1
|
Leboyer M, Foiselle M, Tchitchek N, Tamouza R, Lorenzon R, Richard JR, Arrouasse R, Le Corvoisier P, Le Dudal K, Vicaut E, Ellul P, Rosenzwajg M, Klatzmann D. Low-dose interleukin-2 in patients with bipolar depression: A phase 2 randomised double-blind placebo-controlled trial. Brain Behav Immun 2025; 123:177-184. [PMID: 39242054 DOI: 10.1016/j.bbi.2024.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/28/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024] Open
Abstract
Immune abnormalities including an insufficiency of regulatory T cells (Treg) and increased blood-based inflammatory markers have been observed in bipolar disorders (BD), particularly during depression. As Tregs are pivotal to control inflammation, Treg stimulation by low-dose IL-2 (IL-2LD) could have a therapeutic impact on bipolar depression. We performed a randomized, double-blind, placebo-controlled (2 active: 1 placebo) proof-of-concept trial of add-on IL-2LD in patients with bipolar depression. Patients received a placebo or IL-2LD (1MIU) once a day for 5 days, and then once a week for 4 weeks starting on week 2. The primary objective was to demonstrate a biological Treg response to IL-2LD assessed by fold increase in Treg percentage of CD4 + cells from baseline to day 5. Secondary objectives included safety assessment and mood improvement throughout the study period. This trial is registered with ClinicalTrials.gov, number NCT04133233. Fourteen patients with bipolar depression were included, with 4 receiving placebo and 10 IL-2LD. Baseline clinical and biological characteristics were balanced between groups. The primary evaluation criterion was met, with IL-2LD expanding 1.17 [95 % CI 1.01-1.34] vs 1.01 [95 % CI 0.90-1.12] (p = 0.0421) and activating Tregs. Secondary evaluation criteria were also met with significant improvements of depressive symptoms and global functioning from day-15 onwards in the IL-2LD treated patients. The treatment was well-tolerated, with no serious adverse events related to treatment. This proof-of-concept trial shows that stimulating Tregs in patients with bipolar depression is safe and associated with clinical improvements. This supports a pathophysiological role of inflammation in BD and warrants pursuing the evaluation of IL-2LD as an adjunct treatment of major mood disorders.
Collapse
Affiliation(s)
- Marion Leboyer
- Université Paris Est Créteil (UPEC), AP-HP, Department of Psychiatry, Hôpital Henri Mondor, DMU IMPACT, FHU ADAPT, INSERM U955, IMRB, Translational NeuroPsychiatry Laboratory, 51 Avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France.
| | - Marianne Foiselle
- Université Paris Est Créteil (UPEC), AP-HP, Department of Psychiatry, Hôpital Henri Mondor, DMU IMPACT, FHU ADAPT, INSERM U955, IMRB, Translational NeuroPsychiatry Laboratory, 51 Avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France
| | - Nicolas Tchitchek
- Sorbonne Université, INSERM, UMR_S 959, Immunology-Immunopathology- Immunotherapy (i3), F-75651 Paris, France
| | - Ryad Tamouza
- Université Paris Est Créteil (UPEC), AP-HP, Department of Psychiatry, Hôpital Henri Mondor, DMU IMPACT, FHU ADAPT, INSERM U955, IMRB, Translational NeuroPsychiatry Laboratory, 51 Avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France
| | - Roberta Lorenzon
- Sorbonne Université, INSERM, UMR_S 959, Immunology-Immunopathology- Immunotherapy (i3), F-75651 Paris, France; AP-HP, Pitié-Salpêtrière Hospital, Biotherapy (CIC-BTi) and Inflammation-Immunopathology-Biotherapy Department (i2B), F-75651 Paris, France
| | - Jean-Romain Richard
- Université Paris Est Créteil (UPEC), AP-HP, Department of Psychiatry, Hôpital Henri Mondor, DMU IMPACT, FHU ADAPT, INSERM U955, IMRB, Translational NeuroPsychiatry Laboratory, 51 Avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France
| | - Raphaele Arrouasse
- Inserm, Centre d'Investigation Clinique 1430 et AP-HP, Hôpitaux Universitaires Henri Mondor, Univ Paris Est Creteil, F-94010 Créteil, France
| | - Philippe Le Corvoisier
- Inserm, Centre d'Investigation Clinique 1430 et AP-HP, Hôpitaux Universitaires Henri Mondor, Univ Paris Est Creteil, F-94010 Créteil, France
| | - Katia Le Dudal
- Inserm, Centre d'Investigation Clinique 1430 et AP-HP, Hôpitaux Universitaires Henri Mondor, Univ Paris Est Creteil, F-94010 Créteil, France
| | - Eric Vicaut
- AP-HP, Saint Louis/Lariboisière Hospitals, Unité de recherche clinique and Université Paris 7, F-75010 Paris, France
| | - Pierre Ellul
- Sorbonne Université, INSERM, UMR_S 959, Immunology-Immunopathology- Immunotherapy (i3), F-75651 Paris, France
| | - Michelle Rosenzwajg
- Sorbonne Université, INSERM, UMR_S 959, Immunology-Immunopathology- Immunotherapy (i3), F-75651 Paris, France; AP-HP, Pitié-Salpêtrière Hospital, Biotherapy (CIC-BTi) and Inflammation-Immunopathology-Biotherapy Department (i2B), F-75651 Paris, France
| | - David Klatzmann
- Sorbonne Université, INSERM, UMR_S 959, Immunology-Immunopathology- Immunotherapy (i3), F-75651 Paris, France; AP-HP, Pitié-Salpêtrière Hospital, Biotherapy (CIC-BTi) and Inflammation-Immunopathology-Biotherapy Department (i2B), F-75651 Paris, France.
| |
Collapse
|
2
|
Hofmann T, Schmucker S, Bessei W, Stefanski V. From feather pecking to immunity: Immune differences between lines selected for high and low feather pecking. Brain Behav Immun 2024; 124:S0889-1591(24)00738-4. [PMID: 39674555 DOI: 10.1016/j.bbi.2024.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 11/26/2024] [Accepted: 12/09/2024] [Indexed: 12/16/2024] Open
Abstract
Feather pecking (FP) is a serious behavioral disorder in laying hens, leading to feather damage, skin lesions, and often resulting in cannibalism. The mechanisms underlying FP are not clear yet, but recently the role of the immune system as a cause has been discussed. In humans, the interrelation between personality traits and the immune system is well-documented, with impulsivity and hyperactivity linked to distinct alterations in blood immune cell numbers and to elevated levels of pro-inflammatory cytokines. Similarly, FP in hens is associated with impulsivity and hyperactivity, suggesting a possible connection between FP and immune cell alterations. In this study numbers of leukocyte subsets in blood, spleen and cecal tonsils, along with mitogen-induced lymphocyte proliferative response and antibody concentrations across hens selectively bred for high (HFP) and low (LFP) feather pecking behavior were analyzed. Results showed that divergent selection altered FP behavior, with HFP hens showing about 10 times more pecking behavior than hens of the LFP line. HFP hens had lower numbers of T helper cells, CD4+ CD25high as well as B cells compared to LFP hens. Furthermore, HFP hens demonstrated a stronger proliferation of T cells when stimulated with ConA, while showed a weaker response in T cell-dependent B cell proliferation when stimulated with PWM, compared to LFP hens. Antibody plasma concentrations were similar between both lines. These findings highlight substantial immunological differences between HFP and LFP hens, especially in T cell immunity, and support the hypothesis that FP may be an immune-related behavioral response.
Collapse
Affiliation(s)
- Tanja Hofmann
- Behavioral Physiology of Livestock, Institute of Animal Science, University of Hohenheim, Garbenstr. 17, 70599 Stuttgart, Germany.
| | - Sonja Schmucker
- Behavioral Physiology of Livestock, Institute of Animal Science, University of Hohenheim, Garbenstr. 17, 70599 Stuttgart, Germany
| | - Werner Bessei
- Livestock Population Genomics, Institute of Animal Science, University of Hohenheim, Garbenstr. 17, 70599 Stuttgart, Germany
| | - Volker Stefanski
- Behavioral Physiology of Livestock, Institute of Animal Science, University of Hohenheim, Garbenstr. 17, 70599 Stuttgart, Germany
| |
Collapse
|
3
|
Yao H, Liu Y, Wang Y, Xue Y, Jiang S, Sun X, Ji M, Xu Z, Ding J, Hu G, Lu M. Dural Tregs driven by astrocytic IL-33 mitigate depression through the EGFR signals in mPFC neurons. Cell Death Differ 2024:10.1038/s41418-024-01421-3. [PMID: 39592709 DOI: 10.1038/s41418-024-01421-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/13/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024] Open
Abstract
The dura sinus-resident immune cells can influence the process of central neural system (CNS) diseases by communicating with central nerve cells. In clinical, Tregs are also frequently impaired in depression. However, the significance of this relationship remains unknown. In the present study, we found a significant increase in dural Treg populations in mouse models of depression, whereas depleting them by neutralizing antibodies injection could exacerbate depressive phenotypes. Through RNA sequencing, we identified that the antidepressant effects of dural Tregs are at least in part through the production of amphiregulin, increasing the expression of its receptor EGFR in medial prefrontal cortex (mPFC) pyramidal neurons. Furthermore, dural Tregs expressed high levels of ST2, and their expansion in depressed mice depended on astrocyte-derived IL33 secretion. Our study shows that dural Treg signaling can be enhanced by treatment with fluoxetine, highlighting that dural Tregs can be utilized as a potential target cell in major depressive disorder (MDD).
Collapse
Affiliation(s)
- Hang Yao
- Department of Pharmacology, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
- The Second People's Hospital of Changzhou, Changzhou Medical Center, Nanjing Medical University, Nanjing, China
| | - Yang Liu
- Department of Pharmacology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yueping Wang
- Department of Pharmacology, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - You Xue
- Department of Pharmacology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Siyuan Jiang
- Department of Pharmacology, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Xin Sun
- Department of Pharmacology, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Minjun Ji
- Department of Pathogen Biology, Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing Medical University, Nanjing, China
| | - Zhipeng Xu
- Department of Pathogen Biology, Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing Medical University, Nanjing, China
| | - Jianhua Ding
- Department of Pharmacology, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Gang Hu
- Department of Pharmacology, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China.
- Department of Pharmacology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Ming Lu
- Department of Pharmacology, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China.
- The Second People's Hospital of Changzhou, Changzhou Medical Center, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
4
|
Cepeda Y, Elizondo-Vega R, Garrido C, Tobar C, Araneda M, Oliveros P, Ordenes P, Carril C, Vidal PM, Luz-Crawford P, García-Robles MA, Oyarce K. Regulatory T cells administration reduces anxiety-like behavior in mice submitted to chronic restraint stress. Front Cell Neurosci 2024; 18:1406832. [PMID: 39206016 PMCID: PMC11349540 DOI: 10.3389/fncel.2024.1406832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 07/25/2024] [Indexed: 09/04/2024] Open
Abstract
Background Major depression disorder (MDD) and anxiety are common mental disorders that significantly affect the quality of life of those who suffer from them, altering the person's normal functioning. From the biological perspective, the most classical hypothesis explaining their occurrence relies on neurotransmission and hippocampal excitability alterations. However, around 30% of MDD patients do not respond to medication targeting these processes. Over the last decade, the involvement of inflammatory responses in depression and anxiety pathogenesis has been strongly acknowledged, opening the possibility of tackling these disorders from an immunological point of view. In this context, regulatory T cells (Treg cells), which naturally maintain immune homeostasis by suppressing inflammation could be promising candidates for their therapeutic use in mental disorders. Methods To test this hypothesis, C57BL/6 adult male mice were submitted to classical stress protocols to induce depressive and anxiety-like behavior; chronic restriction stress (CRS), and chronic unpredictable stress (CUS). Some of the stressed mice received a single adoptive transfer of Treg cells during stress protocols. Mouse behavior was analyzed through the open field (OFT) and forced swim test (FST). Blood and spleen samples were collected for T cell analysis using cell cytometry, while brains were collected to study changes in microglia by immunohistochemistry. Results Mice submitted to CRS and CUS develop anxiety and depressive-like behavior, and only CRS mice exhibit lower frequencies of circulating Treg cells. Adoptive transfer of Treg cells decreased anxiety-like behavior in the OFT only in CRS model, but not depressive behavior in FST in neither of the two models. In CRS mice, Treg cells administration lowered the number of microglia in the hippocampus, which increased due this stress paradigm, and restored its arborization. However, in CUS mice, Treg cells administration increased microglia number with no significant effect on their arborization. Conclusion Our results for effector CD4+ T cells in the spleen and microglia number and morphology in the hippocampus add new evidence in favor of the participation of inflammatory responses in the development of depressive and anxiety-like behavior and suggest that the modulation of key immune cells such as Treg cells, could have beneficial effects on these disorders.
Collapse
Affiliation(s)
- Yamila Cepeda
- Laboratorio de Neuroinmunología, Facultad de Medicina y Ciencia, Universidad San Sebastián, Sede Concepción, Concepción, Chile
- Laboratorio de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Roberto Elizondo-Vega
- Laboratorio de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Camila Garrido
- Laboratorio de Neuroinmunología, Facultad de Medicina y Ciencia, Universidad San Sebastián, Sede Concepción, Concepción, Chile
- Laboratorio de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Catalina Tobar
- Laboratorio de Neuroinmunología, Facultad de Medicina y Ciencia, Universidad San Sebastián, Sede Concepción, Concepción, Chile
| | - Matías Araneda
- Laboratorio de Neuroinmunología, Facultad de Medicina y Ciencia, Universidad San Sebastián, Sede Concepción, Concepción, Chile
| | - Patricia Oliveros
- Laboratorio de Neuroinmunología, Facultad de Medicina y Ciencia, Universidad San Sebastián, Sede Concepción, Concepción, Chile
| | - Patricio Ordenes
- Laboratorio de Neuroinmunología, Facultad de Medicina y Ciencia, Universidad San Sebastián, Sede Concepción, Concepción, Chile
| | - Claudio Carril
- Laboratorio de Neuroinmunología, Facultad de Medicina y Ciencia, Universidad San Sebastián, Sede Concepción, Concepción, Chile
| | - Pía M. Vidal
- Neuroimmunology and Regeneration of the Central Nervous System Unit, Biomedical Science Research Laboratory, Department of Basic Sciences, Faculty of Medicine, Universidad Católica de la Santísima Concepción, Concepción, Chile
| | - Patricia Luz-Crawford
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - María. A. García-Robles
- Laboratorio de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Karina Oyarce
- Laboratorio de Neuroinmunología, Facultad de Medicina y Ciencia, Universidad San Sebastián, Sede Concepción, Concepción, Chile
| |
Collapse
|
5
|
Kalisch R, Russo SJ, Müller MB. Neurobiology and systems biology of stress resilience. Physiol Rev 2024; 104:1205-1263. [PMID: 38483288 PMCID: PMC11381009 DOI: 10.1152/physrev.00042.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 03/06/2024] [Accepted: 03/12/2024] [Indexed: 05/16/2024] Open
Abstract
Stress resilience is the phenomenon that some people maintain their mental health despite exposure to adversity or show only temporary impairments followed by quick recovery. Resilience research attempts to unravel the factors and mechanisms that make resilience possible and to harness its insights for the development of preventative interventions in individuals at risk for acquiring stress-related dysfunctions. Biological resilience research has been lagging behind the psychological and social sciences but has seen a massive surge in recent years. At the same time, progress in this field has been hampered by methodological challenges related to finding suitable operationalizations and study designs, replicating findings, and modeling resilience in animals. We embed a review of behavioral, neuroimaging, neurobiological, and systems biological findings in adults in a critical methods discussion. We find preliminary evidence that hippocampus-based pattern separation and prefrontal-based cognitive control functions protect against the development of pathological fears in the aftermath of singular, event-type stressors [as found in fear-related disorders, including simpler forms of posttraumatic stress disorder (PTSD)] by facilitating the perception of safety. Reward system-based pursuit and savoring of positive reinforcers appear to protect against the development of more generalized dysfunctions of the anxious-depressive spectrum resulting from more severe or longer-lasting stressors (as in depression, generalized or comorbid anxiety, or severe PTSD). Links between preserved functioning of these neural systems under stress and neuroplasticity, immunoregulation, gut microbiome composition, and integrity of the gut barrier and the blood-brain barrier are beginning to emerge. On this basis, avenues for biological interventions are pointed out.
Collapse
Affiliation(s)
- Raffael Kalisch
- Leibniz Institute for Resilience Research (LIR), Mainz, Germany
- Neuroimaging Center (NIC), Focus Program Translational Neuroscience (FTN), Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Scott J Russo
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States
- Brain and Body Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Marianne B Müller
- Leibniz Institute for Resilience Research (LIR), Mainz, Germany
- Translational Psychiatry, Department of Psychiatry and Psychotherapy, Johannes Gutenberg University Medical Center, Mainz, Germany
| |
Collapse
|
6
|
Belbézier A, Nguyen TTT, Arnaud M, Ducotterd B, Vangout M, Deroux A, Mansard C, Sarrot-Reynauld F, Bouillet L. Treatment of non-systemic Sjögren's syndrome: Potential prevention of systematization with immunosuppressant agent/biotherapy. J Transl Autoimmun 2024; 8:100238. [PMID: 38496268 PMCID: PMC10940795 DOI: 10.1016/j.jtauto.2024.100238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 02/27/2024] [Accepted: 02/28/2024] [Indexed: 03/19/2024] Open
Abstract
Sjögren's syndrome (SS) is a systemic autoimmune pathology manifested mainly by a dry syndrome, intense asthenia and arthromyalgia. Systemic manifestations may also occur. Since 2019, immunosuppressant agents (IS) or biotherapies are recommended only for patients with systemic involvement. However, before 2019, in some cases, paucisymptomatic patients had been treated with IS/biotherapies, often off-label. Objective: We propose to evaluate the benefit and safety of using IS/biotherapy in patients with SS without systemic involvement. Methods: We retrospectively collected the clinical records of all patients with SS diagnosed according to ACR/EULAR diagnostic criteria followed up between January 1980 and October 2023 at Grenoble University Hospital (France). Results: Eighty-three patients were included: 64 with an initially non-systemic form. Of these patients with an initially non-systemic form, 24 were treated with IS/biotherapy. None of them developed secondary systematization, whereas 11 out of 40 patients in the untreated group did (p < 0.05). On the other hand, IS/biotherapy did not appear to improve dry syndrome. There were no serious adverse events. Conclusion: Early introduction of an IS/biotherapy treatment appears to provide a benefit for the patient without side effects.
Collapse
Affiliation(s)
- Aude Belbézier
- Clinique Universitaire de Médecine Interne, Department of Internal Medicine, Grenoble University Hospital, F-38000, Grenoble, France
| | - Thi Thu Thuy Nguyen
- Clinique Universitaire de Médecine Interne, Department of Internal Medicine, Grenoble University Hospital, F-38000, Grenoble, France
| | - Mélanie Arnaud
- Clinique Universitaire de Médecine Interne, Department of Internal Medicine, Grenoble University Hospital, F-38000, Grenoble, France
| | - Bruna Ducotterd
- Clinique Universitaire de Médecine Interne, Department of Internal Medicine, Grenoble University Hospital, F-38000, Grenoble, France
| | - Marie Vangout
- Clinique Universitaire de Médecine Interne, Department of Internal Medicine, Grenoble University Hospital, F-38000, Grenoble, France
| | - Alban Deroux
- Clinique Universitaire de Médecine Interne, Department of Internal Medicine, Grenoble University Hospital, F-38000, Grenoble, France
| | - Catherine Mansard
- Clinique Universitaire de Médecine Interne, Department of Internal Medicine, Grenoble University Hospital, F-38000, Grenoble, France
| | - Françoise Sarrot-Reynauld
- Clinique Universitaire de Médecine Interne, Department of Internal Medicine, Grenoble University Hospital, F-38000, Grenoble, France
| | - Laurence Bouillet
- Clinique Universitaire de Médecine Interne, Department of Internal Medicine, Grenoble University Hospital, F-38000, Grenoble, France
| |
Collapse
|
7
|
Rachayon M, Jirakran K, Sodsai P, Sughondhabirom A, Maes M. T cell activation and deficits in T regulatory cells are associated with major depressive disorder and severity of depression. Sci Rep 2024; 14:11177. [PMID: 38750122 PMCID: PMC11096341 DOI: 10.1038/s41598-024-61865-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 05/10/2024] [Indexed: 05/18/2024] Open
Abstract
Major depressive disorder (MDD) is associated with T cell activation, but no studies have examined the combined effects of T cell activation and deficits in T regulatory (Treg) cells on the severity of acute phase MDD. Using flow cytometry, we determined the percentage and median fluorescence intensity of CD69, CD71, CD40L, and HLADR-bearing CD3+, CD4+, and CD8+ cells, and cannabinoid type 1 receptor (CB1), CD152 and GARP (glycoprotein A repetitions predominant)-bearing CD25+ FoxP3 T regulatory (Treg) cells in 30 MDD patients and 20 healthy controls in unstimulated and stimulated (anti-CD3/CD28) conditions. Based on cytokine levels, we assessed M1 macrophage, T helper (Th)-1 cell, immune-inflammatory response system (IRS), T cell growth, and neurotoxicity immune profiles. We found that the immune profiles (including IRS and neurotoxicity) were significantly predicted by decreased numbers of CD152 or GARP-bearing CD25+ FoxP3 cells or CD152 and GARP expression in combination with increases in activated T cells (especially CD8+ CD40L+ percentage and expression). MDD patients showed significantly increased numbers of CD3+ CD71+, CD3+ CD40L+, CD4+ CD71+, CD4+ CD40L+, CD4+ HLADR+, and CD8+ HLADR+ T cells, increased CD3+ CD71+, CD4+ CD71+ and CD4+ HLADR+ expression, and lowered CD25+ FoxP3 expression and CD25+ FoxP+ CB1+ numbers as compared with controls. The Hamilton Depression Rating Scale score was strongly predicted (between 30 and 40% of its variance) by a lower number of CB1 or GARP-bearing Treg cells and one or more activated T cell subtypes (especially CD8+ CD40L+). In conclusion, increased T helper and cytotoxic cell activation along with decreased Treg homeostatic defenses are important parts of MDD that lead to enhanced immune responses and, as a result, neuroimmunotoxicity.
Collapse
Affiliation(s)
- Muanpetch Rachayon
- Department of Psychiatry, Faculty of Medicine, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Ketsupar Jirakran
- Department of Psychiatry, Faculty of Medicine, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Pediatrics, Faculty of Medicine, Center of Excellence for Maximizing Children's Developmental Potential, Chulalongkorn University, Bangkok, Thailand
| | - Pimpayao Sodsai
- Department of Microbiology, Faculty of Medicine, Center of Excellence in Immunology and Immune-Mediated Diseases, King Chulalongkorn Memorial Hospital, Chulalongkorn University, Bangkok, Thailand
| | - Atapol Sughondhabirom
- Department of Psychiatry, Faculty of Medicine, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Michael Maes
- Department of Psychiatry, Faculty of Medicine, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Chulalongkorn University, Bangkok, 10330, Thailand.
- Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
- Key Laboratory of Psychosomatic Medicine, Chinese Academy of Medical Sciences, Chengdu, 610072, China.
- Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Korea.
- Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria.
- Research Institute, Medical University Plovdiv, Plovdiv, Bulgaria.
| |
Collapse
|
8
|
Poletti S, Zanardi R, Mandelli A, Aggio V, Finardi A, Lorenzi C, Borsellino G, Carminati M, Manfredi E, Tomasi E, Spadini S, Colombo C, Drexhage HA, Furlan R, Benedetti F. Low-dose interleukin 2 antidepressant potentiation in unipolar and bipolar depression: Safety, efficacy, and immunological biomarkers. Brain Behav Immun 2024; 118:52-68. [PMID: 38367846 DOI: 10.1016/j.bbi.2024.02.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 02/01/2024] [Accepted: 02/08/2024] [Indexed: 02/19/2024] Open
Abstract
Immune-inflammatory mechanisms are promising targets for antidepressant pharmacology. Immune cell abnormalities have been reported in mood disorders showing a partial T cell defect. Following this line of reasoning we defined an antidepressant potentiation treatment with add-on low-dose interleukin 2 (IL-2). IL-2 is a T-cell growth factor which has proven anti-inflammatory efficacy in autoimmune conditions, increasing thymic production of naïve CD4 + T cells, and possibly correcting the partial T cell defect observed in mood disorders. We performed a single-center, randomised, double-blind, placebo-controlled phase II trial evaluating the safety, clinical efficacy and biological responses of low-dose IL-2 in depressed patients with major depressive (MDD) or bipolar disorder (BD). 36 consecutively recruited inpatients at the Mood Disorder Unit were randomised in a 2:1 ratio to receive either aldesleukin (12 MDD and 12 BD) or placebo (6 MDD and 6 BD). Active treatment significantly potentiated antidepressant response to ongoing SSRI/SNRI treatment in both diagnostic groups, and expanded the population of T regulatory, T helper 2, and percentage of Naive CD4+/CD8 + immune cells. Changes in cell frequences were rapidly induced in the first five days of treatment, and predicted the later improvement of depression severity. No serious adverse effect was observed. This is the first randomised control trial (RCT) evidence supporting the hypothesis that treatment to strengthen the T cell system could be a successful way to correct the immuno-inflammatory abnormalities associated with mood disorders, and potentiate antidepressant response.
Collapse
Affiliation(s)
- Sara Poletti
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy; Vita-Salute San Raffaele University, Milano, Italy.
| | - Raffaella Zanardi
- Vita-Salute San Raffaele University, Milano, Italy; Mood Disorder Unit, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Alessandra Mandelli
- Clinical Neuroimmunology, Institute of Experimental Neurology, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Veronica Aggio
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy; Vita-Salute San Raffaele University, Milano, Italy
| | - Annamaria Finardi
- Clinical Neuroimmunology, Institute of Experimental Neurology, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Cristina Lorenzi
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | | | - Matteo Carminati
- Vita-Salute San Raffaele University, Milano, Italy; Mood Disorder Unit, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Elena Manfredi
- Vita-Salute San Raffaele University, Milano, Italy; Mood Disorder Unit, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Enrico Tomasi
- Hospital Pharmacy, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Sara Spadini
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Cristina Colombo
- Vita-Salute San Raffaele University, Milano, Italy; Mood Disorder Unit, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Hemmo A Drexhage
- Coordinator EU consortium MoodStratification, Department of Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Roberto Furlan
- Vita-Salute San Raffaele University, Milano, Italy; Clinical Neuroimmunology, Institute of Experimental Neurology, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Francesco Benedetti
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy; Vita-Salute San Raffaele University, Milano, Italy
| |
Collapse
|
9
|
Bravi B, Melloni EMT, Paolini M, Palladini M, Calesella F, Servidio L, Agnoletto E, Poletti S, Lorenzi C, Colombo C, Benedetti F. Choroid plexus volume is increased in mood disorders and associates with circulating inflammatory cytokines. Brain Behav Immun 2024; 116:52-61. [PMID: 38030049 DOI: 10.1016/j.bbi.2023.11.036] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 11/17/2023] [Accepted: 11/26/2023] [Indexed: 12/01/2023] Open
Abstract
Depressed patients exhibit altered levels of immune-inflammatory markers both in the peripheral blood and in the cerebrospinal fluid (CSF) and inflammatory processes have been widely implicated in the pathophysiology of mood disorders. The Choroid Plexus (ChP), located at the base of each of the four brain ventricles, regulates the exchange of substances between the blood and CSF and several evidence supported a key role for ChP as a neuro-immunological interface between the brain and circulating immune cells. Given the role of ChP as a regulatory gate between periphery, CSF spaces and the brain, we compared ChP volumes in patients with bipolar disorder (BP) or major depressive disorder (MDD) and healthy controls, exploring their association with history of illness and levels of circulating cytokines. Plasma levels of inflammatory markers and MRI scans were acquired for 73 MDD, 79 BD and 72 age- and sex-matched healthy controls (HC). Patients with either BD or MDD had higher ChP volumes than HC. With increasing age, the bilateral ChP volume was larger in patients, an effect driven by the duration of illness; while only minor effects were observed in HC. Right ChP volumes were proportional to higher levels of circulating cytokines in the clinical groups, including IFN-γ, IL-13 and IL-17. Specific effects in the two diagnostic groups were observed when considering the left ChP, with positive association with IL-1ra, IL-13, IL-17, and CCL3 in BD, and negative associations with IL-2, IL-4, IL-1ra, and IFN-γ in MDD. These results suggest that ChP could represent a reliable and easy-to-assess biomarker to evaluate the brain effects of inflammatory status in mood disorders, contributing to personalized diagnosis and tailored treatment strategies.
Collapse
Affiliation(s)
- Beatrice Bravi
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute San Raffaele Hospital, Milan, Italy; PhD Program in Cognitive Neuroscience, University Vita-Salute San Raffaele, Milan, Italy.
| | - Elisa Maria Teresa Melloni
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute San Raffaele Hospital, Milan, Italy; University Vita-Salute San Raffaele, Milan, Italy
| | - Marco Paolini
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute San Raffaele Hospital, Milan, Italy; PhD Program in Molecular Medicine, University Vita-Salute San Raffaele, Milan, Italy
| | - Mariagrazia Palladini
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute San Raffaele Hospital, Milan, Italy; PhD Program in Cognitive Neuroscience, University Vita-Salute San Raffaele, Milan, Italy
| | - Federico Calesella
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute San Raffaele Hospital, Milan, Italy; PhD Program in Cognitive Neuroscience, University Vita-Salute San Raffaele, Milan, Italy
| | - Laura Servidio
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute San Raffaele Hospital, Milan, Italy
| | - Elena Agnoletto
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute San Raffaele Hospital, Milan, Italy
| | - Sara Poletti
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute San Raffaele Hospital, Milan, Italy; University Vita-Salute San Raffaele, Milan, Italy
| | - Cristina Lorenzi
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute San Raffaele Hospital, Milan, Italy
| | - Cristina Colombo
- University Vita-Salute San Raffaele, Milan, Italy; Mood Disorders Unit, IRCCS Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Francesco Benedetti
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute San Raffaele Hospital, Milan, Italy; University Vita-Salute San Raffaele, Milan, Italy
| |
Collapse
|
10
|
Mokhtari T, Uludag K. Role of NLRP3 Inflammasome in Post-Spinal-Cord-Injury Anxiety and Depression: Molecular Mechanisms and Therapeutic Implications. ACS Chem Neurosci 2024; 15:56-70. [PMID: 38109051 DOI: 10.1021/acschemneuro.3c00596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2023] Open
Abstract
The majority of research on the long-term effects of spinal cord injury (SCI) has primarily focused on neuropathic pain (NP), psychological issues, and sensorimotor impairments. Among SCI patients, mood disorders, such as anxiety and depression, have been extensively studied. It has been found that chronic stress and NP have negative consequences and reduce the quality of life for individuals living with SCI. Our review examined both human and experimental evidence to explore the connection between mood changes following SCI and inflammatory pathways, with a specific focus on NLRP3 inflammasome signaling. We observed increased proinflammatory factors in the blood, as well as in the brain and spinal cord tissues of SCI models. The NLRP3 inflammasome plays a crucial role in various diseases by controlling the release of proinflammatory molecules like interleukin 1β (IL-1β) and IL-18. Dysregulation of the NLRP3 inflammasome in key brain regions associated with pain processing, such as the prefrontal cortex and hippocampus, contributes to the development of mood disorders following SCI. In this review, we summarized recent research on the expression and regulation of components related to NLRP3 inflammasome signaling in mood disorders following SCI. Finally, we discussed potential therapeutic approaches that target the NLRP3 inflammasome and regulate proinflammatory cytokines as a way to treat mood disorders following SCI.
Collapse
Affiliation(s)
- Tahmineh Mokhtari
- Hubei Key Laboratory of Embryonic Stem Cell Research, Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, Hubei, People's Republic of China
- Department of Histology and Embryology, Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, Hubei, People's Republic of China
| | - Kadir Uludag
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, People's Republic of China
| |
Collapse
|
11
|
Desmond LW, Holbrook EM, Wright CTO, Zambrano CA, Stamper CE, Bohr AD, Frank MG, Podell BK, Moreno JA, MacDonald AS, Reber SO, Hernández-Pando R, Lowry CA. Effects of Mycobacterium vaccae NCTC 11659 and Lipopolysaccharide Challenge on Polarization of Murine BV-2 Microglial Cells. Int J Mol Sci 2023; 25:474. [PMID: 38203645 PMCID: PMC10779110 DOI: 10.3390/ijms25010474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/15/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
Previous studies have shown that the in vivo administration of soil-derived bacteria with anti-inflammatory and immunoregulatory properties, such as Mycobacterium vaccae NCTC 11659, can prevent a stress-induced shift toward an inflammatory M1 microglial immunophenotype and microglial priming in the central nervous system (CNS). It remains unclear whether M. vaccae NCTC 11659 can act directly on microglia to mediate these effects. This study was designed to determine the effects of M. vaccae NCTC 11659 on the polarization of naïve BV-2 cells, a murine microglial cell line, and BV-2 cells subsequently challenged with lipopolysaccharide (LPS). Briefly, murine BV-2 cells were exposed to 100 µg/mL whole-cell, heat-killed M. vaccae NCTC 11659 or sterile borate-buffered saline (BBS) vehicle, followed, 24 h later, by exposure to 0.250 µg/mL LPS (Escherichia coli 0111: B4; n = 3) in cell culture media vehicle (CMV) or a CMV control condition. Twenty-four hours after the LPS or CMV challenge, cells were harvested to isolate total RNA. An analysis using the NanoString platform revealed that, by itself, M. vaccae NCTC 11659 had an "adjuvant-like" effect, while exposure to LPS increased the expression of mRNAs encoding proinflammatory cytokines, chemokine ligands, the C3 component of complement, and components of inflammasome signaling such as Nlrp3. Among LPS-challenged cells, M. vaccae NCTC 11659 had limited effects on differential gene expression using a threshold of 1.5-fold change. A subset of genes was assessed using real-time reverse transcription polymerase chain reaction (real-time RT-PCR), including Arg1, Ccl2, Il1b, Il6, Nlrp3, and Tnf. Based on the analysis using real-time RT-PCR, M. vaccae NCTC 11659 by itself again induced "adjuvant-like" effects, increasing the expression of Il1b, Il6, and Tnf while decreasing the expression of Arg1. LPS by itself increased the expression of Ccl2, Il1b, Il6, Nlrp3, and Tnf while decreasing the expression of Arg1. Among LPS-challenged cells, M. vaccae NCTC 11659 enhanced LPS-induced increases in the expression of Nlrp3 and Tnf, consistent with microglial priming. In contrast, among LPS-challenged cells, although M. vaccae NCTC 11659 did not fully prevent the effects of LPS relative to vehicle-treated control conditions, it increased Arg1 mRNA expression, suggesting that M. vaccae NCTC 11659 induces an atypical microglial phenotype. Thus, M. vaccae NCTC 11659 acutely (within 48 h) induced immune-activating and microglial-priming effects when applied directly to murine BV-2 microglial cells, in contrast to its long-term anti-inflammatory and immunoregulatory effects observed on the CNS when whole-cell, heat-killed preparations of M. vaccae NCTC 11659 were given peripherally in vivo.
Collapse
Affiliation(s)
- Luke W. Desmond
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; (L.W.D.); (E.M.H.); (C.T.O.W.); (C.A.Z.); (C.E.S.); (A.D.B.); (M.G.F.)
| | - Evan M. Holbrook
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; (L.W.D.); (E.M.H.); (C.T.O.W.); (C.A.Z.); (C.E.S.); (A.D.B.); (M.G.F.)
| | - Caelan T. O. Wright
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; (L.W.D.); (E.M.H.); (C.T.O.W.); (C.A.Z.); (C.E.S.); (A.D.B.); (M.G.F.)
| | - Cristian A. Zambrano
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; (L.W.D.); (E.M.H.); (C.T.O.W.); (C.A.Z.); (C.E.S.); (A.D.B.); (M.G.F.)
| | - Christopher E. Stamper
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; (L.W.D.); (E.M.H.); (C.T.O.W.); (C.A.Z.); (C.E.S.); (A.D.B.); (M.G.F.)
| | - Adam D. Bohr
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; (L.W.D.); (E.M.H.); (C.T.O.W.); (C.A.Z.); (C.E.S.); (A.D.B.); (M.G.F.)
| | - Matthew G. Frank
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; (L.W.D.); (E.M.H.); (C.T.O.W.); (C.A.Z.); (C.E.S.); (A.D.B.); (M.G.F.)
- Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Brendan K. Podell
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO 80523, USA;
| | - Julie A. Moreno
- Prion Research Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA;
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
- Center for Healthy Aging, Colorado State University, Fort Collins, CO 80523, USA
| | - Andrew S. MacDonald
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester M13 9NT, UK;
| | - Stefan O. Reber
- Laboratory for Molecular Psychosomatics, Department of Psychosomatic Medicine and Psychotherapy, Ulm University Medical Center, 89081 Ulm, Germany;
| | - Rogelio Hernández-Pando
- Sección de Patología Experimental, Departamento de Patología, Instituto Nacional De Ciencias Médicas Y Nutrición Salvador Zubirán, Ciudad de México 14080, Mexico;
| | - Christopher A. Lowry
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; (L.W.D.); (E.M.H.); (C.T.O.W.); (C.A.Z.); (C.E.S.); (A.D.B.); (M.G.F.)
- Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA
- Center for Microbial Exploration, University of Colorado Boulder, Boulder, CO 80309, USA
- Department of Physical Medicine and Rehabilitation and Center for Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
12
|
Maldonado-García JL, Pérez-Sánchez G, Becerril-Villanueva E, Alvarez-Herrera S, Pavón L, Sánchez-Torres L, Gutiérrez-Ospina G, Girón-Pérez MI, Damian-Morales G, Maldonado-Tapia JO, López-Santiago R, Moreno-Lafont MC. Imipramine Administration in Brucella abortus 2308-Infected Mice Restores Hippocampal Serotonin Levels, Muscle Strength, and Mood, and Decreases Spleen CFU Count. Pharmaceuticals (Basel) 2023; 16:1525. [PMID: 38004391 PMCID: PMC10674296 DOI: 10.3390/ph16111525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/25/2023] [Accepted: 09/28/2023] [Indexed: 11/26/2023] Open
Abstract
Brucellosis infection causes non-specific symptoms such as fever, chills, sweating, headaches, myalgia, arthralgia, anorexia, fatigue, and mood disorders. In mouse models, it has been associated with increased levels of IL-6, TNF-α, and IFN-γ, a decrease in serotonin and dopamine levels within the hippocampus, induced loss of muscle strength and equilibrium, and increased anxiety and hopelessness. Imipramine (ImiP), a tricyclic antidepressant, is used to alleviate neuropathic pain. This study evaluated the effects of ImiP on Balb/c mice infected with Brucella abortus 2308 (Ba) at 14- and 28-days post-infection. Serum levels of six cytokines (IFN-γ, IL-6, TNF-α, IL-12, MCP-1. and IL-10) were assessed by FACS, while the number of bacteria in the spleen was measured via CFU. Serotonin levels in the hippocampus were analyzed via HPLC, and behavioral tests were conducted to assess strength, equilibrium, and mood. Our results showed that mice infected with Brucella abortus 2308 and treated with ImiP for six days (Im6Ba14) had significantly different outcomes compared to infected mice (Ba14) at day 14 post-infection. The mood was enhanced in the forced swimming test (FST) (p < 0.01), tail suspension test (TST) (p < 0.0001), and open-field test (p < 0.0001). Additionally, there was an increase in serotonin levels in the hippocampus (p < 0.001). Furthermore, there was an improvement in equilibrium (p < 0.0001) and muscle strength (p < 0.01). Lastly, there was a decrease in IL-6 levels (p < 0.05) and CFU count in the spleen (p < 0.0001). At 28 days, infected mice that received ImiP for 20 days (Im20Ba28) showed preservation of positive effects compared to infected mice (Ba28). These effects include the following: (1) improved FST (p < 0.0001) and TST (p < 0.0001); (2) better equilibrium (p < 0.0001) and muscle strength (p < 0.0001); (3) decreased IL-6 levels (p < 0.05); and (4) reduced CFU count in the spleen (p < 0.0001). These findings suggest the potential for ImiP to be used as an adjuvant treatment for the symptoms of brucellosis, which requires future studies.
Collapse
Affiliation(s)
- José Luis Maldonado-García
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias del Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico
- Laboratorio de Inmunología Celular, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 07738, Mexico
| | - Gilberto Pérez-Sánchez
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias del Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico
| | - Enrique Becerril-Villanueva
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias del Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico
| | - Samantha Alvarez-Herrera
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias del Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico
| | - Lenin Pavón
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias del Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico
| | - Luvia Sánchez-Torres
- Laboratorio de Inmunología de los Microorganismos, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 07738, Mexico
| | - Gabriel Gutiérrez-Ospina
- Laboratorio de Biología de Sistemas, Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas y Coordinación de Psicobiología y Neurociencias, Facultad de Psicología, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | | | - Gabriela Damian-Morales
- Laboratorio de Inmunología Celular, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 07738, Mexico
| | - Jesús Octavio Maldonado-Tapia
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias del Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico
- Laboratorio de Inmunología Celular, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 07738, Mexico
| | - Rubén López-Santiago
- Laboratorio de Inmunología Celular, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 07738, Mexico
| | - Martha C Moreno-Lafont
- Laboratorio de Inmunología Celular, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 07738, Mexico
| |
Collapse
|
13
|
Luarte A, Nardocci G, Chakraborty A, Batiz LF, Pino-Lagos K, Wyneken Ú. Astrocyte-derived extracellular vesicles in stress-associated mood disorders. Does the immune system get astrocytic? Pharmacol Res 2023; 194:106833. [PMID: 37348692 DOI: 10.1016/j.phrs.2023.106833] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 06/15/2023] [Accepted: 06/19/2023] [Indexed: 06/24/2023]
Abstract
Life stressors can wreak havoc on our health, contributing to mood disorders like major depressive disorder (MDD), a widespread and debilitating condition. Unfortunately, current treatments and diagnostic strategies fall short of addressing these disorders, highlighting the need for new approaches. In this regard, the relationship between MDD, brain inflammation (neuroinflammation), and systemic inflammation in the body may offer novel insights. Recent research has uncovered the crucial role of astrocytes in coordinating the inflammatory response through the release of extracellular vesicles (ADEVs) during different neuroinflammatory conditions. While the contribution of ADEVs to stress and MDD remains largely unexplored, their potential to modulate immune cells and contribute to MDD pathogenesis is significant. In this article, we delve into the immunomodulatory role of ADEVs, their potential impact on peripheral immune cells, and how their microRNA (miRNA) landscape may hold the key to controlling immune cell activity. Together, these mechanisms may constitute an opportunity to develop novel therapeutic pharmacological approaches to tackle mood disorders.
Collapse
Affiliation(s)
- Alejandro Luarte
- Faculty of Medicine, Universidad de los Andes, Santiago 7620001, Chile; Program in Neuroscience, Center for Biomedical Research and Innovation (CiiB), Universidad de los Andes, Santiago 7620001, Chile.
| | - Gino Nardocci
- Faculty of Medicine, Universidad de los Andes, Santiago 7620001, Chile; Molecular Biology and Bioinformatics Lab, Program in Molecular Biology and Bioinformatics, Center for Biomedical Research and Innovation (CiiB), Universidad de los Andes, Santiago 7620001, Chile; IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago 7620001, Chile
| | - Ankush Chakraborty
- Program in Neuroscience, Center for Biomedical Research and Innovation (CiiB), Universidad de los Andes, Santiago 7620001, Chile
| | - Luis Federico Batiz
- Faculty of Medicine, Universidad de los Andes, Santiago 7620001, Chile; Program in Neuroscience, Center for Biomedical Research and Innovation (CiiB), Universidad de los Andes, Santiago 7620001, Chile; IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago 7620001, Chile
| | - Karina Pino-Lagos
- Faculty of Medicine, Universidad de los Andes, Santiago 7620001, Chile; Program in Immunology, Center for Biomedical Research and Innovation (CiiB), Universidad de los Andes, Santiago 7620001, Chile
| | - Úrsula Wyneken
- Faculty of Medicine, Universidad de los Andes, Santiago 7620001, Chile; Program in Neuroscience, Center for Biomedical Research and Innovation (CiiB), Universidad de los Andes, Santiago 7620001, Chile; IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago 7620001, Chile.
| |
Collapse
|
14
|
Holbrook EM, Zambrano CA, Wright CTO, Dubé EM, Stewart JR, Sanders WJ, Frank MG, MacDonald AS, Reber SO, Lowry CA. Mycobacterium vaccae NCTC 11659, a Soil-Derived Bacterium with Stress Resilience Properties, Modulates the Proinflammatory Effects of LPS in Macrophages. Int J Mol Sci 2023; 24:ijms24065176. [PMID: 36982250 PMCID: PMC10049321 DOI: 10.3390/ijms24065176] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/15/2023] [Accepted: 02/20/2023] [Indexed: 03/10/2023] Open
Abstract
Inflammatory conditions, including allergic asthma and conditions in which chronic low-grade inflammation is a risk factor, such as stress-related psychiatric disorders, are prevalent and are a significant cause of disability worldwide. Novel approaches for the prevention and treatment of these disorders are needed. One approach is the use of immunoregulatory microorganisms, such as Mycobacterium vaccae NCTC 11659, which have anti-inflammatory, immunoregulatory, and stress-resilience properties. However, little is known about how M. vaccae NCTC 11659 affects specific immune cell targets, including monocytes, which can traffic to peripheral organs and the central nervous system and differentiate into monocyte-derived macrophages that, in turn, can drive inflammation and neuroinflammation. In this study, we investigated the effects of M. vaccae NCTC 11659 and subsequent lipopolysaccharide (LPS) challenge on gene expression in human monocyte-derived macrophages. THP-1 monocytes were differentiated into macrophages, exposed to M. vaccae NCTC 11659 (0, 10, 30, 100, 300 µg/mL), then, 24 h later, challenged with LPS (0, 0.5, 2.5, 250 ng/mL), and assessed for gene expression 24 h following challenge with LPS. Exposure to M. vaccae NCTC 11659 prior to challenge with higher concentrations of LPS (250 ng/mL) polarized human monocyte-derived macrophages with decreased IL12A, IL12B, and IL23A expression relative to IL10 and TGFB1 mRNA expression. These data identify human monocyte-derived macrophages as a direct target of M. vaccae NCTC 11659 and support the development of M. vaccae NCTC 11659 as a potential intervention to prevent stress-induced inflammation and neuroinflammation implicated in the etiology and pathophysiology of inflammatory conditions and stress-related psychiatric disorders.
Collapse
Affiliation(s)
- Evan M. Holbrook
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Cristian A. Zambrano
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Caelan T. O. Wright
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Elizabeth M. Dubé
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Jessica R. Stewart
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA
| | - William J. Sanders
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Matthew G. Frank
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Andrew S. MacDonald
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester M13 9NT, UK
| | - Stefan O. Reber
- Laboratory for Molecular Psychosomatics, Department of Psychosomatic Medicine and Psychotherapy, Ulm University Medical Center, 89081 Ulm, Germany
| | - Christopher A. Lowry
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA
- Center for Neuroscience and Center for Microbial Exploration, University of Colorado, Boulder, CO 80309, USA
- Department of Physical Medicine and Rehabilitation and Center for Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), The Rocky Mountain Regional Veterans Affairs Medical Center (RMRVAMC), Aurora, CO 80045, USA
- Military and Veteran Microbiome Consortium for Research and Education (MVM-CoRE), Aurora, CO 80045, USA
- Senior Fellow, inVIVO Planetary Health, of the Worldwide Universities Network (WUN), West New York, NJ 07093, USA
- Correspondence: ; Tel.: +1-303-492-6029; Fax: +1-303-492-0811
| |
Collapse
|
15
|
Qin Z, Shi DD, Li W, Cheng D, Zhang YD, Zhang S, Tsoi B, Zhao J, Wang Z, Zhang ZJ. Berberine ameliorates depression-like behaviors in mice via inhibiting NLRP3 inflammasome-mediated neuroinflammation and preventing neuroplasticity disruption. J Neuroinflammation 2023; 20:54. [PMID: 36859349 PMCID: PMC9976521 DOI: 10.1186/s12974-023-02744-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 02/21/2023] [Indexed: 03/03/2023] Open
Abstract
OBJECTIVES Neuroinflammation has been suggested that affects the processing of depression. There is renewed interest in berberine owing to its anti-inflammatory effects. Herein, we investigated whether berberine attenuate depressive-like behaviors via inhibiting NLRP3 inflammasome activation in mice model of depression. METHODS Adult male C57BL/6N mice were administrated corticosterone (CORT, 20 mg/kg/day) for 35 days. Two doses (100 mg/kg/day and 200 mg/kg/day) of berberine were orally administrated from day 7 until day 35. Behavioral tests were performed to measure the depression-like behaviors alterations. Differentially expressed gene analysis was performed for RNA-sequencing data in the prefrontal cortex. NLRP3 inflammasome was measured by quantitative reverse transcription polymerase chain reaction, western blotting, and immunofluorescence labeling. The neuroplasticity and synaptic function were measured by immunofluorescence labeling, Golgi-Cox staining, transmission electron microscope, and whole-cell patch-clamp recordings. RESULTS The results of behavioral tests demonstrated that berberine attenuated the depression-like behaviors induced by CORT. RNA-sequencing identified that NLRP3 was markedly upregulated after long-term CORT exposure. Berberine reversed the concentrations of peripheral and brain cytokines, NLRP3 inflammasome elicited by CORT in the prefrontal cortex and hippocampus were decreased by berberine. In addition, the lower frequency of neuronal excitation as well as the dendritic spine reduction were reversed by berberine treatment. Together, berberine increases hippocampal adult neurogenesis and synaptic plasticity induced by CORT. CONCLUSION The anti-depressants effects of berberine were accompanied by reduced the neuroinflammatory response via inhibiting the activation of NLRP3 inflammasome and rescued the neuronal deterioration via suppression of impairments in synaptic plasticity and neurogenesis.
Collapse
Affiliation(s)
- Zongshi Qin
- grid.11135.370000 0001 2256 9319Peking University Clinical Research Institute, Peking University, Beijing, China ,grid.194645.b0000000121742757School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Dong-Dong Shi
- grid.16821.3c0000 0004 0368 8293Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenqi Li
- grid.194645.b0000000121742757School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Dan Cheng
- grid.194645.b0000000121742757School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Ying-Dan Zhang
- grid.16821.3c0000 0004 0368 8293Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sen Zhang
- grid.16821.3c0000 0004 0368 8293Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bun Tsoi
- grid.16890.360000 0004 1764 6123Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Jia Zhao
- grid.194645.b0000000121742757School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Zhen Wang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Zhang-Jin Zhang
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
16
|
Rodriguez-Gonzalez A, Hernández R, Cruz-Castellanos P, Fernández-Montes A, Castillo-Trujillo O, Muñoz MM, Cano-Cano JM, Corral MJ, Esteban E, Jiménez-Fonseca P, Calderon C. Using the emotional functioning in clinical practice to detect psychological distress in patients with advanced thoracic and colorectal cancer. Health Qual Life Outcomes 2023; 21:15. [PMID: 36800957 PMCID: PMC9936733 DOI: 10.1186/s12955-023-02099-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 02/13/2023] [Indexed: 02/19/2023] Open
Abstract
PURPOSE Patients with advanced cancer suffer significant decline of their psychological state. A rapid and reliable evaluation of this state is essential to detect and treat it and improve quality of life. The aim was to probe the usefulness of the emotional function (EF) subscale of the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire C30 (EF-EORTC-QLQ-C30) to assess psychological distress in cancer patients. METHODS This is a multicenter, prospective, observational study involving 15 Spanish hospitals. Patients diagnosed with unresectable advanced thoracic or colorectal cancer were included. Participants completed the Brief Symptom Inventory 18 (BSI-18), the current the gold standard, and the EF-EORTC-QLQ-C30 to assess their psychological distress prior to initiating systemic antineoplastic treatment. Accuracy, sensitivity, positive predictive value (PPV), specificity, and negative predictive value (NPV) were calculated. RESULTS The sample comprised 639 patients: 283 with advanced thoracic cancer and 356 with advanced colorectal cancer. According to the BSI scale, 74% and 66% displayed psychological distress with an EF-EORTC-QLQ-C30 accuracy of 79% and 76% in detecting psychological distress in individuals with advanced thoracic and colorectal cancer, respectively. Sensitivity was 79 and 75% and specificity was 79 and 77% with a PPV of 92 and 86% and a NPV of 56 and 61% (scale cut-off point, 75) for patients with advanced thoracic and colorectal cancer, respectively. The mean AUC for thoracic cancer was 0.84 and, for colorectal cancer, it was 0.85. CONCLUSION This study reveals that the EF-EORTC-QLQ-C30 subscale is a simple and effective tool for detecting psychological distress in people with advanced cancer.
Collapse
Affiliation(s)
- Adán Rodriguez-Gonzalez
- grid.411052.30000 0001 2176 9028Department of Medical Oncology, Hospital Universitario Central de Asturias, ISPA, Oviedo, Spain
| | - Raquel Hernández
- grid.411220.40000 0000 9826 9219Department of Medical Oncology, Hospital Universitario de Canarias, Tenerife, Spain
| | - Patricia Cruz-Castellanos
- grid.81821.320000 0000 8970 9163Department of Oncology Medical. Hospital, Universitario La Paz, Madrid, Spain
| | - Ana Fernández-Montes
- grid.418883.e0000 0000 9242 242XDepartment of Medical Oncology, Complejo Hospitalario Universitario de Ourense – CHUO, Orense, Spain
| | - Oscar Castillo-Trujillo
- grid.411052.30000 0001 2176 9028Department of Medical Oncology, Hospital Universitario Central de Asturias, ISPA, Oviedo, Spain
| | - María M. Muñoz
- Department of Medical Oncology, Hospital General Virgen de La Luz, Cuenca, Spain
| | - Juana M. Cano-Cano
- grid.411096.bDepartment of Medical Oncology, Hospital General Universitario de Ciudad Real, Ciudad Real, Spain
| | - María J. Corral
- grid.5841.80000 0004 1937 0247Department of Clinical Psychology and Psychobiology, University of Barcelona, Barcelona, Spain
| | - Emilio Esteban
- grid.411052.30000 0001 2176 9028Department of Medical Oncology, Hospital Universitario Central de Asturias, ISPA, Oviedo, Spain
| | - Paula Jiménez-Fonseca
- grid.411052.30000 0001 2176 9028Department of Medical Oncology, Hospital Universitario Central de Asturias, ISPA, Oviedo, Spain
| | - Caterina Calderon
- Department of Clinical Psychology and Psychobiology, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
17
|
Dawud LM, Holbrook EM, Lowry CA. Evolutionary Aspects of Diverse Microbial Exposures and Mental Health: Focus on "Old Friends" and Stress Resilience. Curr Top Behav Neurosci 2023; 61:93-117. [PMID: 35947354 PMCID: PMC9918614 DOI: 10.1007/7854_2022_385] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
The prevalence of inflammatory disease conditions, including allergies, asthma, and autoimmune disorders, increased during the latter half of the twentieth century, as societies transitioned from rural to urban lifestyles. A number of hypotheses have been put forward to explain the increasing prevalence of inflammatory disease in modern urban societies, including the hygiene hypothesis and the "Old Friends" hypothesis. In 2008, Rook and Lowry proposed, based on the evidence that increased inflammation was a risk factor for stress-related psychiatric disorders, that the hygiene hypothesis or "Old Friends" hypothesis may be relevant to psychiatric disorders. Since then, it has become more clear that chronic low-grade inflammation is a risk factor for stress-related psychiatric disorders, including anxiety disorders, mood disorders, and trauma- and stressor-related disorders, such as posttraumatic stress disorder (PTSD). Evidence now indicates that persons raised in modern urban environments without daily contact with pets, relative to persons raised in rural environments in proximity to farm animals, respond with greater systemic inflammation to psychosocial stress. Here we consider the possibility that increased inflammation in persons living in modern urban environments is due to a failure of immunoregulation, i.e., a balanced expression of regulatory and effector T cells, which is known to be dependent on microbial signals. We highlight evidence that microbial signals that can drive immunoregulation arise from phylogenetically diverse taxa but are strain specific. Finally, we highlight Mycobacterium vaccae NCTC 11659, a soil-derived bacterium with anti-inflammatory and immunoregulatory properties, as a case study of how single strains of bacteria might be used in a psychoneuroimmunologic approach for prevention and treatment of stress-related psychiatric disorders.
Collapse
Affiliation(s)
- Lamya'a M Dawud
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Evan M Holbrook
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Christopher A Lowry
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA.
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA.
- Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Rocky Mountain Regional VA Medical Center (RMRVAMC), Aurora, CO, USA.
- Department of Physical Medicine and Rehabilitation, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, CO, USA.
- Center for Neuroscience, University of Colorado Boulder, Boulder, CO, USA.
- Center for Microbial Exploration, University of Colorado Boulder, Boulder, CO, USA.
- inVIVO Planetary Health, Worldwide Universities Network (WUN), West New York, NJ, USA.
| |
Collapse
|
18
|
von Mücke-Heim IA, Martin J, Uhr M, Ries C, Deussing JM. The human P2X7 receptor alters microglial morphology and cytokine secretion following immunomodulation. Front Pharmacol 2023; 14:1148190. [PMID: 37101546 PMCID: PMC10123291 DOI: 10.3389/fphar.2023.1148190] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 03/30/2023] [Indexed: 04/28/2023] Open
Abstract
Introduction: In recent years, purinergic signaling via the P2X7 receptor (P2X7R) on microglia has repeatedly been implicated in depression genesis. However, it remains unclear which role the human P2X7R (hP2X7R) plays in regulating both microglia morphology and cytokine secretion upon different environmental and immune stimuli, respectively. Methods: For this purpose, we used primary microglial cultures derived from a humanized microglia-specific conditional P2X7R knockout mouse line to emulate different gene-environment interactions between microglial hP2X7R and molecular proxies of psychosocial and pathogen-derived immune stimuli. Microglial cultures were subjected to treatments with the agonists 2'(3')-O-(4-benzoylbenzoyl)-ATP (BzATP) and lipopolysaccharides (LPS) combined with specific P2X7R antagonists (JNJ-47965567, A-804598). Results: Morphotyping revealed overall high baseline activation due to the in vitro conditions. Both BzATP and LPS + BzATP treatment increased round/ameboid microglia and decreased polarized and ramified morphotypes. This effect was stronger in hP2X7R-proficient (CTRL) compared to knockout (KO) microglia. Aptly, we found antagonism with JNJ-4796556 and A-804598 to reduce round/ameboid microglia and increase complex morphologies only in CTRL but not KO microglia. Single cell shape descriptor analysis confirmed the morphotyping results. Compared to KO microglia, hP2X7R-targeted stimulation in CTRLs led to a more pronounced increase in microglial roundness and circularity along with an overall higher decrease in aspect ratio and shape complexity. JNJ-4796556 and A-804598, on the other hand, led to opposite dynamics. In KO microglia, similar trends were observed, yet the magnitude of responses was much smaller. Parallel assessment of 10 cytokines demonstrated the proinflammatory properties of hP2X7R. Following LPS + BzATP stimulation, IL-1β, IL-6, and TNFα levels were found to be higher and IL-4 levels lower in CTRL than in KO cultures. Vice versa, hP2X7R antagonists reduced proinflammatory cytokine levels and increased IL-4 secretion. Discussion: Taken together, our results help disentangle the complex function of microglial hP2X7R downstream of various immune stimuli. In addition, this is the first study in a humanized, microglia-specific in vitro model identifying a so far unknown potential link between microglial hP2X7R function and IL-27 levels.
Collapse
Affiliation(s)
| | - Jana Martin
- Molecular Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Manfred Uhr
- Core Unit Analytics and Mass Spectrometry, Max Planck Institute of Psychiatry, Munich, Germany
| | - Clemens Ries
- Molecular Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Jan M. Deussing
- Molecular Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
- *Correspondence: Jan M. Deussing,
| |
Collapse
|
19
|
Khantakova JN, Bondar NP, Antontseva EV, Reshetnikov VV. Once induced, it lasts for a long time: the structural and molecular signatures associated with depressive-like behavior after neonatal immune activation. Front Cell Neurosci 2022; 16:1066794. [PMID: 36619667 PMCID: PMC9812963 DOI: 10.3389/fncel.2022.1066794] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 11/29/2022] [Indexed: 12/24/2022] Open
Abstract
Adverse factors such as stress or inflammation in the neonatal period can affect the development of certain brain structures and have negative delayed effects throughout the lifespan of an individual, by reducing cognitive abilities and increasing the risk of psychopathologies. One possible reason for these delayed effects is the neuroinflammation caused by neonatal immune activation (NIA). Neuroinflammation can lead to disturbances of neurotransmission and to reprogramming of astroglial and microglial brain cells; when combined, the two problems can cause changes in the cytoarchitecture of individual regions of the brain. In addition, neuroinflammation may affect the hypothalamic-pituitary-adrenal (HPA) axis and processes of oxidative stress, thereby resulting in higher stress reactivity. In our review, we tried to answer the questions of whether depressive-like behavior develops after NIA in rodents and what the molecular mechanisms associated with these disorders are. Most studies indicate that NIA does not induce depressive-like behavior in a steady state. Nonetheless, adult males (but not females or adolescents of both sexes) with experience of NIA exhibit marked depressive-like behavior when exposed to aversive conditions. Analyses of molecular changes have shown that NIA leads to an increase in the amount of activated microglia and astroglia in the frontal cortex and hippocampus, an increase in oxidative-stress parameters, a change in stress reactivity of the HPA axis, and an imbalance of cytokines in various regions of the brain, but not in blood plasma, thus confirming the local nature of the inflammation. Therefore, NIA causes depressive-like behavior in adult males under aversive testing conditions, which are accompanied by local inflammation and have sex- and age-specific effects.
Collapse
Affiliation(s)
- Julia N. Khantakova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk, Russia,Federal Government-Funded Scientific Institution Research Institute of Fundamental and Clinical Immunology (RIFCI), Novosibirsk, Russia,*Correspondence: Julia N. Khantakova
| | - Natalia P. Bondar
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk, Russia,Department of Natural Sciences, Novosibirsk State University, Novosibirsk, Russia
| | - Elena V. Antontseva
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk, Russia
| | - Vasiliy V. Reshetnikov
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk, Russia,Department of Biotechnology, Sirius University of Science and Technology, Sochi, Russia
| |
Collapse
|
20
|
Fecal microbiota transplantation from patients with rheumatoid arthritis causes depression-like behaviors in mice through abnormal T cells activation. Transl Psychiatry 2022; 12:223. [PMID: 35650202 PMCID: PMC9160267 DOI: 10.1038/s41398-022-01993-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 05/19/2022] [Accepted: 05/25/2022] [Indexed: 02/08/2023] Open
Abstract
Depression is common in patients with rheumatoid arthritis (RA); however, the precise mechanisms underlying a link between depression and RA remain unclear. Accumulating evidence suggests the role of gut-microbiota-brain axis in depression. In this study, we investigated whether collagen-induced arthritis (CIA) mice produce depression-like behaviors and abnormal composition of gut microbiota. Furthermore, we investigated whether fecal microbiota transplantation (FMT) from RA patients causes depression-like phenotypes in antibiotic cocktail (ABX)-treated mice. CIA mice displayed depression-like behaviors, increased blood levels of pro-inflammatory cytokine interleukin-6 (IL-6), decreased expression of synaptic proteins in the prefrontal cortex (PFC), and abnormal composition of gut microbiota. Furthermore, FMT from RA patients caused depression-like phenotypes, alterations of gut microbiota composition, increased levels of IL-6 and tumor necrosis factor-α (TNF-α), and downregulation of synaptic proteins in the PFC compared to FMT from healthy controls. There were correlations between relative abundance of microbiota and plasma cytokines, expression of synaptic proteins in the PFC or depression-like behaviors. Interestingly, FMT from RA patients induced T cells differentiation in Peyer's patches and spleen. Reduced percentage of Treg cells with an increase of Th1/Th2 index was observed in the mice after FMT from RA patients. These findings suggest that CIA mice exhibit depression-like behaviors, systemic inflammation, and abnormal composition of gut microbiota, and that FMT from RA patients produces depression-like behaviors in ABX-treated mice via T cells differentiation. Therefore, abnormalities in gut microbiota in RA patients may contribute to depression via gut-microbiota-brain axis.
Collapse
|
21
|
Depression in breast cancer patients: Immunopathogenesis and immunotherapy. Cancer Lett 2022; 536:215648. [DOI: 10.1016/j.canlet.2022.215648] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/13/2022] [Accepted: 03/14/2022] [Indexed: 01/10/2023]
|
22
|
Mindus C, van Staaveren N, Fuchs D, Gostner JM, Kjaer JB, Kunze W, Mian MF, Shoveller AK, Forsythe P, Harlander-Matauschek A. Regulatory T Cell Modulation by Lactobacillus rhamnosus Improves Feather Damage in Chickens. Front Vet Sci 2022; 9:855261. [PMID: 35478602 PMCID: PMC9036099 DOI: 10.3389/fvets.2022.855261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 03/17/2022] [Indexed: 11/17/2022] Open
Abstract
It is currently unclear whether potential probiotics such as lactic acid bacteria could affect behavioral problems in birds. To this end, we assessed whether a supplementation of Lactobacillus rhamnosus JB-1 can reduce stress-induced severe feather pecking (SFP), feather damage and fearfulness in adult birds kept for egg laying. In parallel, we assessed SFP genotypic and phenotypic-related immune responses and aromatic amino acid status linked to neurotransmitter production. Social stress aggravated plumage damage, while L. rhamnosus treatment improved the birds' feather cover in non-stressed birds, but did not impact fearfulness. Our data demonstrate the significant impact of L. rhamnosus supplementation on the immune system. L. rhamnosus supplementation induced immunosuppressive regulatory T cells and cytotoxic T cells in both the cecal tonsils and the spleen. Birds exhibiting the SFP phenotype possessed lower levels of cecal tonsils regulatory T cells, splenic T helper cells and a lower TRP:(PHE+TYR). Together, these results suggest that bacteria may have beneficial effects on the avian immune response and may be useful therapeutic adjuncts to counteract SFP and plumage damage, thus increasing animal health and welfare.
Collapse
Affiliation(s)
- Claire Mindus
- Department of Animal Biosciences, Ontario Agricultural College, University of Guelph, Guelph, ON, Canada
| | - Nienke van Staaveren
- Department of Animal Biosciences, Ontario Agricultural College, University of Guelph, Guelph, ON, Canada
| | - Dietmar Fuchs
- Biocenter, Institute of Biological Chemistry, Medical University of Innsbruck, Innsbruck, Austria
| | - Johanna M. Gostner
- Biocenter, Institute of Medical Biochemistry, Medical University of Innsbruck, Innsbruck, Austria
| | - Joergen B. Kjaer
- Institute of Animal Welfare and Animal Husbandry, Friedrich-Loeffler-Institut, Celle, Germany
| | - Wolfgang Kunze
- Brain-Body Institute, St. Joseph's Healthcare, McMaster University, Hamilton, ON, Canada
| | - M. Firoz Mian
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Anna K. Shoveller
- Department of Animal Biosciences, Ontario Agricultural College, University of Guelph, Guelph, ON, Canada
| | - Paul Forsythe
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, Canada
- Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Alexandra Harlander-Matauschek
- Department of Animal Biosciences, Ontario Agricultural College, University of Guelph, Guelph, ON, Canada
- *Correspondence: Alexandra Harlander-Matauschek
| |
Collapse
|
23
|
Old and New Biomarkers for Infection, Inflammation, and Autoimmunity in Treatment-Resistant Affective and Schizophrenic Spectrum Disorders. Pharmaceuticals (Basel) 2022; 15:ph15030299. [PMID: 35337097 PMCID: PMC8949012 DOI: 10.3390/ph15030299] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/24/2022] [Accepted: 02/24/2022] [Indexed: 02/06/2023] Open
Abstract
Affective (AF) and Schizophrenic (SZ) Spectrum disorders manifest with risk factors, involving inflammatory processes linked to infections and autoimmunity. This study searched for novel biomarkers in cerebrospinal fluid (CSF) and peripheral blood. A total of 29 AF and 39 SZ patients with treatment-resistant disease were included. In CSF, the chemokine IL-8 was significantly elevated in AF and SZ patients. IL-8 promotes chemotaxis by neutrophils and may originate from different tissues. S100B, a glia-derived brain damage marker, was higher in CSF from AF than SZ patients. Among the plasma-derived biomarkers, ferritin was elevated in AF and SZ. Soluble CD25, indicating Treg dysfunction, was higher in SZ than in AF patients. Interferon-γ, implying virus-specific immune activation, was positive in selective AF patients, only. Both groups showed elevated expression of immunosuppressive CD33 on monocytes, but higher amounts of CD123+ plasmacytoid dendritic cells were restricted to SZ. In conclusion, chemotactic IL-8 indicates neuronal stress and inflammation in the CSF of both groups. Novel plasma-derived biomarkers such as sCD25 and monocytic CD33 distinguish SZ from AF with an autoimmune phenotype.
Collapse
|
24
|
Transcriptomic signatures of treatment response to the combination of escitalopram and memantine or placebo in late-life depression. Mol Psychiatry 2021; 26:5171-5179. [PMID: 32382137 PMCID: PMC9922535 DOI: 10.1038/s41380-020-0752-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 04/17/2020] [Accepted: 04/22/2020] [Indexed: 12/17/2022]
Abstract
Drugs that target glutamate neuronal transmission, such as memantine, offer a novel approach to the treatment of late-life depression, which is frequently comorbid with cognitive impairment. The results of our recently published double-blind, randomized, placebo-controlled trial of escitalopram or escitalopram/memantine in late-life depression with subjective memory complaints (NCT01902004) indicated no differences between treatments in depression remission, but additional benefits in cognition at 12-month follow-up with combination treatment. To identify pathways and biological functions uniquely induced by combination treatment that may explain cognitive improvements, we generated transcriptional profiles of remission compared with non-remission from whole blood samples. Remitters to escitalopram compared with escitalopram/memantine combination treatment display unique patterns of gene expression at baseline and 6 months after treatment initiation. Functional enrichment analysis demonstrates that escitalopram-based remission associates to functions related to cellular proliferation, apoptosis, and inflammatory response. Escitalopram/memantine-based remission, however, is characterized by processes related to cellular clearance, metabolism, and cytoskeletal dynamics. Both treatments modulate inflammatory responses, albeit via different effector pathways. Additional research is needed to understand the implications of these results in explaining the observed superior effects of combination treatment on cognition observed with prolonged treatment.
Collapse
|
25
|
Gasnier M, Ellul P, Plaze M, Ahad PA. A New Look on an Old Issue: Comprehensive Review of Neurotransmitter Studies in Cerebrospinal Fluid of Patients with Schizophrenia and Antipsychotic Effect on Monoamine's Metabolism. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE 2021; 19:395-410. [PMID: 34294610 PMCID: PMC8316661 DOI: 10.9758/cpn.2021.19.3.395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/30/2020] [Accepted: 12/28/2020] [Indexed: 11/18/2022]
Abstract
Neurotransmitters metabolism has a key role in the physiopathology of schizophrenia as demonstrated by studies measuring monoamine metabolites in patient’s cerebrospinal fluid (CSF) since the beginning of the antipsychotic use. This comprehensive review aims to understand the anomalies of CSF monoamines in schizophrenia and their correlation with clinical and paraclinical features. We also review the influence of antipsychotic treatment on CSF monoamines and discuss the connection with metabolic and inflammatory processes. Studies comparing CSF homovanillic acid (HVA) levels between patients and controls are miscellaneous, due to the heterogeneity of samples studies. However, low HVA is associated with more positive symptoms and a poorer outcome and negatively correlated with brain ventricle size. Based on humans and animals’ studies, antipsychotic treatments increase HVA during the first week of administration and decrease progressively over the time with a fall-off after withdrawal. 5‐hydroxyindolacetic acetic acid levels do not seem to be different in the patient’s CSF compared to controls. Considering metabolic co-factors of neurotrans-mitters synthesis, there is evidence supporting an increase of kynurenic acid in the CSF of patients with schizophrenia. Few studies explore folate metabolism in CSF. Literature also emphasizes the relationship between folate metabolism, inflammation and monoamine’s metabolism. Those results suggest that the CSF monoamines could be correlated with schizophrenia symptoms and treatment outcome. However, further studies, exploring the role of CSF monoamines as biomarkers of disease severity and response to treatment are needed. They should assess the antipsychotic prescription, inflammatory markers and folate metabolism as potential confounding factors.
Collapse
Affiliation(s)
- Matthieu Gasnier
- Department of Psychiatry, MOODS Team, Paris Saclay University, Bicetre Hospital, AP-HP, Paris, France
| | - Pierre Ellul
- Department of Child and Adolescent Psychiatry, Robert Debré Hospital, AP-HP, Paris, France
| | - Marion Plaze
- Department of Psychiatry, Service Hospitalo Universitaire, Sainte Anne Hospital, Paris, France
| | - Pierre Abdel Ahad
- Department of Psychiatry, Service Hospitalo Universitaire, Sainte Anne Hospital, Paris, France
| |
Collapse
|
26
|
Enrico P, Delvecchio G, Turtulici N, Pigoni A, Villa FM, Perlini C, Rossetti MG, Bellani M, Lasalvia A, Bonetto C, Scocco P, D’Agostino A, Torresani S, Imbesi M, Bellini F, Veronese A, Bocchio-Chiavetto L, Gennarelli M, Balestrieri M, Colombo GI, Finardi A, Ruggeri M, Furlan R, Brambilla P. Classification of Psychoses Based on Immunological Features: A Machine Learning Study in a Large Cohort of First-Episode and Chronic Patients. Schizophr Bull 2021; 47:1141-1155. [PMID: 33561292 PMCID: PMC8266656 DOI: 10.1093/schbul/sbaa190] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
For several years, the role of immune system in the pathophysiology of psychosis has been well-recognized, showing differences from the onset to chronic phases. Our study aims to implement a biomarker-based classification model suitable for the clinical management of psychotic patients. A machine learning algorithm was used to classify a cohort of 362 subjects, including 160 first-episode psychosis patients (FEP), 70 patients affected by chronic psychiatric disorders (schizophrenia, bipolar disorder, and major depressive disorder) with psychosis (CRO) and 132 health controls (HC), based on mRNA transcript levels of 56 immune genes. Models distinguished between FEP, CRO, and HC and between the subgroup of drug-free FEP and HC with a mean accuracy of 80.8% and 90.4%, respectively. Interestingly, by using the feature importance method, we identified some immune gene transcripts that contribute most to the classification accuracy, possibly giving new insights on the immunopathogenesis of psychosis. Therefore, our results suggest that our classification model has a high translational potential, which may pave the way for a personalized management of psychosis.
Collapse
Affiliation(s)
- Paolo Enrico
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Giuseppe Delvecchio
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Nunzio Turtulici
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Alessandro Pigoni
- MoMiLab Research Unit, IMT School for Advanced Studies Lucca, Lucca, Italy
| | | | - Cinzia Perlini
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Clinical Psychology, University of Verona, Verona, Italy
| | - Maria Gloria Rossetti
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Psychiatry, University of Verona, Verona, Italy
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Marcella Bellani
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Psychiatry, University of Verona, Verona, Italy
- AOUI – Verona Hospital Trust, Verona, Italy
| | - Antonio Lasalvia
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Psychiatry, University of Verona, Verona, Italy
- AOUI – Verona Hospital Trust, Verona, Italy
| | - Chiara Bonetto
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Psychiatry, University of Verona, Verona, Italy
| | - Paolo Scocco
- Department of Mental Health, AULSS 6 Euganea, Padua, Italy
| | - Armando D’Agostino
- Department of Health Sciences, San Paolo University Hospital, University of Milan, Milan, Italy
| | - Stefano Torresani
- Department of Psychiatry, ULSS, Bolzano Suedtiroler Sanitaetbetrieb- Azienda Sanitaria dell’Alto Adige, Bolzano, Italy
| | | | | | | | - Luisella Bocchio-Chiavetto
- Genetics Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
- Faculty of Psychology, eCampus University, Novedrate, Como, Italy
| | - Massimo Gennarelli
- Genetics Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Matteo Balestrieri
- Unit of Psychiatry, Department of Medicine (DAME), University of Udine, Udine, Italy
| | - Gualtiero I Colombo
- Centro Cardiologico Monzino IRCCS, Immunology and Functional Genomics Unit, Milan, Italy
| | - Annamaria Finardi
- Clinical Neuroimmunology Unit, Institute of Experimental Neurology, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Mirella Ruggeri
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Psychiatry, University of Verona, Verona, Italy
- AOUI – Verona Hospital Trust, Verona, Italy
| | - Roberto Furlan
- Clinical Neuroimmunology Unit, Institute of Experimental Neurology, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Paolo Brambilla
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
27
|
Sluiter F, Incollingo Rodriguez AC, Nephew BC, Cali R, Murgatroyd C, Santos HP. Pregnancy associated epigenetic markers of inflammation predict depression and anxiety symptoms in response to discrimination. Neurobiol Stress 2020; 13:100273. [PMID: 33344726 PMCID: PMC7739167 DOI: 10.1016/j.ynstr.2020.100273] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 11/16/2020] [Accepted: 11/16/2020] [Indexed: 02/06/2023] Open
Abstract
Latina mothers, who have one of the highest fertility rates among ethnic groups in the United States (US), often experience discrimination. Psychosocial influences during pregnancy, such as discrimination stress, promotes inflammation. However, the role of epigenetic markers of inflammation as a mediator between, and predictor of, maternal discrimination stress and neuropsychiatric outcomes has not been extensively studied. The current study investigates the role of DNA methylation at FOXP3 Treg-cell-specific demethylated region (TSDR), as a marker of regulatory T (Treg) cells that are important negative regulators of inflammation, and the promoter of tumour necrosis factor-alpha (TNF-α) gene, an important pro-inflammatory cytokine, in relation to discrimination stress during pregnancy and depression and anxiety symptomatology. A sample of 148 Latina women residing in the US (mean age 27.6 years) were assessed prenatally at 24–32 weeks’ gestation and 4–6 weeks postnatally for perceived discrimination exposure (Everyday Discrimination Scale, EDS), emotional distress (depression, anxiety, perinatal-specific depression), acculturation, and acculturative stress. DNA methylation levels at the FOXP3 and TNFα promoter regions from blood samples collected at the prenatal stage were assessed by bisulphite pyrosequencing. Regression analyses showed that prenatal EDS associated with postnatal emotional distress, depression and anxiety symptoms only in those individuals with higher than mean levels of FOXP3 TSDR and TNFα promoter methylation; no such significant associations were found in those with lower than mean levels of methylation for either. We further found that these relationships were mediated by TNFα only in those with high FOXP3 TSDR methylation, implying that immunosuppression via TNFα promoter methylation buffers the impact of discrimination stress on postpartum symptomatology. These results indicate that epigenetic markers of immunosuppression and inflammation play an important role in resilience or sensitivity, respectively, to prenatal stress.
Collapse
Affiliation(s)
- Femke Sluiter
- Department of Life Sciences, Manchester Metropolitan University, Manchester, United Kingdom
| | | | - Benjamin C Nephew
- Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester, MA, United States
| | - Ryan Cali
- University of Massachusetts Medical School, Worcester, MA, United States
| | - Chris Murgatroyd
- Department of Life Sciences, Manchester Metropolitan University, Manchester, United Kingdom
| | - Hudson P Santos
- Biobehavioral Laboratory, School of Nursing, University of North Carolina at Chapel Hill, North Carolina, United States
| |
Collapse
|
28
|
Abstract
The immune system consists of a complex biological and psychological network designed for fighting against infections and to protect the body from pathogen factors, including the internal ones. In the past, for a long time inflammation and infectious diseases were thought to be only the result of the genetic heritage and the biological functioning of the body, when the pathogenic factors acted within the body. Studies in recent decades stressed the importance of psychological balance and mental health on the body immunity. Psychoneuroimmunology studies indicated the thoughts and emotional patterns, and the psychological dynamics are strongly interrelated with the immune response. Moreover, the immunological mechanisms not only regulates the health of the person, but they are also an important part of the individual adaptive process in the environment. In various studies, the results of each treatment modality (drug interventions and psychosocial interventions) were observed and compared in patients with mental health problems associated with immune reactions (inflammation). Psychosocial interventions suggest increased efficiency in reducing inflammation and improving immune system function.
Collapse
Affiliation(s)
- Cristian Vasile
- Educational Sciences Department, Petroleum-Gas University of Ploiesti, 100680 Ploiesti, Romania
| |
Collapse
|
29
|
Berben L, Floris G, Kenis C, Dalmasso B, Smeets A, Vos H, Neven P, Antoranz Martinez A, Laenen A, Wildiers H, Hatse S. Age-related remodelling of the blood immunological portrait and the local tumor immune response in patients with luminal breast cancer. Clin Transl Immunology 2020; 9:e1184. [PMID: 33024560 PMCID: PMC7532981 DOI: 10.1002/cti2.1184] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 09/07/2020] [Accepted: 09/07/2020] [Indexed: 12/26/2022] Open
Abstract
Objectives Aging is associated with altered immune function and chronic low-grade inflammation, referred to as immunosenescence. As breast cancer is an age-related disease, the impact of aging on tumor immune responses may have important consequences. However, effects of immunosenescence on breast tumor immune infiltration remain largely unknown. Methods This exploratory study investigated a broad panel of immune/senescence markers in peripheral blood and in the tumor microenvironment of young, middle-aged and old patients diagnosed with early invasive luminal (hormone-sensitive, HER2-negative) breast cancer. In the old group, G8-scores were computed as a correlate for clinical frailty. Results Significant age-related changes in plasma levels of several inflammatory mediators (IL-1α, IP-10, IL-8, MCP-1, CRP), immune checkpoint markers (Gal-9, sCD25, TIM-3, PD-L1), IGF-1 and circulating miRs (miR-18a, miR-19b, miR-20, miR-155, miR-195 and miR-326) were observed. Shifts were observed in distinct peripheral blood mononuclear cell populations, particularly naive CD8+ T-cells. At the tumor level, aging was associated with lower total lymphocytic infiltration, together with decreased abundance of several immune cell markers, especially CD8. The relative fractions of cell subsets in the immune infiltrate were also altered. Clinical frailty was associated with higher frequencies of exhausted/senescent (CD27-CD28- and/or CD57+) terminally differentiated CD8+ cells in the blood and with increased tumor infiltration by FOXP3+ cells. Conclusion Aging and frailty are associated with profound changes of the blood and tumor immune profile in luminal breast cancer, pointing to a different interplay between tumor cells, immune cells and inflammatory mediators at higher age.
Collapse
Affiliation(s)
- Lieze Berben
- Laboratory of Experimental Oncology Department of Oncology KU Leuven Leuven Belgium
| | - Giuseppe Floris
- Department of Pathology University Hospitals Leuven Leuven Belgium.,Laboratory of Translational Cell and Tissue Research Department of Imaging and Pathology KU Leuven Leuven Belgium
| | - Cindy Kenis
- Department of General Medical Oncology and Geriatric Medicine University Hospitals Leuven Leuven Belgium
| | - Bruna Dalmasso
- Genetis of Rare Cancers Department of Internal Medicine and Medical Specialties (DiMI) University of Genoa and IRCCS Ospedale Policlinico San Martino Genoa Italy
| | - Ann Smeets
- Department of Surgical Oncology KU Leuven, University Hospitals Leuven Leuven Belgium
| | - Hanne Vos
- Department of Surgical Oncology KU Leuven, University Hospitals Leuven Leuven Belgium
| | - Patrick Neven
- Department of Gynaecology and Obstetrics University Hospitals Leuven Leuven Belgium
| | - Asier Antoranz Martinez
- Laboratory of Translational Cell and Tissue Research Department of Imaging and Pathology KU Leuven Leuven Belgium
| | - Annouschka Laenen
- Interuniversity Centre for Biostatistics and Statistical Bioinformatics Leuven Belgium
| | - Hans Wildiers
- Laboratory of Experimental Oncology Department of Oncology KU Leuven Leuven Belgium.,Department of General Medical Oncology University Hospitals Leuven Leuven Belgium
| | - Sigrid Hatse
- Laboratory of Experimental Oncology Department of Oncology KU Leuven Leuven Belgium
| |
Collapse
|
30
|
Çakici N, Sutterland AL, Penninx BWJH, Dalm VA, de Haan L, van Beveren NJM. Altered peripheral blood compounds in drug-naïve first-episode patients with either schizophrenia or major depressive disorder: a meta-analysis. Brain Behav Immun 2020; 88:547-558. [PMID: 32330592 DOI: 10.1016/j.bbi.2020.04.039] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 03/30/2020] [Accepted: 04/15/2020] [Indexed: 12/30/2022] Open
Abstract
IMPORTANCE Schizophrenia and major depressive disorder (MDD) are associated with increased risks of immunologic disease and metabolic syndrome. It is unclear to what extent growth, immune or glucose dysregulations are similarly present in these disorders without the influence of treatment or chronicity. OBJECTIVE To conduct a meta-analysis investigating whether there are altered peripheral growth, immune or glucose metabolism compounds in drug-naïve first-episode patients with schizophrenia or MDD compared with controls. DATA SOURCES AND STUDY SELECTION Case-control studies reporting compound measures in drug-naïve first-episode patients with schizophrenia or MDD compared with controls in the Embase, PubMed and PsycINFO databases. DATA EXTRACTION AND SYNTHESIS Two independent authors extracted data for a random-effects meta-analysis. MAIN OUTCOMES AND MEASURES Peripheral growth, immune or glucose metabolism compounds in schizophrenia or MDD compared with controls. Standardized mean differences were quantified with Hedges' g (g). RESULTS 74 studies were retrieved comprising 3453 drug-naïve first-episode schizophrenia patients and 4152 controls, and 29 studies were retrieved comprising 1095 drug-naïve first-episode MDD patients and 1399 controls. Growth factors: brain-derived neurotrophic factor (BDNF) (g = -0.77, P < .001) and nerve growth factor (NGF) (g = -2.51, P = .03) were decreased in schizophrenia. For MDD, we observed a trend toward decreased BDNF (g = -0.47, P = .19) and NGF (g = -0.33, P = .08) levels, and elevated vascular endothelial growth factor levels (g = 0.40, P = .03). Immune factors: interleukin (IL)-6 (g = 0.95, P < .001), IL-8 (g = 0.59, P = .001) and tumor necrosis factor alpha (TNFα) (g = 0.48, P = .002) were elevated in schizophrenia. For C-reactive protein (CRP) (g = 0.57, P = .09), IL-4 (g = 0.44, P = .10) and interferon gamma (g = 0.33, P = .11) we observed a trend toward elevated levels in schizophrenia. In MDD, IL-6 (g = 0.62, P = .007), TNFα (g = 1.21, P < .001), CRP (g = 0.53, P < .001), IL-1β (g = 1.52, P = .009) and IL-2 (g = 4.41, P = .04) were elevated, whereas IL-8 (g = -0.84, P = .01) was decreased. The fasting glucose metabolism factors glucose (g = 0.24, P = .003) and insulin (g = 0.38, P = .003) were elevated in schizophrenia. CONCLUSIONS AND RELEVANCE Both schizophrenia and MDD show alterations in growth and immune factors from disease onset. An altered glucose metabolism seems to be present from onset in schizophrenia. These findings support efforts for further research into transdiagnostic preventive strategies and augmentation therapy for those with immune or metabolic dysfunctions.
Collapse
Affiliation(s)
- Nuray Çakici
- Department of Psychiatry and Amsterdam Neuroscience, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Parnassia Academy, Parnassia Psychiatric Institute, The Hague, the Netherlands.
| | - Arjen L Sutterland
- Department of Psychiatry and Amsterdam Neuroscience, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Brenda W J H Penninx
- Department of Psychiatry, Amsterdam Public Health Research Institute and Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit, Amsterdam, the Netherlands
| | - Virgil A Dalm
- Department of Internal Medicine, Division of Clinical Immunology and Department of Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Lieuwe de Haan
- Department of Psychiatry and Amsterdam Neuroscience, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Nico J M van Beveren
- Parnassia Academy, Parnassia Psychiatric Institute, The Hague, the Netherlands; Department of Psychiatry, Department of Neuroscience, Erasmus Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
31
|
Vasileva AV. [Pandemic and mental adjustment disorders. Therapy options]. Zh Nevrol Psikhiatr Im S S Korsakova 2020; 120:146-152. [PMID: 32621481 DOI: 10.17116/jnevro2020120051146] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The article considers the main triggers of adjustment and anxiety disorders in the time of pandemic that brings significant changes in all life dimensions with the high uncertainty level. The article highlights a role of coronavirus infodemic in the development of anxiety disorders. The main targets for cognitive, emotional and behavioral interventions are suggested. Recommendations for anxiety adjustment disorders psychopharmacotherapy with the use of tranquillizers as treatment of choice are given. Prevention measures for medical doctors working in conditions of pandemic are proposed.
Collapse
Affiliation(s)
- A V Vasileva
- Bekhterev National Research Medical Center for Psychiatry and Neurology, St. Petersburg, Russia.,Mechnikov North-Western State Medical University, St. Petersburg, Russia
| |
Collapse
|
32
|
Petralia MC, Mazzon E, Fagone P, Basile MS, Lenzo V, Quattropani MC, Di Nuovo S, Bendtzen K, Nicoletti F. The cytokine network in the pathogenesis of major depressive disorder. Close to translation? Autoimmun Rev 2020; 19:102504. [PMID: 32173514 DOI: 10.1016/j.autrev.2020.102504] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 12/25/2019] [Indexed: 12/18/2022]
Abstract
Major depressive disorder (MDD) is a common condition that afflicts the general population across a broad spectrum of ages and social backgrounds. MDD has been identified by the World Health Organization as a leading cause of disability worldwide. Approximately 30% of patients are poor responsive to standard of care (SOC) treatment and novel therapeutic approaches are warranted. Since chronic inflammation, as it is often observed in certain cancers, type 2 diabetes, psoriasis and chronic arthritis, are accompanied by depression, it has been suggested that immunoinflammatory processes may be involved in the pathogenesis of MDD. Cytokines are a group of glycoproteins secreted from lymphoid and non-lymphoid cells that orchestrate immune responses. It has been suggested that a dysregulated production of cytokines may be implicated in the pathogenesis and maintenance of MDD. On the basis of their functions, cytokines can be subdivided in pro-inflammatory and anti-inflammatory cytokines. Since abnormal blood and cerebrospinal fluid of both pro and anti-inflammatory cytokines are altered in MDD, it has been suggested that abnormal cytokine homeostasis may be implicated in the pathogenesis of MDD and possibly to induction of therapeutic resistance. We review current data that indicate that cytokines may represent a useful tool to identify MDD patients that may benefit from tailored immunotherapeutic approaches and may represent a potential tailored therapeutic target.
Collapse
Affiliation(s)
| | | | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Maria Sofia Basile
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Vittorio Lenzo
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | | | - Santo Di Nuovo
- Department of Educational Sciences, University of Catania, Catania, Italy
| | | | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.
| |
Collapse
|
33
|
Zhang W, Sun Q, Jia L, Li M. Ketamine exerts a protective role in a cell-based model of major depressive disorder via the inhibition of apoptosis and inflammation and activation of the Krebs cycle. Bosn J Basic Med Sci 2020; 20:44-55. [PMID: 31215856 PMCID: PMC7029208 DOI: 10.17305/bjbms.2019.4222] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 05/13/2019] [Indexed: 11/26/2022] Open
Abstract
Major depressive disorder (MDD) is one of the most common psychiatric disorders characterized by major depressive episodes. Although great efforts have been made to develop antidepressant drugs that target the monoaminergic system, these drugs are effective in only approximately 50% of MDD patients. In this study, we established a model of depression in PC12 cells using corticosterone to investigate the effect of ketamine and nuclear factor-κB (NF-κB) on the cell viability, apoptosis, levels of pro-inflammatory cytokines, apoptosis-related molecules, and enzymes of the Krebs cycle. PC12 cells were divided into control (no treatment, NC), ketamine treatment (KT), ketamine treatment with the inhibition of NF-κB (KI), and ketamine treatment with the overexpression of NF-κB (KO) group. Blood serum samples were collected from patients with MDD (n = 10) and healthy controls (n = 10) between 2015 and 2017. Ketamine significantly increased the viability and decreased the apoptosis of PC12 cells in KT and KI vs. NC group, but not in KO group. The levels of anti-apoptotic molecules and Krebs cycle enzymes were significantly increased in KI vs. KT group, while the levels of pro-apoptotic molecules and pro-inflammatory cytokines were decreased in KI vs. KT group. In addition, the levels of pro-inflammatory cytokines in the serum of MDD patients were significantly increased. The antidepressant effect of ketamine was enhanced in KI and reduced in KO group. Our results indicate that ketamine exerts its antidepressant effect via the inhibition of apoptosis and inflammation and the activation of the Krebs cycle in PC12 cells. NF-κB might be a potential therapeutic target in MDD.
Collapse
Affiliation(s)
- Wenfei Zhang
- Psychological Outpatient Department, Dezhou People's Hospital, Dezhou, China.
| | - Qian Sun
- Department of Pharmacy, Dezhou People's Hospital, Dezhou, China.
| | - Lingling Jia
- Department of Pharmacy, Dezhou People's Hospital, Dezhou, China.
| | - Ming Li
- Department of Orthopedics, Dezhou People's Hospital, Dezhou, China.
| |
Collapse
|
34
|
McFarland DC. Neutrophil to Lymphocyte Ratio in Lung Cancer: Implications for Depressive Symptoms and Survival. ACTA ACUST UNITED AC 2020; 3. [PMID: 34056574 PMCID: PMC8162915 DOI: 10.31487/j.cor.2020.06.12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Background: Depression very commonly appears in the presence of lung cancer. Multiple contexts have shown that depression is associated with inflammation. The Neutrophil to Lymphocyte Ratio (NLR) provides an easy to interpret the measure of both inflammation and immunity, but its use as an inflammatory biomarker has not been evaluated in patients with lung cancer. We hypothesize that NLR will be elevated in depressed patients with lung cancer and that both elevated NRL and depression will have prognostic implications. Methods: Patients (n=109) were assessed for depression and anxiety using the Hospital .Anxiety and Depression Scale (HADS) and for distress using the Distress Thermometer. NLR was derived from a complete blood count obtained on the day of the cross-sectional survey. Data were dichotomized (NLR ≥5 and HADS-D ≥8) and analysed for survival estimations using Kaplan-Meier plots. Results: NLR was found to be significantly correlated with depression (r=.21, p=.03) and was associated with depression while controlling for age, sex, and marital status (β=.21, p=.004). NLR trended toward correlation with anxiety (r=.19, p=.07). Elevated NLR (≥5) predicted for worse survival (chi square= 10.08, p=.001), which was similarly seen when combined with meeting depression criteria (chi square = 16.00,p<.001). Conclusion: NLR provides a reasonable assessment of lung cancer related inflammation with survival implications that may indicate the presence of depression. These results warrant further research.
Collapse
Affiliation(s)
- Daniel C McFarland
- Department of Psychiatry and Behavioral Sciences, Memorial Sloan Kettering Cancer Center, New York, USA
| |
Collapse
|
35
|
Peripheral blood GILZ mRNA levels in depression and following electroconvulsive therapy. Psychoneuroendocrinology 2019; 101:304-310. [PMID: 30602137 DOI: 10.1016/j.psyneuen.2018.12.234] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 12/20/2018] [Accepted: 12/22/2018] [Indexed: 12/12/2022]
Abstract
Dysregulation of the hypothalamic-pituitary-adrenocortical (HPA)-axis is commonly observed in patients with depression. The delayed feedback system that mediates inhibition of HPA-axis activation is regulated by glucocorticoid receptors (GRs) found in stress-responsive areas of the brain. Glucocorticoid-induced leucine zipper (GILZ) is a key molecule in glucocorticoid biology and is thought to mediate the downstream anti-inflammatory effects of GRs. Previous reports suggest that GILZ levels are altered in the blood and brains of patients with, and animal models of, depression. However, no study has yet investigated the effects of antidepressant treatment on GILZ. Therefore, our aim was to examine peripheral blood GILZ mRNA levels in patients with depression (n = 88) compared to age- and sex-matched healthy controls (n = 63), and in patients with depression following treatment with a course of electroconvulsive therapy (ECT). We also assessed the relationship between GILZ and mood and clinical outcomes following ECT. GILZ mRNA levels were assessed using qRT-PCR. GILZ levels were found to be significantly lower in patients with depression compared to controls (p < 0.002), and ECT further decreased GILZ levels (p = 0.05). Both of these results survived adjustment for potential covariates. However, we found no association between GILZ and mood scores. Overall, these results suggest that GILZ is involved in the pathophysiology of depression and the peripheral molecular response to ECT.
Collapse
|
36
|
Wu H, Lv W, Pan Q, Kalavagunta PK, Liu Q, Qin G, Cai M, Zhou L, Wang T, Xia Z, Shang J. Simvastatin therapy in adolescent mice attenuates HFD-induced depression-like behavior by reducing hippocampal neuroinflammation. J Affect Disord 2019; 243:83-95. [PMID: 30236762 DOI: 10.1016/j.jad.2018.09.022] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 08/06/2018] [Accepted: 09/10/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND A high-fat diet (HFD)-induced obesity/hyperlipidemia is accompanied by hormonal and neurochemical changes that can be associated with depression. Emerging studies indicate that simvastatin (SMV, decreasing cholesterol levels) has therapeutic effects on neurological and neuropsychiatric diseases through hippocampal-dependent function. However, the studies on the HFD exposure in adolescent animals, which investigate the neuroprotective effects of SMV on the hippocampal morphology, serotonin (5-HT) system and inflammation, are limited. Hence, the aim of this study was to determine whether SMV attenuates HFD-induced major depressive disorders in adolescent animals and, more specifically, acts as an anti-neuroinflammatory response. METHODS Twenty-four male C57BL/6 mice were fed a control (n = 8), HFD (n = 8) and HFD + SMV (n = 8) for 14 weeks. In HFD + SMV group, SMV (10 mg/kg) was administrated from the 10th week of HFD feeding. The open field test (OFT) and the tail suspension test (TST) were used to examine the effect of SMV on behavioral performance. HE and Nissl staining were conducted to detect hippocampal morphology and neural survival. Expression of the inflammatory cytokine genes was assayed by quantitative polymerase chain reaction (Q-PCR). RESULTS Firstly, alterations in lipid parameters were minimized after SMV treatment. HFD-induced depression-like behavior, which was evidenced by an increase in immobility time in TST along with considerable decrease in locomotion activity, was significantly attenuated by SMV therapy for 4 weeks. Additionally, SMV could reduce HFD-induced structural abnormality, neuronal injury, serotonergic system disturbance and pro-inflammatory cytokine over-expression in the hippocampus. Neuroimmunological changes in central hippocampus displayed a similar characteristic (only IL-1β, IL-6, TNF-α) with that in periphery spleen, whereas they appeared in an entirely opposite trend with that in cerebral cortex. CONCLUSION Our results suggest that SMV may be a promising treatment for HFD-induced depression-like behavior during adolescent period through brain region-specific neuroninflammatory mechanisms.
Collapse
Affiliation(s)
- Huali Wu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wenting Lv
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing 211198, China; School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Qi Pan
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing 211198, China; School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Praveen Kumar Kalavagunta
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing 211198, China; School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Qiongzhen Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing 211198, China; School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Guohong Qin
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing 211198, China; School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Minxuan Cai
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing 211198, China; School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Liangliang Zhou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing 211198, China; School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Tao Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing 211198, China; School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Zhenjiang Xia
- Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810008, Qinghai Province, China; Qinghai Key Laboratory of Tibetan Medicine Pharmacology and Safety Evaluation, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810008, Qinghai Province, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jing Shang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing 211198, China; School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Qinghai Key Laboratory of Tibetan Medicine Pharmacology and Safety Evaluation, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810008, Qinghai Province, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|