryland Health Care System, Baltimore, MD
| |
Collapse
|
74
|
Li J, Zhao R, Li X, Sun W, Qu M, Tang Q, Yang X, Zhang S. Shen-Qi-Jie-Yu-Fang exerts effects on a rat model of postpartum depression by regulating inflammatory cytokines and CD4(+)CD25(+) regulatory T cells. Neuropsychiatr Dis Treat 2016; 12:883-96. [PMID: 27143890 PMCID: PMC4841396 DOI: 10.2147/ndt.s98131] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Shen-Qi-Jie-Yu-Fang (SJF) is composed of eight Chinese medicinal herbs. It is widely used in traditional Chinese medicine for treating postpartum depression (PPD). Previous studies have shown that SJF treats PPD through the neuroendocrine mechanism. AIM To further investigate the effect of SJF on the immune system, including the inflammatory response system and CD4(+)CD25(+) regulatory T (Treg) cells. MATERIALS AND METHODS Sprague Dawley rats were used to create an animal model of PPD by inducing hormone-simulated pregnancy followed by hormone withdrawal. After hormone withdrawal, the PPD rats were treated with SJF or fluoxetine for 1, 2, and 4 weeks. Levels of Treg cells in peripheral blood were measured by flow cytometry analysis. Serum interleukin (IL)-1β and IL-6 were evaluated by enzyme-linked immunosorbent assay, and gene and protein expressions of IL-1RI, IL-6Rα, and gp130 in the hippocampus were observed by reverse-transcription polymerase chain reaction and Western blot. RESULTS Serum IL-1β in PPD rats increased at 2 weeks and declined from then on, while serum IL-6 increased at 1, 2, and 4 weeks. Both IL-1β and IL-6 were downregulated by SJF and fluoxetine. Changes in gene and protein expressions of IL-1RI and gp130 in PPD rats were consistent with changes in serum IL-1β, and were able to be regulated by SJF and fluoxetine. The levels of Treg cells were negatively correlated with serum IL-1β and IL-6, and were decreased in PPD rats. The levels of Treg cells were increased by SJF and fluoxetine. CONCLUSION Dysfunction of proinflammatory cytokines and Tregs in different stages of PPD was attenuated by SJF and fluoxetine through the modulation of serum concentrations of IL-1β and IL-6, expressions of IL-1RI, and gp130 in the hippocampus, and CD4(+)CD25(+) Treg cells in peripheral blood.
Collapse
Affiliation(s)
- Jingya Li
- Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, People's Republic of China
| | - Ruizhen Zhao
- Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, People's Republic of China
| | - Xiaoli Li
- Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, People's Republic of China
| | - Wenjun Sun
- Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, People's Republic of China
| | - Miao Qu
- Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, People's Republic of China
| | - Qisheng Tang
- Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, People's Republic of China
| | - Xinke Yang
- Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, People's Republic of China
| | - Shujing Zhang
- School of Basic Medical Sciences, Beijing University of Chinese Medicine, Beijing, People's Republic of China
| |
Collapse
|
75
|
Duggal NA, Upton J, Phillips AC, Lord JM. Development of depressive symptoms post hip fracture is associated with altered immunosuppressive phenotype in regulatory T and B lymphocytes. Biogerontology 2015; 17:229-39. [PMID: 26112234 PMCID: PMC4723613 DOI: 10.1007/s10522-015-9587-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 06/13/2015] [Indexed: 12/18/2022]
Abstract
Hip fracture is a common physical trauma in older adults that is also associated with a high incidence of new onset depression. The immune system declines with age and is also compromised by physical and psychological stress. This study examined whether hip fracture and depressive symptoms had additive effects upon the aged immune system that might contribute to poor health outcomes after hip fracture. We assessed the frequency of regulatory T cells, Tregs (CD4+ CD25+ Foxp3+) and IL10 production by CD4 T cells, and the frequency and IL10 production by regulatory B cells, Bregs (CD19+ CD24hi CD38hi) in 101 hip fracture patients (81 female) 6 weeks after injury and 43 healthy age-matched controls (28 female). 38 hip fracture patients (37 %) developed depressive symptoms. Hip fracture did not have an effect on circulating Tregs frequency but a significant reduction in the frequency of Bregs was observed in patients who developed depression compared with non-depressed patients (p = 0.001) or healthy controls (p < 0.001). Bregs also showed a significant decline in IL10 production in depressed hip fracture patients compared with controls (p = 0.04) and non-depressed patients (p = 0.01). In contrast, there was an increase in IL10 production by CD4 T cells in hip fracture patients with new onset depression compared to hip fracture patients without depression (p = .04) and healthy controls (p = .02). We conclude that the reduced immunity associated with new onset depression post hip fracture could include a contribution by heightened Tregs function.
Collapse
Affiliation(s)
| | - Jane Upton
- School of Sport and Exercise Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Anna C Phillips
- School of Sport and Exercise Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Janet M Lord
- School of Immunity and Infection, University of Birmingham, Birmingham, B15 2TT, UK.
- NIHR Surgical Reconstruction and Microbiology Research Centre, Birmingham, UK.
| |
Collapse
|
76
|
Evidence for a distinct neuro-immune signature in rats that develop behavioural disability after nerve injury. J Neuroinflammation 2015; 12:96. [PMID: 25986444 PMCID: PMC4506439 DOI: 10.1186/s12974-015-0318-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 05/05/2015] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Chronic neuropathic pain is a neuro-immune disorder, characterised by allodynia, hyperalgesia and spontaneous pain, as well as debilitating affective-motivational disturbances (e.g., reduced social interactions, sleep-wake cycle disruption, anhedonia, and depression). The role of the immune system in altered sensation following nerve injury is well documented. However, its role in the development of affective-motivational disturbances remains largely unknown. Here, we aimed to characterise changes in the immune response at peripheral and spinal sites in a rat model of neuropathic pain and disability. METHODS Sixty-two rats underwent sciatic nerve chronic constriction injury (CCI) and were characterised as either Pain and disability, Pain and transient disability or Pain alone on the basis of sensory threshold testing and changes in post-CCI dominance behaviour in resident-intruder interactions. Nerve ultrastructure was assessed and the number of T lymphocytes and macrophages were quantified at the site of injury on day six post-CCI. ATF3 expression was quantified in the dorsal root ganglia (DRG). Using a multiplex assay, eight cytokines were quantified in the sciatic nerve, DRG and spinal cord. RESULTS All CCI rats displayed equal levels of mechanical allodynia, structural nerve damage, and reorganisation. All CCI rats had significant infiltration of macrophages and T lymphocytes to both the injury site and the DRG. Pain and disability rats had significantly greater numbers of T lymphocytes. CCI increased IL-6 and MCP-1 in the sciatic nerve. Examination of disability subgroups revealed increases in IL-6 and MCP-1 were restricted to Pain and disability rats. Conversely, CCI led to a decrease in IL-17, which was restricted to Pain and transient disability and Pain alone rats. CCI significantly increased IL-6 and MCP-1 in the DRG, with IL-6 restricted to Pain and disability rats. CCI rats had increased IL-1β, IL-6 and MCP-1 in the spinal cord. Amongst subgroups, only Pain and disability rats had increased IL-1β. CONCLUSIONS This study has defined individual differences in the immune response at peripheral and spinal sites following CCI in rats. These changes correlated with the degree of disability. Our data suggest that individual immune signatures play a significant role in the different behavioural trajectories following nerve injury, and in some cases may lead to persistent affective-motivational disturbances.
Collapse
|
77
|
Lymphocytes from chronically stressed mice confer antidepressant-like effects to naive mice. J Neurosci 2015; 35:1530-8. [PMID: 25632130 DOI: 10.1523/jneurosci.2278-14.2015] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
We examined whether cells of the adaptive immune system retain the memory of psychosocial stress and thereby alter mood states and CNS function in the host. Lymphocytes from mice undergoing chronic social defeat stress or from unstressed control mice were isolated and adoptively transferred into naive lymphopenic Rag2(-/-) mice. Changes in affective behavior, hippocampal cell proliferation, microglial activation states, and blood cytokine levels were examined in reconstituted stress-naive mice. The mice receiving lymphocytes from defeated donors showed less anxiety, more social behavior, and increased hippocampal cell proliferation compared with those receiving no cells or cells from unstressed donors. Mice receiving stressed immune cells had reduced pro-inflammatory cytokine levels in the blood relative to the other groups, an effect opposite to the elevated donor pro-inflammatory cytokine profile. Furthermore, mice receiving stressed immune cells had microglia skewed toward an anti-inflammatory, neuroprotective M2-like phenotype, an effect opposite the stressed donors' M1-like pro-inflammatory profile. However, stress had no effect on lymphocyte surface marker profiles in both donor and recipient mice. The data suggest that chronic stress-induced changes in the adaptive immune system, contrary to conferring anxiety and depressive behavior, protect against the deleterious effects of stress. Improvement in affective behavior is potentially mediated by reduced peripheral pro-inflammatory cytokine load, protective microglial activity, and increased hippocampal cell proliferation. The data identify the peripheral adaptive immune system as putatively involved in the mechanisms underlying stress resilience and a potential basis for developing novel rapid-acting antidepressant therapies.
Collapse
|
78
|
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
|
79
|
Inflammatory cytokine-associated depression. Brain Res 2014; 1617:113-25. [PMID: 25003554 DOI: 10.1016/j.brainres.2014.06.032] [Citation(s) in RCA: 135] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 06/23/2014] [Accepted: 06/26/2014] [Indexed: 02/07/2023]
Abstract
Inflammatory cytokines can sometimes trigger depression in humans, are often associated with depression, and can elicit some behaviors in animals that are homologous to major depression. Moreover, these cytokines can affect monoaminergic and glutamatergic systems, supporting an overlapping pathoetiology with major depression. This suggests that there could be a specific major depression subtype, inflammatory cytokine-associated depression (ICAD), which may require different therapeutic approaches. However, most people do not develop depression, even when exposed to sustained elevations in inflammatory cytokines. Thus several vulnerabilities and sources of resilience to inflammation-associated depression have been identified. These range from genetic differences in neurotrophic and serotonergic systems to sleep quality and omega-3 fatty acid levels. Replicating these sources of resilience as treatments could be one approach for preventing "ICAD". This article is part of a Special Issue entitled SI: Neuroimmunology in Health And Disease.
Collapse
|
80
|
Maes M, Anderson G, Kubera M, Berk M. Targeting classical IL-6 signalling or IL-6 trans-signalling in depression? Expert Opin Ther Targets 2014; 18:495-512. [PMID: 24548241 DOI: 10.1517/14728222.2014.888417] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
INTRODUCTION Increased IL-6 and soluble IL-6 receptor (sIL-6R) levels in depressed patients was first shown over 20 years ago. The pro-inflammatory effects of IL-6 are predominantly mediated by IL-6 trans-signalling via the sIL-6R, whereas IL-6R membrane signalling has anti-inflammatory effects. AREAS COVERED We review data on IL-6 and sIL-6R in inflammation, depression, animal models of depression and the effects of different classes of antidepressants. The biological context for IL-6 trans-signalling as a pathogenic factor in depression involves its role in the acute phase response, disorders in zinc and the erythron, hypothalamic-pituitary-adrenal axis activation, induction of the tryptophan catabolite pathway, oxidative stress, bacterial translocation, transition towards sensitisation, autoimmune processes and neuroprogression and the multicausal aetiology of depression, considering that psychosocial stressors and comorbid immune-inflammatory diseases are associated with the onset of depression. EXPERT OPINION The homeostatic functions of IL-6 imply that ubiquitous IL-6 inhibitors, for example, tocilizumab, may not be the optimal treatment target in depression. A more promising target may be to increase soluble glycoprotein 130 (sgp130) inhibition of IL-6 trans-signalling, while allowing the maintenance of IL-6R membrane signalling. Future research should delineate the effects of treatments with sgp130Fc in combination with antidepressants in various animal models of chronic depression.
Collapse
Affiliation(s)
- Michael Maes
- Deakin University, Department of Psychiatry , Geelong , Australia
| | | | | | | |
Collapse
|
81
|
Wright L, Simpson W, Van Lieshout RJ, Steiner M. Depression and cardiovascular disease in women: is there a common immunological basis? A theoretical synthesis. Ther Adv Cardiovasc Dis 2014; 8:56-69. [DOI: 10.1177/1753944714521671] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Clinical studies have established an inherent comorbidity between depression and the development of cardiovascular disease (CVD). Furthermore, this comorbidity seems to be more amplified in women than in men. To further investigate this comorbidity, a thorough literature review was conducted on studies from 1992 to date. The PubMed database was accessed using the keywords: cardiovascular disease, inflammation, depression, and sex differences. Both human and animal studies were considered. This review takes the standpoint that depression and CVD are both inflammatory disorders, and that their co-occurrence may be related to how the hypothalamic–pituitary–adrenal axis, serotonergic transmission and circulation, and the renin–angiotensin–aldosterone system via angiotensin II are affected by the excess secretion of proinflammatory cytokines. More recently, preliminary research attributes this systemic inflammation to a global deficiency in CD4+CD25+FOXP3 regulatory T cells. 17-β estradiol and progesterone mediated modulation of cytokine secretion may partially explain the sex differences observed. These hormones and reproductive events associated with hormonal fluctuations are discussed in depth, including the analysis of perinatal models of depression and CVD, including preeclampsia. However, as evidenced by this review, there is a need for mechanistic research in humans to truly understand the nature and directionality of the relationship between depression and CVD.
Collapse
Affiliation(s)
- Lauren Wright
- MiNDS Neuroscience Program, McMaster University, Canada and Women’s Health Concerns Clinic, St Joseph’s Healthcare, Hamilton, ON, Canada
| | - William Simpson
- MiNDS Neuroscience Program, McMaster University, Canada and Women’s Health Concerns Clinic, St Joseph’s Healthcare, Hamilton, ON, Canada
| | - Ryan J. Van Lieshout
- MiNDS Neuroscience Program, McMaster University, Canada and Women’s Health Concerns Clinic, St Joseph’s Healthcare, and Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
| | - Meir Steiner
- MiNDS Neuroscience Program, Department of Psychiatry and Behavioural Neurosciences, McMaster University, Canada and Women’s Health Concerns Clinic, St Joseph’s Healthcare, 301 James Street South, Hamilton, ON, Canada L8P 3B6
| |
Collapse
|
82
|
Jia W, Liu R, Shi J, Wu B, Dang W, Du Y, Zhou Q, Wang J, Zhang R. Differential Regulation of MAPK Phosphorylation in the Dorsal Hippocampus in Response to Prolonged Morphine Withdrawal-Induced Depressive-Like Symptoms in Mice. PLoS One 2013; 8:e66111. [PMID: 23823128 PMCID: PMC3688859 DOI: 10.1371/journal.pone.0066111] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Accepted: 04/30/2013] [Indexed: 12/20/2022] Open
Abstract
Depression is one of the most frequent neuropsychiatric comorbidities associated with opiate addiction. Mitogen activated protein kinase (MAPK) and MAPK phosphatase (MKP) are involved in drug addiction and depression. However, the potential role of MAPK and MKP in depression caused by morphine withdrawal remains unclear. We utilized a mouse model of repeated morphine administration to examine the molecular mechanisms that contribute to prolonged withdrawal induced depressive-like behaviors. Depressive-like behaviors were significant at 1 week after withdrawal and worsened over time. Phospho-ERK (extracellular signal-regulated protein kinase) was decreased and MKP-1 was elevated in the hippocampus, and JNK (c-Jun N-terminal protein kinase), p38 (p38 protein kinase) and MKP-3 were unaffected. A pharmacological blockade of MKP-1 by intra-hippocampal sanguinarine (SA) infusion prevented the development of depressive-like behaviors and resulted in relatively normal levels of MKP-1 and phospho-ERK after withdrawal. Our findings support the association between hippocampal MAPK phosphorylation and prolonged morphine withdrawal-induced depression, and emphasize the MKP-1 as an negative regulator of the ERK phosphorylation that contributes to depression.
Collapse
Affiliation(s)
- Wei Jia
- Methadone Maintenance Treatment Clinic, Xi’an Mental Health Center, Xi’an, China
| | - Rui Liu
- Department of Geriatrics, Tangdu Hospital, Fourth Military Medical University, Xi’an, China
- * E-mail:
| | - Jianguo Shi
- Methadone Maintenance Treatment Clinic, Xi’an Mental Health Center, Xi’an, China
| | - Bin Wu
- Methadone Maintenance Treatment Clinic, Xi’an Mental Health Center, Xi’an, China
| | - Wei Dang
- Methadone Maintenance Treatment Clinic, Xi’an Mental Health Center, Xi’an, China
| | - Ying Du
- Department of Neurology, Tangdu Hospital, Fourth Military Medical University, Xi’an, China
| | - Qiong Zhou
- Department of Neurology, Tangdu Hospital, Fourth Military Medical University, Xi’an, China
| | - Jianhua Wang
- Department of Geriatrics, Tangdu Hospital, Fourth Military Medical University, Xi’an, China
| | - Rui Zhang
- Methadone Maintenance Treatment Clinic, Xi’an Mental Health Center, Xi’an, China
| |
Collapse
|
83
|
Voorhees JL, Tarr AJ, Wohleb ES, Godbout JP, Mo X, Sheridan JF, Eubank TD, Marsh CB. Prolonged restraint stress increases IL-6, reduces IL-10, and causes persistent depressive-like behavior that is reversed by recombinant IL-10. PLoS One 2013; 8:e58488. [PMID: 23520517 PMCID: PMC3592793 DOI: 10.1371/journal.pone.0058488] [Citation(s) in RCA: 172] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Accepted: 02/05/2013] [Indexed: 12/19/2022] Open
Abstract
Altered inflammatory cytokine profiles are often observed in individuals suffering from major depression. Recent clinical work reports on elevated IL-6 and decreased IL-10 in depression. Elevated IL-6 has served as a consistent biomarker of depression and IL-10 is proposed to influence depressive behavior through its ability to counterbalance pro-inflammatory cytokine expression. Clinical and animal studies suggest a role for IL-10 in modifying depressive behavior. Murine restraint stress (RST) is regularly employed in the study of behavioral and biological symptoms associated with depressive disorders. While responses to acute RST exposure have been widely characterized, few studies have examined the ongoing and longitudinal effects of extended RST and fewer still have examined the lasting impact during the post-stress period. Consistent with clinical data, we report that a protocol of prolonged murine RST produced altered cytokine profiles similar to those observed in major depressive disorder. Parallel to these changes in circulating cytokines, IL-10 mRNA expression was diminished in the cortex and hippocampus throughout the stress period and following cessation of RST. Moreover, chronic RST promoted depressive-like behavior throughout the 28-day stress period and these depressive-like complications were maintained weeks after cessation of RST. Because of the correlation between IL-10 suppression and depressive behavior and because many successful antidepressant therapies yield increases in IL-10, we examined the effects of IL-10 treatment on RST-induced behavioral changes. Behavioral deficits induced by RST were reversed by exogenous administration of recombinant IL-10. This work provides one of the first reports describing the biological and behavioral impact following prolonged RST and, taken together, this study provides details on the correlation between responses to chronic RST and those seen in depressive disorders.
Collapse
Affiliation(s)
- Jeffrey L. Voorhees
- The Dorothy M. Davis Heart and Lung Research Institute, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, Ohio, United States of America
- Department of Internal Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Andrew J. Tarr
- Division of Oral Biology, The Ohio State University, Columbus, Ohio, United States of America
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio, United States of America
| | - Eric S. Wohleb
- Division of Oral Biology, The Ohio State University, Columbus, Ohio, United States of America
- Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus, Ohio, United States of America
| | - Jonathan P. Godbout
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio, United States of America
- Department of Neuroscience, The Ohio State University, Columbus, Ohio, United States of America
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, Ohio, United States of America
| | - Xiaokui Mo
- Center for Biostatistics, The Ohio State University, Columbus, Ohio, United States of America
| | - John F. Sheridan
- Division of Oral Biology, The Ohio State University, Columbus, Ohio, United States of America
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio, United States of America
- Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus, Ohio, United States of America
| | - Timothy D. Eubank
- The Dorothy M. Davis Heart and Lung Research Institute, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, Ohio, United States of America
- Department of Internal Medicine, The Ohio State University, Columbus, Ohio, United States of America
- * E-mail: (TDE); (CBM)
| | - Clay B. Marsh
- The Dorothy M. Davis Heart and Lung Research Institute, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, Ohio, United States of America
- Department of Internal Medicine, The Ohio State University, Columbus, Ohio, United States of America
- * E-mail: (TDE); (CBM)
| |
Collapse
|
84
|
Eyre HA, Papps E, Baune BT. Treating depression and depression-like behavior with physical activity: an immune perspective. Front Psychiatry 2013; 4:3. [PMID: 23382717 PMCID: PMC3562851 DOI: 10.3389/fpsyt.2013.00003] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 01/07/2013] [Indexed: 12/18/2022] Open
Abstract
The increasing burden of major depressive disorder makes the search for an extended understanding of etiology, and for the development of additional treatments highly significant. Biological factors may be useful biomarkers for treatment with physical activity (PA), and neurobiological effects of PA may herald new therapeutic development in the future. This paper provides a thorough and up-to-date review of studies examining the neuroimmunomodulatory effects of PA on the brain in depression and depression-like behaviors. From a neuroimmune perspective, evidence suggests PA does enhance the beneficial and reduce the detrimental effects of the neuroimmune system. PA appears to increase the following factors: interleukin (IL)-10, IL-6 (acutely), macrophage migration inhibitory factor, central nervous system-specific autoreactive CD4+ T cells, M2 microglia, quiescent astrocytes, CX3CL1, and insulin-like growth factor-1. On the other hand, PA appears to reduce detrimental neuroimmune factors such as: Th1/Th2 balance, pro-inflammatory cytokines, C-reactive protein, M1 microglia, and reactive astrocytes. The effect of other mechanisms is unknown, such as: CD4+CD25+ T regulatory cells (T regs), CD200, chemokines, miRNA, M2-type blood-derived macrophages, and tumor necrosis factor (TNF)-α [via receptor 2 (R2)]. The beneficial effects of PA are likely to occur centrally and peripherally (e.g., in visceral fat reduction). The investigation of the neuroimmune effects of PA on depression and depression-like behavior is a rapidly developing and important field.
Collapse
Affiliation(s)
- Harris A Eyre
- Discipline of Psychiatry, School of Medicine, University of Adelaide Adelaide, SA, Australia ; School of Medicine and Dentistry, James Cook University Townsville, QLD, Australia
| | | | | |
Collapse
|