51
|
Xue R, Zhang H, Pan J, Du Z, Zhou W, Zhang Z, Tian Z, Zhou R, Bai L. Peripheral Dopamine Controlled by Gut Microbes Inhibits Invariant Natural Killer T Cell-Mediated Hepatitis. Front Immunol 2018; 9:2398. [PMID: 30386344 PMCID: PMC6199378 DOI: 10.3389/fimmu.2018.02398] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Accepted: 09/27/2018] [Indexed: 12/31/2022] Open
Abstract
Neurotransmitters have been shown to regulate immune responses, and thereby are critically related to autoimmune diseases. Here we showed that depletion of dopaminergic neurons significantly promoted activation of hepatic iNKT cells and augmented concanavalin A (Con A)-induced liver injury. The suppressive effect of dopamine on iNKT cells was mediated by D1-like receptor-PKA pathway. Clearance of gut microbiota by antibiotic cocktail reduced synthesis of dopamine in intestines and exacerbated liver damage, and that could be restored by recovery of gut microbiota or replenishment of D1-like receptor agonist. Our results demonstrate that peripheral dopamine controlled by gut microbes inhibits IL4 and IFNγ production in iNKT cells and suppresses iNKT cell-mediated hepatitis. Together, we propose a gut microbe-nervous system-immune system regulatory axis in modulating autoimmune hepatitis.
Collapse
Affiliation(s)
- Rufeng Xue
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Huimin Zhang
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Jun Pan
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Zhiwei Du
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Wenjie Zhou
- Key Laboratory of Brain Function and Disease of Chinese Academy of Science, Department of Biophysics and Neurobiology, University of Science and Technology of China, Hefei, China
| | - Zhi Zhang
- Key Laboratory of Brain Function and Disease of Chinese Academy of Science, Department of Biophysics and Neurobiology, University of Science and Technology of China, Hefei, China
| | - Zhigang Tian
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Rongbin Zhou
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Innovation Center for Cell Signaling Network, Hefei, China
| | - Li Bai
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Innovation Center for Cell Signaling Network, Hefei, China
| |
Collapse
|
52
|
Osorio-Barrios F, Prado C, Contreras F, Pacheco R. Dopamine Receptor D5 Signaling Plays a Dual Role in Experimental Autoimmune Encephalomyelitis Potentiating Th17-Mediated Immunity and Favoring Suppressive Activity of Regulatory T-Cells. Front Cell Neurosci 2018; 12:192. [PMID: 30042660 PMCID: PMC6048279 DOI: 10.3389/fncel.2018.00192] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 06/14/2018] [Indexed: 01/11/2023] Open
Abstract
A number of studies have shown pharmacologic evidence indicating that stimulation of type I dopamine receptor (DR), favors T-helper-17 (Th17)-mediated immunity involved in experimental autoimmune encephalomyelitis (EAE) and in some other inflammatory disorders. Nevertheless, the lack of drugs that might discriminate between DRD1 and DRD5 has made the pharmacological distinction between the two receptors difficult. We have previously shown genetic evidence demonstrating a relevant role of DRD5-signaling in dendritic cells (DCs) favoring the CD4+ T-cell-driven inflammation in EAE. However, the role of DRD5-signaling confined to CD4+ T-cells in the development of EAE is still unknown. Here, we analyzed the functional role of DRD5-signaling in CD4+ T-cell-mediated responses and its relevance in EAE by using a genetic approach. Our results show that DRD5-signaling confined to naive CD4+ T-cells exerts a pro-inflammatory effect promoting the development of EAE with a stronger disease severity. This pro-inflammatory effect observed for DRD5-signaling in naive CD4+ T-cells was related with an exacerbated proliferation in response to T-cell activation and to an increased ability to differentiate toward the Th17 inflammatory phenotype. On the other hand, quite unexpected, our results show that DRD5-signaling confined to Tregs strengthens their suppressive activity, thereby dampening the development of EAE manifestation. This anti-inflammatory effect of DRD5-signaling in Tregs was associated with a selective increase in the expression of glucocorticoid-induced tumor necrosis factor receptor-related protein (GITR), which has been described to play a critical role in the expansion of Tregs. Our findings here indicate a complex role for DRD5-signaling in CD4+ T-cells-driven responses potentiating early inflammation mediated by effector T-cells in EAE, but exacerbating suppressive activity in Tregs and thereby dampening disease manifestation in late EAE stages.
Collapse
Affiliation(s)
| | - Carolina Prado
- Laboratorio de Neuroinmunología, Fundación Ciencia & Vida, Santiago, Chile
| | | | - Rodrigo Pacheco
- Laboratorio de Neuroinmunología, Fundación Ciencia & Vida, Santiago, Chile.,Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| |
Collapse
|
53
|
Talhada D, Rabenstein M, Ruscher K. The role of dopaminergic immune cell signalling in poststroke inflammation. Ther Adv Neurol Disord 2018; 11:1756286418774225. [PMID: 29774058 PMCID: PMC5952273 DOI: 10.1177/1756286418774225] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 04/06/2018] [Indexed: 01/08/2023] Open
Abstract
Upon ischaemic stroke, brain-resident and peripheral immune cells accumulate in the central nervous system (CNS). Interestingly, these cells express pattern specific to neurotransmitter receptors and, therefore, seem to be susceptible to neurotransmitter stimulation, potentially modulating their properties and functions. One of the principal neurotransmitters in the CNS, dopamine, is involved in the regulation of processes of brain development, motor control and higher brain functions. It is constantly released in the brain and there is experimental and clinical evidence that dopaminergic signalling is involved in recovery of lost neurological function after stroke. Independent studies have revealed specific but different patterns of dopamine receptor subtypes on different populations of immune cells. Those patterns are dependent on the activation status of cells. Generally, exposure to dopamine or dopamine receptor agonists decreases detrimental actions of immune cells. In contrast, a reduction of dopaminergic inputs perpetuates a pro-inflammatory state associated with increased release of pro-inflammatory molecules. In addition, subsets of immune cells have been identified to synthesize and release dopamine, suggesting autoregulatory mechanisms. Evidence supports that inflammatory processes activated following ischaemic stroke are modulated by dopaminergic signalling.
Collapse
Affiliation(s)
- Daniela Talhada
- LUBIN Lab – Lund Brain Injury Laboratory for Neurosurgical Research, Department of Clinical Sciences, Lund University, Lund, Sweden CICS-UBI-Health Sciences Research Centre, Faculdade de Ciências da Saúde, Av. Infante D. Henrique, Universidade da Beira Interior, Portugal
| | - Monika Rabenstein
- Department of Neurology, University Hospital Cologne, Cologne, Germany
| | - Karsten Ruscher
- Lund Brain Injury Laboratory for Neurosurgical Research, Wallenberg Neuroscience Center, Lund University, BMC A13, S-22184 Lund, Sweden
| |
Collapse
|
54
|
Mackie P, Lebowitz J, Saadatpour L, Nickoloff E, Gaskill P, Khoshbouei H. The dopamine transporter: An unrecognized nexus for dysfunctional peripheral immunity and signaling in Parkinson's Disease. Brain Behav Immun 2018; 70:21-35. [PMID: 29551693 PMCID: PMC5953824 DOI: 10.1016/j.bbi.2018.03.020] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 03/13/2018] [Accepted: 03/14/2018] [Indexed: 02/06/2023] Open
Abstract
The second-most common neurodegenerative disease, Parkinson's Disease (PD) has three hallmarks: dysfunctional dopamine transmission due, at least in part, to dopamine neuron degeneration; intracellular inclusions of α-synuclein aggregates; and neuroinflammation. The origin and interplay of these features remains a puzzle, as does the underlying mechanism of PD pathogenesis and progression. When viewed in the context of neuroimmunology, dopamine also plays a role in regulating peripheral immune cells. Intriguingly, plasma dopamine levels are altered in PD, suggesting collateral dysregulation of peripheral dopamine transmission. The dopamine transporter (DAT), the main regulator of dopaminergic tone in the CNS, is known to exist in lymphocytes and monocytes/macrophages, but little is known about peripheral DAT biology or how DAT regulates the dopaminergic tone, much less how peripheral DAT alters immune function. Our review is guided by the hypothesis that dysfunctional peripheral dopamine signaling might be linked to the dysfunctional immune responses in PD and thereby suggests a potential bidirectional communication between central and peripheral dopamine systems. This review seeks to foster new perspectives concerning PD pathogenesis and progression.
Collapse
Affiliation(s)
- Phillip Mackie
- University of Florida College of Medicine, Department of Neuroscience, Gainesville, FL 32611, United States
| | - Joe Lebowitz
- University of Florida College of Medicine, Department of Neuroscience, Gainesville, FL 32611, United States
| | - Leila Saadatpour
- University of Florida College of Medicine, Department of Neuroscience, Gainesville, FL 32611, United States
| | - Emily Nickoloff
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, United States
| | - Peter Gaskill
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, United States
| | - Habibeh Khoshbouei
- University of Florida College of Medicine, Department of Neuroscience, Gainesville, FL 32611, United States.
| |
Collapse
|
55
|
Marino F, Scanzano A, Pulze L, Pinoli M, Rasini E, Luini A, Bombelli R, Legnaro M, de Eguileor M, Cosentino M. β 2 -Adrenoceptors inhibit neutrophil extracellular traps in human polymorphonuclear leukocytes. J Leukoc Biol 2018; 104:603-614. [PMID: 29668114 DOI: 10.1002/jlb.3a1017-398rr] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 03/22/2018] [Accepted: 03/22/2018] [Indexed: 12/31/2022] Open
Abstract
This study tests the hypothesis that in isolated human polymorphonuclear leukocytes (PMN) adrenergic ligands can affect neutrophil extracellular trap (NET) formation. We have previously shown that, in PMN, adrenaline (A), through the activation of adrenergic receptors (AR), reduces stimulus-dependent cell activation; we have, therefore, planned to investigate if AR are involved in NET production. PMN were obtained from venous blood of healthy subject. The ability of adrenergic ligands to affect reactive oxygen species (ROS) production, NET production, and cell migration was investigated in cells cultured under resting conditions or after activation with N-formyl-methionyl-leucyl-phenylalanine (fMLP), LPS, or IL-8. Stimuli-induced NET production measured as ROS, microscopic evaluation, and elastase production was reverted by A and this effect was blocked by the selective β2 -AR antagonist ICI-118,551. The stimulus-induced ROS generation and migration was prevented by A and by isoprenaline (ISO), and these effects were counteracted only by ICI-118,551 and not by the other two selective ligands for the β1 and β3 -AR. Finally, the presence of the β-ARs on PMN was confirmed, by means of microscopy and flow cytometry. The data of the present study suggest that adrenergic compounds, through the interaction of mainly β2 -AR, are able to affect neutrophil functions. These data are suggestive of a possible therapeutic role of β2 -AR ligands (in addition to their classical use), promoting the possible therapeutic relevance of adrenergic system in the modulation of innate immunity proposing their possible use as anti-inflammatory drugs.
Collapse
Affiliation(s)
- Franca Marino
- Center of Research in Medical Pharmacology, Varese, Italy
| | | | - Laura Pulze
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Monica Pinoli
- Center of Research in Medical Pharmacology, Varese, Italy
| | | | | | | | | | - Magda de Eguileor
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | | |
Collapse
|
56
|
Prado C, Gaiazzi M, González H, Ugalde V, Figueroa A, Osorio-Barrios FJ, López E, Lladser A, Rasini E, Marino F, Zaffaroni M, Cosentino M, Pacheco R. Dopaminergic Stimulation of Myeloid Antigen-Presenting Cells Attenuates Signal Transducer and Activator of Transcription 3-Activation Favouring the Development of Experimental Autoimmune Encephalomyelitis. Front Immunol 2018; 9:571. [PMID: 29619030 PMCID: PMC5871671 DOI: 10.3389/fimmu.2018.00571] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 03/06/2018] [Indexed: 12/19/2022] Open
Abstract
The dual potential to promote tolerance or inflammation to self-antigens makes dendritic cells (DCs) fundamental players in autoimmunity. Previous results have shown that stimulation of dopamine receptor D5 (DRD5) in DCs potentiates their inflammatory behaviour, favouring the development of experimental autoimmune encephalomyelitis (EAE). Here, we aimed to decipher the underlying mechanism and to test its relevance in multiple sclerosis (MS) patients. Our data shows that DRD5-deficiency confined to DCs in EAE mice resulted in reduced frequencies of CD4+ T-cell subsets with inflammatory potential in the central nervous system, including not only Th1 and Th17 cells but also granulocyte-macrophage colony-stimulating factor producers. Importantly, ex vivo depletion of dopamine from DCs resulted in a dramatic reduction of EAE severity, highlighting the relevance of an autocrine loop promoting inflammation in vivo. Mechanistic analyses indicated that DRD5-signalling in both mouse DCs and human monocytes involves the attenuation of signal transducer and activator of transcription 3-activation, a transcription factor that limits the production of the inflammatory cytokines interleukin (IL)-12 and IL-23. Furthermore, we found an exacerbated expression of all dopamine receptors in peripheral blood pro-inflammatory monocytes obtained from MS patients. These findings illustrate a novel mechanism by which myeloid antigen-presenting cells may trigger the onset of their inflammatory behaviour promoting the development of autoimmunity.
Collapse
Affiliation(s)
- Carolina Prado
- Laboratorio de Neuroinmunología, Fundación Ciencia and Vida, Santiago, Chile
| | - Michela Gaiazzi
- Center for Research in Medical Pharmacology, University of Insubria, Varese, Italy
| | - Hugo González
- Laboratorio de Neuroinmunología, Fundación Ciencia and Vida, Santiago, Chile
| | - Valentina Ugalde
- Laboratorio de Neuroinmunología, Fundación Ciencia and Vida, Santiago, Chile
| | - Alicia Figueroa
- Laboratorio de Neuroinmunología, Fundación Ciencia and Vida, Santiago, Chile
| | | | - Ernesto López
- Laboratorio de Inmunoterapia Génica, Fundación Ciencia and Vida, Santiago, Chile
| | - Alvaro Lladser
- Laboratorio de Inmunoterapia Génica, Fundación Ciencia and Vida, Santiago, Chile
| | - Emanuela Rasini
- Center for Research in Medical Pharmacology, University of Insubria, Varese, Italy
| | - Franca Marino
- Center for Research in Medical Pharmacology, University of Insubria, Varese, Italy
| | - Mauro Zaffaroni
- Multiple Sclerosis Centre, ASST della Valle Olona, Hospital of Gallarate, Gallarate, Italy
| | - Marco Cosentino
- Center for Research in Medical Pharmacology, University of Insubria, Varese, Italy
| | - Rodrigo Pacheco
- Laboratorio de Neuroinmunología, Fundación Ciencia and Vida, Santiago, Chile.,Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas, Universidad Andres Bello, Santiago, Chile
| |
Collapse
|
57
|
Olesen MN, Christiansen JR, Petersen SV, Jensen PH, Paslawski W, Romero-Ramos M, Sanchez-Guajardo V. CD4 T cells react to local increase of α-synuclein in a pathology-associated variant-dependent manner and modify brain microglia in absence of brain pathology. Heliyon 2018; 4:e00513. [PMID: 29560431 PMCID: PMC5857520 DOI: 10.1016/j.heliyon.2018.e00513] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Revised: 01/03/2018] [Accepted: 01/11/2018] [Indexed: 12/31/2022] Open
Abstract
We have previously shown that immunological processes in the brain during α-synuclein-induced neurodegeneration vary depending on the presence or absence of cell death. This suggests that the immune system is able to react differently to the different stages of α-synuclein pathology. However, it was unclear whether these immune changes were governed by brain processes or by a direct immune response to α-synuclein modifications. We have herein locally increased the peripheral concentration of α-synuclein or its pathology-associated variants, nitrated or fibrillar, to characterize the modulation of the CD4 T cell pool by α-synuclein and brain microglia in the absence of any α-synuclein brain pathology. We observed that α-synuclein changed the CD4:CD8 ratio by contracting the CD3+CD4+ T cell pool and reducing the pool of memory Regulatory T cells (Treg). Nitrated α-synuclein induced the expansion of both the CD3+CD4+ and CD3+CD4- T cells, while fibrils increased the percentage of Foxp3+ Treg cells and induced anti-α-synuclein antibodies. Furthermore, the activation pattern of CD3+CD4+ T cells was modulated in a variant-dependent manner; while nitrated and fibrillar α-synuclein expanded the fraction of activated Treg, all three α-synuclein variants reduced the expression levels of STAT3, CD25 and CD127 on CD3+CD4+ T cells. Additionally, while monomeric α-synuclein increased CD103 expression, the fibrils decreased it, and CCR6 expression was decreased by nitrated and fibrillar α-synuclein, indicating that α-synuclein variants affect the homing and tolerance capacities of CD3+CD4+ T cells. Indeed, this correlated with changes in brain microglia phenotype, as determined by FACS analysis, in an α-synuclein variant-specific manner and coincided in time with CD4+ T cell infiltration into brain parenchyma. We have shown that the peripheral immune system is able to sense and react specifically to changes in the local concentration and structure of α-synuclein, which results in variant-specific T cell migration into the brain. This may have a specific repercussion for brain microglia.
Collapse
Affiliation(s)
- Mads N Olesen
- Neuroimmunology of Degenerative Diseases Group, Department of Biomedicine, Aarhus University, Aarhus, Denmark.,AUideas Pilot Center NEURODIN, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Josefine R Christiansen
- Neuroimmunology of Degenerative Diseases Group, Department of Biomedicine, Aarhus University, Aarhus, Denmark.,AUideas Pilot Center NEURODIN, Department of Biomedicine, Aarhus University, Aarhus, Denmark.,CNS Disease Modeling Group, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Steen Vang Petersen
- Laboratory for Redox Regulation, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Wojciech Paslawski
- iNANO, Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Marina Romero-Ramos
- AUideas Pilot Center NEURODIN, Department of Biomedicine, Aarhus University, Aarhus, Denmark.,CNS Disease Modeling Group, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Vanesa Sanchez-Guajardo
- Neuroimmunology of Degenerative Diseases Group, Department of Biomedicine, Aarhus University, Aarhus, Denmark.,AUideas Pilot Center NEURODIN, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
58
|
Zhang X, Liu Q, Liao Q, Zhao Y. Potential Roles of Peripheral Dopamine in Tumor Immunity. J Cancer 2017; 8:2966-2973. [PMID: 28928888 PMCID: PMC5604448 DOI: 10.7150/jca.20850] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 06/29/2017] [Indexed: 12/12/2022] Open
Abstract
Recent years, immunotherapy has turned out to be a promising strategy against tumors. Peripheral dopamine (DA) has important roles in immune system among tumor patients. Accumulated reports demonstrate variable expression and distribution of DA receptors (DRs) in diverse immune cells. Interestingly, peripheral DA also involves in tumor progression and it exerts anticancer effects on immunomodulation, which includes inflammasomes in cancer, function of immune effector cells, such as T lymphocytes, myeloid-derived suppressor cells (MDSCs), tumor-associated macrophages (TAMs) and natural killer (NK) cells. Given the specific immunologic status, DA medication may be a valuable candidate in pancreatic cancer treatment. The major purpose of this review is to discuss the novel potential interactions between peripheral dopamine and tumor immunity.
Collapse
Affiliation(s)
- Xiang Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Qiaofei Liu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Quan Liao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
59
|
Pardo E, Cárcamo C, Uribe-San Martín R, Ciampi E, Segovia-Miranda F, Curkovic-Peña C, Montecino F, Holmes C, Tichauer JE, Acuña E, Osorio-Barrios F, Castro M, Cortes P, Oyanadel C, Valenzuela DM, Pacheco R, Naves R, Soza A, González A. Galectin-8 as an immunosuppressor in experimental autoimmune encephalomyelitis and a target of human early prognostic antibodies in multiple sclerosis. PLoS One 2017. [PMID: 28650992 PMCID: PMC5484466 DOI: 10.1371/journal.pone.0177472] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Galectin-8 (Gal-8) is a member of a glycan-binding protein family that regulates the immune system, among other functions, and is a target of antibodies in autoimmune disorders. However, its role in multiple sclerosis (MS), an autoimmune inflammatory disease of the central nervous system (CNS), remains unknown. We study the consequences of Gal-8 silencing on lymphocyte subpopulations and the development of experimental autoimmune encephalitis (EAE), to then assess the presence and clinical meaning of anti-Gal-8 antibodies in MS patients. Lgals8/Lac-Z knock-in mice lacking Gal-8 expression have higher polarization toward Th17 cells accompanied with decreased CCR6+ and higher CXCR3+ regulatory T cells (Tregs) frequency. These conditions result in exacerbated MOG35-55 peptide-induced EAE. Gal-8 eliminates activated Th17 but not Th1 cells by apoptosis and ameliorates EAE in C57BL/6 wild-type mice. β-gal histochemistry reflecting the activity of the Gal-8 promoter revealed Gal-8 expression in a wide range of CNS regions, including high expression in the choroid-plexus. Accordingly, we detected Gal-8 in human cerebrospinal fluid, suggesting a role in the CNS immune-surveillance circuit. In addition, we show that MS patients generate function-blocking anti-Gal-8 antibodies with pathogenic potential. Such antibodies block cell adhesion and Gal-8-induced Th17 apoptosis. Furthermore, circulating anti-Gal-8 antibodies associate with relapsing-remitting MS (RRMS), and not with progressive MS phenotypes, predicting clinical disability at diagnosis within the first year of follow-up. Our results reveal that Gal-8 has an immunosuppressive protective role against autoimmune CNS inflammation, modulating the balance of Th17 and Th1 polarization and their respective Tregs. Such a role can be counteracted during RRMS by anti-Gal-8 antibodies, worsening disease prognosis. Even though anti-Gal-8 antibodies are not specific for MS, our results suggest that they could be a potential early severity biomarker in RRMS.
Collapse
MESH Headings
- Animals
- Apoptosis/physiology
- Autoantibodies/immunology
- Brain/immunology
- Brain/metabolism
- Cell Adhesion/physiology
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Female
- Galectins/genetics
- Galectins/immunology
- Galectins/metabolism
- Gene Silencing
- Humans
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Multiple Sclerosis/genetics
- Multiple Sclerosis/immunology
- Multiple Sclerosis/metabolism
- Prognosis
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Th17 Cells/immunology
- Th17 Cells/metabolism
Collapse
Affiliation(s)
- Evelyn Pardo
- Center for Aging and Regeneration (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Anatomy and Developmental Biology Program, Institute of Biomedical Sciences, Geroscience Center for Brain Health and Metabolism, University of Chile, Santiago, Chile
| | - Claudia Cárcamo
- Departamento de Neurología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Reinaldo Uribe-San Martín
- Departamento de Neurología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ethel Ciampi
- Departamento de Neurología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Fabián Segovia-Miranda
- Center for Aging and Regeneration (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cristobal Curkovic-Peña
- Center for Aging and Regeneration (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Fabián Montecino
- Center for Aging and Regeneration (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Christopher Holmes
- Center for Aging and Regeneration (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan Enrique Tichauer
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Eric Acuña
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | | | - Marjorie Castro
- Facultad de Ciencias de la Salud, Universidad San Sebastián, Santiago, Chile
| | - Priscilla Cortes
- Center for Aging and Regeneration (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Facultad de Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Claudia Oyanadel
- Center for Aging and Regeneration (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Fundación Ciencia & Vida, Santiago, Chile
- Facultad de Ciencia, Universidad San Sebastián, Santiago, Chile
| | | | - Rodrigo Pacheco
- Fundación Ciencia & Vida, Santiago, Chile
- Facultad de Ciencias Biológicas, Departamento de Ciencias Biológicas, Universidad Andres Bello, Santiago, Chile
| | - Rodrigo Naves
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Andrea Soza
- Center for Aging and Regeneration (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Facultad de Ciencia, Universidad San Sebastián, Santiago, Chile
- * E-mail: (AG); (AS)
| | - Alfonso González
- Center for Aging and Regeneration (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Facultad de Medicina, Universidad San Sebastián, Santiago, Chile
- * E-mail: (AG); (AS)
| |
Collapse
|
60
|
Leng ZG, Lin SJ, Wu ZR, Guo YH, Cai L, Shang HB, Tang H, Xue YJ, Lou MQ, Zhao W, Le WD, Zhao WG, Zhang X, Wu ZB. Activation of DRD5 (dopamine receptor D5) inhibits tumor growth by autophagic cell death. Autophagy 2017; 13:1404-1419. [PMID: 28613975 DOI: 10.1080/15548627.2017.1328347] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Dopamine agonists such as bromocriptine and cabergoline have been successfully used in the treatment of pituitary prolactinomas and other neuroendocrine tumors. However, their therapeutic mechanisms are not fully understood. In this study we demonstrated that DRD5 (dopamine receptor D5) agonists were potent inhibitors of pituitary tumor growth. We further found that DRD5 activation increased production of reactive oxygen species (ROS), inhibited the MTOR pathway, induced macroautophagy/autophagy, and led to autophagic cell death (ACD) in vitro and in vivo. In addition, DRD5 protein was highly expressed in the majority of human pituitary adenomas, and treatment of different human pituitary tumor cell cultures with the DRD5 agonist SKF83959 resulted in growth suppression, and the efficacy was correlated with the expression levels of DRD5 in the tumors. Furthermore, we found that DRD5 was expressed in other human cancer cells such as glioblastomas, colon cancer, and gastric cancer. DRD5 activation in these cell lines suppressed their growth, inhibited MTOR activity, and induced autophagy. Finally, in vivo SKF83959 also inhibited human gastric cancer cell growth in nude mice. Our studies revealed novel mechanisms for the tumor suppressive effects of DRD5 agonists, and suggested a potential use of DRD5 agonists as a novel therapeutic approach in the treatment of different human tumors and cancers.
Collapse
Affiliation(s)
- Zhi Gen Leng
- a Department of Neurosurgery , First Affiliated Hospital of Wenzhou Medical University , Wenzhou , China
| | - Shao Jian Lin
- b Department of Neurosurgery , Ruijin Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Ze Rui Wu
- a Department of Neurosurgery , First Affiliated Hospital of Wenzhou Medical University , Wenzhou , China
| | - Yu Hang Guo
- a Department of Neurosurgery , First Affiliated Hospital of Wenzhou Medical University , Wenzhou , China
| | - Lin Cai
- a Department of Neurosurgery , First Affiliated Hospital of Wenzhou Medical University , Wenzhou , China
| | - Han Bing Shang
- b Department of Neurosurgery , Ruijin Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Hao Tang
- b Department of Neurosurgery , Ruijin Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Ya Jun Xue
- c Department of Neurosurgery, Shanghai General Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Mei Qing Lou
- c Department of Neurosurgery, Shanghai General Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Wenxiu Zhao
- e Neuroendocrine Research Laboratory , Massachusetts General Hospital and Harvard Medical School , Boston , MA , USA
| | - Wei-Dong Le
- d Center for Clinical Research on Neurological Diseases , First Affiliated Hospital of Dalian Medical University , Dalian , China
| | - Wei Guo Zhao
- b Department of Neurosurgery , Ruijin Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Xun Zhang
- e Neuroendocrine Research Laboratory , Massachusetts General Hospital and Harvard Medical School , Boston , MA , USA
| | - Zhe Bao Wu
- a Department of Neurosurgery , First Affiliated Hospital of Wenzhou Medical University , Wenzhou , China.,b Department of Neurosurgery , Ruijin Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , China
| |
Collapse
|
61
|
Pinoli M, Marino F, Cosentino M. Dopaminergic Regulation of Innate Immunity: a Review. J Neuroimmune Pharmacol 2017; 12:602-623. [PMID: 28578466 DOI: 10.1007/s11481-017-9749-2] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 04/28/2017] [Indexed: 12/13/2022]
Abstract
Dopamine (DA) is a neurotransmitter in the central nervous system as well as in peripheral tissues. Emerging evidence however points to DA also as a key transmitter between the nervous system and the immune system as well as a mediator produced and released by immune cells themselves. Dopaminergic pathways have received so far extensive attention in the adaptive branch of the immune system, where they play a role in health and disease such as multiple sclerosis, rheumatoid arthritis, cancer, and Parkinson's disease. Comparatively little is known about DA and the innate immune response, although DA may affect innate immune system cells such as dendritic cells, macrophages, microglia, and neutrophils. The present review aims at providing a complete and exhaustive summary of currently available evidence about DA and innate immunity, and to become a reference for anyone potentially interested in the fields of immunology, neurosciences and pharmacology. A wide array of dopaminergic drugs is used in therapeutics for non-immune indications, such as Parkinson's disease, hyperprolactinemia, shock, hypertension, with a usually favorable therapeutic index, and they might be relatively easily repurposed for immune-mediated disease, thus leading to innovative treatments at low price, with benefit for patients as well as for the healthcare systems.
Collapse
Affiliation(s)
- Monica Pinoli
- Center of Research in Medical Pharmacology, University of Insubria, Via Ottorino Rossi n. 9, 21100, Varese, VA, Italy
| | - Franca Marino
- Center of Research in Medical Pharmacology, University of Insubria, Via Ottorino Rossi n. 9, 21100, Varese, VA, Italy.
| | - Marco Cosentino
- Center of Research in Medical Pharmacology, University of Insubria, Via Ottorino Rossi n. 9, 21100, Varese, VA, Italy
| |
Collapse
|
62
|
Levite M, Marino F, Cosentino M. Dopamine, T cells and multiple sclerosis (MS). J Neural Transm (Vienna) 2017; 124:525-542. [DOI: 10.1007/s00702-016-1640-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 10/31/2016] [Indexed: 01/11/2023]
|
63
|
Pacheco R. Targeting dopamine receptor D3 signalling in inflammation. Oncotarget 2017; 8:7224-7225. [PMID: 28086229 PMCID: PMC5352314 DOI: 10.18632/oncotarget.14601] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 01/10/2017] [Indexed: 11/25/2022] Open
Affiliation(s)
- Rodrigo Pacheco
- Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas, Universidad Andres Bello, Santiago, Chile
- Laboratorio de Neuroinmunología, Fundación Ciencia & Vida, Ñuñoa, Santiago, Chile
| |
Collapse
|
64
|
Inhibition of dopamine receptor D3 signaling in dendritic cells increases antigen cross-presentation to CD8 + T-cells favoring anti-tumor immunity. J Neuroimmunol 2017; 303:99-107. [PMID: 28077213 DOI: 10.1016/j.jneuroim.2016.12.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 12/06/2016] [Accepted: 12/28/2016] [Indexed: 11/23/2022]
Abstract
Dendritic cells (DCs) display the unique ability for cross-presenting antigens to CD8+ T-cells, promoting their differentiation into cytotoxic T-lymphocytes (CTLs), which play a pivotal role in anti-tumor immunity. Emerging evidence points to dopamine receptor D3 (D3R) as a key regulator of immunity. Accordingly, we studied how D3R regulates DCs function in anti-tumor immunity. The results show that D3R-deficiency in DCs enhanced expansion of CTLs in vivo and induced stronger anti-tumor immunity. Co-culture experiments indicated that D3R-inhibition in DCs potentiated antigen cross-presentation and CTLs activation. Our findings suggest that D3R in DCs constitutes a new therapeutic target to strengthen anti-tumor immunity.
Collapse
|
65
|
Abstract
ABSTRACT
The aim of this review is to provide a coherent framework for understanding dendritic cells (DCs). It has seven sections. The introduction provides an overview of the immune system and essential concepts, particularly for the nonspecialist reader. Next, the “History” section outlines the early evolution of ideas about DCs and highlights some sources of confusion that still exist today. The “Lineages” section then focuses on five different populations of DCs: two subsets of “classical” DCs, plasmacytoid DCs, monocyte-derived DCs, and Langerhans cells. It highlights some cellular and molecular specializations of each, and also notes other DC subsets that have been proposed. The following “Tissues” section discusses the distribution and behavior of different DC subsets within nonlymphoid and secondary lymphoid tissues that are connected by DC migration pathways between them. In the “Tolerance” section, the role of DCs in central and peripheral tolerance is considered, including their ability to drive the differentiation of different populations of regulatory T cells. In contrast, the “Immunity” section considers the roles of DCs in sensing of infection and tissue damage, the initiation of primary responses, the T-cell effector phase, and the induction of immunological memory. The concluding section provides some speculative ideas about the evolution of DCs. It also revisits earlier concepts of generation of diversity and clonal selection in terms of DCs driving the evolution of T-cell responses. Throughout, this review highlights certain areas of uncertainty and suggests some avenues for future investigation.
Collapse
|
66
|
Immunomodulatory Effects Mediated by Dopamine. J Immunol Res 2016; 2016:3160486. [PMID: 27795960 PMCID: PMC5067323 DOI: 10.1155/2016/3160486] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 07/29/2016] [Accepted: 08/08/2016] [Indexed: 01/11/2023] Open
Abstract
Dopamine (DA), a neurotransmitter in the central nervous system (CNS), has modulatory functions at the systemic level. The peripheral and central nervous systems have independent dopaminergic system (DAS) that share mechanisms and molecular machinery. In the past century, experimental evidence has accumulated on the proteins knowledge that is involved in the synthesis, reuptake, and transportation of DA in leukocytes and the differential expression of the D1-like (D1R and D5R) and D2-like receptors (D2R, D3R, and D4R). The expression of these components depends on the state of cellular activation and the concentration and time of exposure to DA. Receptors that are expressed in leukocytes are linked to signaling pathways that are mediated by changes in cAMP concentration, which in turn triggers changes in phenotype and cellular function. According to the leukocyte lineage, the effects of DA are associated with such processes as respiratory burst, cytokine and antibody secretion, chemotaxis, apoptosis, and cytotoxicity. In clinical conditions such as schizophrenia, Parkinson disease, Tourette syndrome, and multiple sclerosis (MS), there are evident alterations during immune responses in leukocytes, in which changes in DA receptor density have been observed. Several groups have proposed that these findings are useful in establishing clinical status and clinical markers.
Collapse
|
67
|
Polcwiartek C, Vang T, Bruhn CH, Hashemi N, Rosenzweig M, Nielsen J. Diabetic ketoacidosis in patients exposed to antipsychotics: a systematic literature review and analysis of Danish adverse drug event reports. Psychopharmacology (Berl) 2016; 233:3663-3672. [PMID: 27592232 DOI: 10.1007/s00213-016-4411-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 08/11/2016] [Indexed: 12/21/2022]
Abstract
RATIONALE Patients exposed to second-generation antipsychotics (SGAs) have approximately 10 times increased risk of diabetic ketoacidosis (DKA) compared with the general population. However, as DKA is a rare complication of type 2 diabetes mellitus, and susceptible patients exposed to antipsychotics may rapidly develop DKA independently of treatment duration and weight gain, this is rather suggestive of type 1 diabetes mellitus (T1DM) or latent autoimmune diabetes in adults. OBJECTIVES We performed a systematic review of current studies regarding antipsychotic-associated DKA with type 1 etiology and analyzed Danish adverse drug event (ADE) reports (previously unpublished cases). METHODS PubMed, Embase, and the Cochrane Library were searched for all relevant studies, and the Danish Medicines Agency retrieved ADE reports using the Danish ADE database (up to date as of June 28, 2016). Diagnosis of antipsychotic-associated DKA with type 1 etiology was either considered confirmed or possible depending on authors' conclusions in the studies and/or clinical aspects. In addition, clinico-demographic risk factors were extracted. RESULTS A total of 655 records and 11 ADE reports were identified, and after screening for eligibility, we included 21 case reports/series and two ADE reports (n = 24). No relevant clinical studies were included. Although fatal cases were identified, these were excluded because of diagnostic uncertainties (n = 15). DKA occurred in 15 males (62.5 %) and nine females (37.5 %), with a mean age ± standard deviation of 34.8 ± 12.4 years. Median time to DKA was 5 months (interquartile range: 1.4-11 months). Associated antipsychotics were olanzapine (n = 9, 36 %), aripiprazole (n = 6, 24 %), risperidone (n = 6, 24 %), clozapine (n = 3, 12 %), and quetiapine (n = 1, 4 %). Nine patients (37.5 %) were confirmedly diagnosed with T1DM following DKA resolution, whereas 15 patients (62.5 %) had possible T1DM. In 22 patients (91.7 %), ongoing insulin treatment was required for glycemic control. CONCLUSIONS Increased awareness of the potential risk of antipsychotic-associated DKA and subsequent T1DM diagnosis, with insulin requirements for glycemic control, is warranted. The underlying mechanisms are poorly understood but most probably multifactorial. Certainly, further studies are warranted. Clinicians must utilize appropriate monitoring in susceptible patients and consider the possibility of continuing antipsychotic treatment with appropriate diabetic care.
Collapse
Affiliation(s)
- Christoffer Polcwiartek
- Department of Psychiatry, Aalborg University Hospital, Aalborg, Denmark.,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Torkel Vang
- Department of Psychiatry, Aalborg University Hospital, Aalborg, Denmark
| | | | - Nasseh Hashemi
- Department of Psychiatry, Aalborg University Hospital, Aalborg, Denmark.,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | | | - Jimmi Nielsen
- Department of Psychiatry, Aalborg University Hospital, Aalborg, Denmark. .,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.
| |
Collapse
|
68
|
Doñas C, Carrasco M, Fritz M, Prado C, Tejón G, Osorio-Barrios F, Manríquez V, Reyes P, Pacheco R, Bono MR, Loyola A, Rosemblatt M. The histone demethylase inhibitor GSK-J4 limits inflammation through the induction of a tolerogenic phenotype on DCs. J Autoimmun 2016; 75:105-117. [PMID: 27528513 DOI: 10.1016/j.jaut.2016.07.011] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 07/21/2016] [Accepted: 07/25/2016] [Indexed: 10/21/2022]
Abstract
As it has been established that demethylation of lysine 27 of histone H3 by the lysine-specific demethylase JMJD3 increases immune responses and thus elicits inflammation, we hypothesize that inhibition of JMJD3 may attenuate autoimmune disorders. We found that in vivo administration of GSK-J4, a selective inhibitor of JMJD3 and UTX, ameliorates the severity of experimental autoimmune encephalomyelitis (EAE). In vitro experiments revealed that the anti-inflammatory effect of GSK-J4 was exerted through an effect on dendritic cells (DCs), promoting a tolerogenic profile characterized by reduced expression of costimulatory molecules CD80/CD86, an increased expression of tolerogenic molecules CD103 and TGF-β1, and reduced secretion of proinflammatory cytokines IL-6, IFN-γ, and TNF. Adoptive transfer of GSK-J4-treated DCs into EAE mice reduced the clinical manifestation of the disease and decreased the extent of inflammatory CD4+ T cells infiltrating the central nervous system. Notably, Treg generation, stability, and suppressive activity were all exacerbated by GSK-J4-treated DCs without affecting Th1 and Th17 cell production. Our data show that GSK-J4-mediated modulation of inflammation is achieved by a direct effect on DCs and that systemic treatment with GSK-J4 or adoptive transfer of GSK-J4-treated DCs ex vivo may be promising approaches for the treatment of inflammatory and autoimmune disorders.
Collapse
Affiliation(s)
- Cristian Doñas
- Fundación Ciencia & Vida, Ñuñoa, 7780272, Santiago, Chile; Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas, Universidad Andrés Bello, 8370146, Santiago, Chile
| | - Macarena Carrasco
- Fundación Ciencia & Vida, Ñuñoa, 7780272, Santiago, Chile; Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas, Universidad Andrés Bello, 8370146, Santiago, Chile
| | - Macarena Fritz
- Fundación Ciencia & Vida, Ñuñoa, 7780272, Santiago, Chile; Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas, Universidad Andrés Bello, 8370146, Santiago, Chile
| | - Carolina Prado
- Fundación Ciencia & Vida, Ñuñoa, 7780272, Santiago, Chile
| | - Gabriela Tejón
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | | | - Valeria Manríquez
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Paz Reyes
- Fundación Ciencia & Vida, Ñuñoa, 7780272, Santiago, Chile
| | - Rodrigo Pacheco
- Fundación Ciencia & Vida, Ñuñoa, 7780272, Santiago, Chile; Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas, Universidad Andrés Bello, 8370146, Santiago, Chile
| | - María Rosa Bono
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | | | - Mario Rosemblatt
- Fundación Ciencia & Vida, Ñuñoa, 7780272, Santiago, Chile; Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile; Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas, Universidad Andrés Bello, 8370146, Santiago, Chile.
| |
Collapse
|
69
|
Liu X, Bretou M, Lennon-Duménil AM, Lemaître F, Guille-Collignon M. Indium Tin Oxide Microsystem for Electrochemical Detection of Exocytosis of Migratory Dendritic Cells. ELECTROANAL 2016. [DOI: 10.1002/elan.201600360] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Xiaoqing Liu
- Ecole normale supérieure; PSL Research University; UPMC Univ Paris 06 CNRS; Département de Chimie PASTEUR; 24, rue Lhomond 75005 Paris France
- Sorbonne Universités; UPMC Univ Paris 06, ENS, CNRS, PASTEUR; 75005 Paris France
| | - Marine Bretou
- INSERM U932; Inst Curie; 12, rue Lhomond 75005 Paris France
| | | | - Frédéric Lemaître
- Ecole normale supérieure; PSL Research University; UPMC Univ Paris 06 CNRS; Département de Chimie PASTEUR; 24, rue Lhomond 75005 Paris France
- Sorbonne Universités; UPMC Univ Paris 06, ENS, CNRS, PASTEUR; 75005 Paris France
| | - Manon Guille-Collignon
- Ecole normale supérieure; PSL Research University; UPMC Univ Paris 06 CNRS; Département de Chimie PASTEUR; 24, rue Lhomond 75005 Paris France
- Sorbonne Universités; UPMC Univ Paris 06, ENS, CNRS, PASTEUR; 75005 Paris France
| |
Collapse
|
70
|
Fernandez-Egea E, Vértes PE, Flint SM, Turner L, Mustafa S, Hatton A, Smith KGC, Lyons PA, Bullmore ET. Peripheral Immune Cell Populations Associated with Cognitive Deficits and Negative Symptoms of Treatment-Resistant Schizophrenia. PLoS One 2016; 11:e0155631. [PMID: 27244229 PMCID: PMC4887013 DOI: 10.1371/journal.pone.0155631] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Accepted: 05/01/2016] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Hypothetically, psychotic disorders could be caused or conditioned by immunological mechanisms. If so, one might expect there to be peripheral immune system phenotypes that are measurable in blood cells as biomarkers of psychotic states. METHODS We used multi-parameter flow cytometry of venous blood to quantify and determine the activation state of 73 immune cell subsets for 18 patients with chronic schizophrenia (17 treated with clozapine), and 18 healthy volunteers matched for age, sex, BMI and smoking. We used multivariate methods (partial least squares) to reduce dimensionality and define populations of differentially co-expressed cell counts in the cases compared to controls. RESULTS Schizophrenia cases had increased relative numbers of NK cells, naïve B cells, CXCR5+ memory T cells and classical monocytes; and decreased numbers of dendritic cells (DC), HLA-DR+ regulatory T-cells (Tregs), and CD4+ memory T cells. Likewise, within the patient group, more severe negative and cognitive symptoms were associated with decreased relative numbers of dendritic cells, HLA-DR+ Tregs, and CD4+ memory T cells. Motivated by the importance of central nervous system dopamine signalling for psychosis, we measured dopamine receptor gene expression in separated CD4+ cells. Expression of the dopamine D3 (DRD3) receptor was significantly increased in clozapine-treated schizophrenia and covaried significantly with differentiated T cell classes in the CD4+ lineage. CONCLUSIONS Peripheral immune cell populations and dopaminergic signalling are disrupted in clozapine-treated schizophrenia. Immuno-phenotypes may provide peripherally accessible and mechanistically specific biomarkers of residual cognitive and negative symptoms in this treatment-resistant subgroup of patients.
Collapse
Affiliation(s)
- Emilio Fernandez-Egea
- NIHR Cambridge Biomedical Research Centre, Cambridge University Hospitals NHS Foundation Trust and the University of Cambridge, Cambridge, United Kingdom
- University of Cambridge, Behavioural & Clinical Neuroscience Institute, Department of Psychiatry, Cambridge, United Kingdom
- Centro de Investigación Biomedica en Red de Salud Mental (CIBERSAM), G04, Barcelona, Spain
- Cambridgeshire & Peterborough NHS Foundation Trust, Cambridge, United Kingdom
| | - Petra E. Vértes
- University of Cambridge, Behavioural & Clinical Neuroscience Institute, Department of Psychiatry, Cambridge, United Kingdom
| | - Shaun M. Flint
- Department of Medicine and Cambridge Institute for Medical Research, University of Cambridge, School of Clinical Medicine, Cambridge, United Kingdom
| | - Lorinda Turner
- NIHR Cambridge Biomedical Research Centre, Cambridge University Hospitals NHS Foundation Trust and the University of Cambridge, Cambridge, United Kingdom
- Department of Medicine and Cambridge Institute for Medical Research, University of Cambridge, School of Clinical Medicine, Cambridge, United Kingdom
| | - Syed Mustafa
- University of Cambridge, Behavioural & Clinical Neuroscience Institute, Department of Psychiatry, Cambridge, United Kingdom
| | - Alex Hatton
- Department of Medicine and Cambridge Institute for Medical Research, University of Cambridge, School of Clinical Medicine, Cambridge, United Kingdom
| | - Kenneth G. C. Smith
- NIHR Cambridge Biomedical Research Centre, Cambridge University Hospitals NHS Foundation Trust and the University of Cambridge, Cambridge, United Kingdom
- Department of Medicine and Cambridge Institute for Medical Research, University of Cambridge, School of Clinical Medicine, Cambridge, United Kingdom
| | - Paul A. Lyons
- Department of Medicine and Cambridge Institute for Medical Research, University of Cambridge, School of Clinical Medicine, Cambridge, United Kingdom
| | - Edward T. Bullmore
- NIHR Cambridge Biomedical Research Centre, Cambridge University Hospitals NHS Foundation Trust and the University of Cambridge, Cambridge, United Kingdom
- University of Cambridge, Behavioural & Clinical Neuroscience Institute, Department of Psychiatry, Cambridge, United Kingdom
- Cambridgeshire & Peterborough NHS Foundation Trust, Cambridge, United Kingdom
- GlaxoSmithKline, ImmunoPsychiatry, Alternative Discovery & Development, Pharmaceutical R&D, Cambridge, United Kingdom
| |
Collapse
|
71
|
Ciencewicki JM, Verhein KC, Gerrish K, McCaw ZR, Li J, Bushel PR, Kleeberger SR. Effects of mannose-binding lectin on pulmonary gene expression and innate immune inflammatory response to ozone. Am J Physiol Lung Cell Mol Physiol 2016; 311:L280-91. [PMID: 27106289 DOI: 10.1152/ajplung.00205.2015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 04/20/2016] [Indexed: 02/07/2023] Open
Abstract
Ozone is a common, potent oxidant pollutant in industrialized nations. Ozone exposure causes airway hyperreactivity, lung hyperpermeability, inflammation, and cell damage in humans and laboratory animals, and exposure to ozone has been associated with exacerbation of asthma, altered lung function, and mortality. The mechanisms of ozone-induced lung injury and differential susceptibility are not fully understood. Ozone-induced lung inflammation is mediated, in part, by the innate immune system. We hypothesized that mannose-binding lectin (MBL), an innate immunity serum protein, contributes to the proinflammatory events caused by ozone-mediated activation of the innate immune system. Wild-type (Mbl(+/+)) and MBL-deficient (Mbl(-/-)) mice were exposed to ozone (0.3 ppm) for up to 72 h, and bronchoalveolar lavage fluid was examined for inflammatory markers. Mean numbers of eosinophils and neutrophils and levels of the neutrophil attractants C-X-C motif chemokines 2 [Cxcl2 (major intrinsic protein 2)] and 5 [Cxcl5 (limb expression, LIX)] in the bronchoalveolar lavage fluid were significantly lower in Mbl(-/-) than Mbl(+/+) mice exposed to ozone. Using genome-wide mRNA microarray analyses, we identified significant differences in transcript response profiles and networks at baseline [e.g., nuclear factor erythroid-related factor 2 (NRF2)-mediated oxidative stress response] and after exposure (e.g., humoral immune response) between Mbl(+/+) and Mbl(-/-) mice. The microarray data were further analyzed to discover several informative differential response patterns and subsequent gene sets, including the antimicrobial response and the inflammatory response. We also used the lists of gene transcripts to search the LINCS L1000CDS(2) data sets to identify agents that are predicted to perturb ozone-induced changes in gene transcripts and inflammation. These novel findings demonstrate that targeted deletion of Mbl caused differential levels of inflammation-related gene sets at baseline and after exposure to ozone and significantly reduced pulmonary inflammation, thus indicating an important innate immunomodulatory role of the gene in this model.
Collapse
Affiliation(s)
- Jonathan M Ciencewicki
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | - Kirsten C Verhein
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | - Kevin Gerrish
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina; and
| | - Zachary R McCaw
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | - Jianying Li
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | - Pierre R Bushel
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | - Steven R Kleeberger
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina;
| |
Collapse
|
72
|
Contreras F, Prado C, González H, Franz D, Osorio-Barrios F, Osorio F, Ugalde V, Lopez E, Elgueta D, Figueroa A, Lladser A, Pacheco R. Dopamine Receptor D3 Signaling on CD4+ T Cells Favors Th1- and Th17-Mediated Immunity. THE JOURNAL OF IMMUNOLOGY 2016; 196:4143-9. [PMID: 27183640 DOI: 10.4049/jimmunol.1502420] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 03/18/2016] [Indexed: 01/10/2023]
Abstract
Dopamine receptor D3 (DRD3) expressed on CD4(+) T cells is required to promote neuroinflammation in a murine model of Parkinson's disease. However, how DRD3 signaling affects T cell-mediated immunity remains unknown. In this study, we report that TCR stimulation on mouse CD4(+) T cells induces DRD3 expression, regardless of the lineage specification. Importantly, functional analyses performed in vivo using adoptive transfer of OVA-specific OT-II cells into wild-type recipients show that DRD3 deficiency in CD4(+) T cells results in attenuated differentiation of naive CD4(+) T cells toward the Th1 phenotype, exacerbated generation of Th2 cells, and unaltered Th17 differentiation. The reciprocal regulatory effect of DRD3 signaling in CD4(+) T cells favoring Th1 generation and impairing the acquisition of Th2 phenotype was also reproduced using in vitro approaches. Mechanistic analysis indicates that DRD3 signaling evokes suppressor of cytokine signaling 5 expression, a negative regulator of Th2 development, which indirectly favors acquisition of Th1 phenotype. Accordingly, DRD3 deficiency results in exacerbated eosinophil infiltration into the airways of mice undergoing house dust mite-induced allergic response. Interestingly, our results show that, upon chronic inflammatory colitis induced by transfer of naive CD4(+) T cells into lymphopenic recipients, DRD3 deficiency not only affects Th1 response, but also the frequency of Th17 cells, suggesting that DRD3 signaling also contributes to Th17 expansion under chronic inflammatory conditions. In conclusion, our findings indicate that DRD3-mediated signaling in CD4(+) T cells plays a crucial role in the balance of effector lineages, favoring the inflammatory potential of CD4(+) T cells.
Collapse
Affiliation(s)
- Francisco Contreras
- Laboratorio de Neuroinmunología, Fundación Ciencia y Vida, Ñuñoa 7780272, Santiago, Chile; Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Santiago 8370146, Chile
| | - Carolina Prado
- Laboratorio de Neuroinmunología, Fundación Ciencia y Vida, Ñuñoa 7780272, Santiago, Chile
| | - Hugo González
- Laboratorio de Neuroinmunología, Fundación Ciencia y Vida, Ñuñoa 7780272, Santiago, Chile
| | - Dafne Franz
- Laboratorio de Neuroinmunología, Fundación Ciencia y Vida, Ñuñoa 7780272, Santiago, Chile
| | | | - Fabiola Osorio
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; and
| | - Valentina Ugalde
- Laboratorio de Neuroinmunología, Fundación Ciencia y Vida, Ñuñoa 7780272, Santiago, Chile
| | - Ernesto Lopez
- Laboratorio de Inmunoterapia Génica, Fundación Ciencia y Vida, Ñuñoa 7780272, Santiago, Chile
| | - Daniela Elgueta
- Laboratorio de Neuroinmunología, Fundación Ciencia y Vida, Ñuñoa 7780272, Santiago, Chile; Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Santiago 8370146, Chile
| | - Alicia Figueroa
- Laboratorio de Neuroinmunología, Fundación Ciencia y Vida, Ñuñoa 7780272, Santiago, Chile
| | - Alvaro Lladser
- Laboratorio de Inmunoterapia Génica, Fundación Ciencia y Vida, Ñuñoa 7780272, Santiago, Chile
| | - Rodrigo Pacheco
- Laboratorio de Neuroinmunología, Fundación Ciencia y Vida, Ñuñoa 7780272, Santiago, Chile; Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Santiago 8370146, Chile;
| |
Collapse
|
73
|
Christiansen JR, Olesen MN, Otzen DE, Romero-Ramos M, Sanchez-Guajardo V. α-Synuclein vaccination modulates regulatory T cell activation and microglia in the absence of brain pathology. J Neuroinflammation 2016; 13:74. [PMID: 27055651 PMCID: PMC4825077 DOI: 10.1186/s12974-016-0532-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 03/18/2016] [Indexed: 12/22/2022] Open
Abstract
Background Passive and active immunization with α-synuclein has been shown to be neuroprotective in animal models of Parkinson’s disease. We have previously shown that vaccination with α-synuclein, long before α-synuclein-induced brain pathology, prevents striatal degeneration by inducing regulatory T cell infiltration in parenchyma and antibody deposition on α-synuclein overexpressing neurons. However, the effect of peripheral α-synuclein on the immune system is unknown, as are the mechanistic changes induced in the CD4 T cell population during successful neuroprotective animal studies. We have studied the changes induced by vaccination with α-synuclein in the CD4 T cell pool and its impact on brain microglia to understand the immune mechanisms behind successful vaccination strategies in Parkinson’s disease animal models. Methods Mice were immunized with WT or nitrated α-synuclein at a dose equivalent to the one used in our previous successful vaccination strategy and at a higher dose to determine potential dose-dependent effects. Animals were re-vaccinated 4 weeks after and sacrificed 5 days later. These studies were conducted in naive animals in the absence of human α-synuclein expression. Results The CD4 T cell response was modulated by α-synuclein in a dose-dependent manner, in particular the regulatory T cell population. Low-dose α-synuclein induced expansion of naive (Foxp3 + CCR6-CD127lo/neg) and dopamine receptor type D3+ regulatory T cells, as well as an increase in Stat5 protein levels. On the other hand, high dose promoted activation of regulatory T cells (Foxp3CCR6 + CD127lo/neg), which were dopamine receptor D2+D3-, and induced up-regulation of Stat5 and production of anti-α-synuclein antibodies. These effects were specific to the variant of α-synuclein used as the pathology-associated nitrated form induced distinct effects at both doses. The changes observed in the periphery after vaccination with low-dose α-synuclein correlated with an increase in CD154+, CD103+, and CD54+ microglia and the reduction of CD200R+ microglia. This resulted in the induction of a polarized tolerogenic microglia population that was CD200R-CD54CD103CD172a+ (82 % of total microglia). Conclusions We have shown for the first time the mechanisms behind α-synuclein vaccination and, importantly, how we can modulate microglia’s phenotype by regulating the CD4 T cell pool, thus shedding invaluable light on the design of neuroimmunoregulatory therapies for Parkinson’s disease. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0532-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Josefine R Christiansen
- Neuroimmunology of Degenerative Diseases group, Department of Biomedicine, HEALTH, Aarhus University, Aarhus, Denmark.,CNS Disease Modeling group, Department of Biomedicine, HEALTH , Aarhus University, Aarhus, Denmark.,AU Ideas Pilot Center NEURODIN, Department of Biomedicine, HEALTH, Aarhus University, Aarhus, Denmark
| | - Mads N Olesen
- Neuroimmunology of Degenerative Diseases group, Department of Biomedicine, HEALTH, Aarhus University, Aarhus, Denmark.,AU Ideas Pilot Center NEURODIN, Department of Biomedicine, HEALTH, Aarhus University, Aarhus, Denmark
| | - Daniel E Otzen
- Interdisciplinary Nanoscience Center - iNANO, Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Marina Romero-Ramos
- CNS Disease Modeling group, Department of Biomedicine, HEALTH , Aarhus University, Aarhus, Denmark.,AU Ideas Pilot Center NEURODIN, Department of Biomedicine, HEALTH, Aarhus University, Aarhus, Denmark
| | - Vanesa Sanchez-Guajardo
- Neuroimmunology of Degenerative Diseases group, Department of Biomedicine, HEALTH, Aarhus University, Aarhus, Denmark. .,AU Ideas Pilot Center NEURODIN, Department of Biomedicine, HEALTH, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
74
|
Pramipexole, a Dopamine D2/D3 Receptor-Preferring Agonist, Prevents Experimental Autoimmune Encephalomyelitis Development in Mice. Mol Neurobiol 2016; 54:1033-1045. [PMID: 26801190 DOI: 10.1007/s12035-016-9717-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 01/12/2016] [Indexed: 01/16/2023]
Abstract
Experimental autoimmune encephalomyelitis (EAE) is the most used animal model of multiple sclerosis (MS) for the development of new therapies. Dopamine receptors can modulate EAE and MS development, thus highlighting the potential use of dopaminergic agonists in the treatment of MS, which has been poorly explored. Herein, we hypothesized that pramipexole (PPX), a dopamine D2/D3 receptor-preferring agonist commonly used to treat Parkinson's disease (PD), would be a suitable therapeutic drug for EAE. Thus, we report the effects and the underlying mechanisms of action of PPX in the prevention of EAE. PPX (0.1 and 1 mg/kg) was administered intraperitoneally (i.p.) from day 0 to 40 post-immunization (p.i.). Our results showed that PPX 1 mg/kg prevented EAE development, abolishing EAE signs by blocking neuroinflammatory response, demyelination, and astroglial activation in spinal cord. Moreover, PPX inhibited the production of inflammatory cytokines, such as IL-17, IL-1β, and TNF-α in peripheral lymphoid tissue. PPX was also able to restore basal levels of a number of EAE-induced effects in spinal cord and striatum, such as reactive oxygen species, glutathione peroxidase, parkin, and α-synuclein (α-syn). Thus, our findings highlight the usefulness of PPX in preventing EAE-induced motor symptoms, possibly by modulating immune cell responses, such as those found in MS and other T helper cell-mediated inflammatory diseases.
Collapse
|
75
|
Levite M. Dopamine and T cells: dopamine receptors and potent effects on T cells, dopamine production in T cells, and abnormalities in the dopaminergic system in T cells in autoimmune, neurological and psychiatric diseases. Acta Physiol (Oxf) 2016; 216:42-89. [PMID: 25728499 DOI: 10.1111/apha.12476] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 10/07/2014] [Accepted: 02/23/2015] [Indexed: 12/12/2022]
Abstract
Dopamine, a principal neurotransmitter, deserves upgrading to 'NeuroImmunotransmitter' thanks to its multiple, direct and powerful effects on most/all immune cells. Dopamine by itself is a potent activator of resting effector T cells (Teffs), via two independent ways: direct Teffs activation, and indirect Teffs activation by suppression of regulatory T cells (Tregs). The review covers the following findings: (i) T cells express functional dopamine receptors (DRs) D1R-D5R, but their level and function are dynamic and context-sensitive, (ii) DR membranal protein levels do not necessarily correlate with DR mRNA levels, (iii) different T cell types/subtypes have different DR levels and composition and different responses to dopamine, (iv) autoimmune and pro-inflammatory T cells and T cell leukaemia/lymphoma also express functional DRs, (v) dopamine (~10(-8) M) activates resting/naive Teffs (CD8(+) >>>CD4(+) ), (vi) dopamine affects Th1/Th2/Th17 differentiation, (vii) dopamine inhibits already activated Teffs (i.e. T cells that have been already activated by either antigen, mitogen, anti-CD3 antibodies cytokines or other molecules), (viii) dopamine inhibits activated Tregs in an autocrine/paracrine manner. Thus, dopamine 'suppresses the suppressors' and releases the inhibition they exert on Teffs, (ix) dopamine affects intracellular signalling molecules and cascades in T cells (e.g. ERK, Lck, Fyn, NF-κB, KLF2), (x) T cells produce dopamine (Tregs>>>Teffs), can release dopamine, mainly after activation (by antigen, mitogen, anti-CD3 antibodies, PKC activators or other), uptake extracellular dopamine, and most probably need dopamine, (xi) dopamine is important for antigen-specific interactions between T cells and dendritic cells, (xii) in few autoimmune diseases (e.g. multiple sclerosis/SLE/rheumatoid arthritis), and neurological/psychiatric diseases (e.g. Parkinson disease, Alzheimer's disease, Schizophrenia and Tourette), patient's T cells seem to have abnormal DRs expression and/or responses to dopamine or production of dopamine, (xiii) drugs that affect the dopaminergic system have potent effects on T cells (e.g. dopamine=Intropin, L-dopa, bromocriptine, haloperidol, quinpirole, reserpine, pergolide, ecopipam, pimozide, amantadine, tetrabenazine, nomifensine, butaclamol). Dopamine-induced activation of resting Teffs and suppression of Tregs seem beneficial for health and may also be used for immunotherapy of cancer and infectious diseases. Independently, suppression of DRs in autoimmune and pro-inflammatory T cells, and also in cancerous T cells, may be advantageous. The review is relevant to Immunologists, Neurologists, Neuroimmunologists, Hematologists, Psychiatrists, Psychologists and Pharmacologists.
Collapse
Affiliation(s)
- M. Levite
- School of Pharmacy; Faculty of Medicine; The Hebrew University; Jerusalem Israel
- Institute of Gene Therapy; Hadassah Hebrew University Hospital; Jerusalem Israel
- School of Behavioral Sciences; Academic College of Tel-Aviv-Yaffo; Tel Aviv Israel
| |
Collapse
|
76
|
Berberine is a dopamine D1- and D2-like receptor antagonist and ameliorates experimentally induced colitis by suppressing innate and adaptive immune responses. J Neuroimmunol 2015; 289:43-55. [DOI: 10.1016/j.jneuroim.2015.10.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 09/24/2015] [Accepted: 10/05/2015] [Indexed: 12/17/2022]
|
77
|
Goldeck D, Maetzler W, Berg D, Oettinger L, Pawelec G. Altered dendritic cell subset distribution in patients with Parkinson's disease: Impact of CMV serostatus. J Neuroimmunol 2015; 290:60-5. [PMID: 26711571 DOI: 10.1016/j.jneuroim.2015.11.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 11/07/2015] [Accepted: 11/12/2015] [Indexed: 12/12/2022]
Abstract
Parkinson's disease (PD) is characterised by low-level systemic inflammation, which may be at least partly due to pathophysiological activation of immunity. Here, the frequencies of different types of circulating dendritic cells (DCs) with and without a pro-inflammatory phenotype were determined in PD patients and controls. A high proportion of older people is infected with cytomegalovirus (CMV), which acts as a chronic antigenic stressor that could also contribute to increased inflammation. Following this idea, we found higher frequencies of myeloid DCs with a pro-inflammatory CD16+ILT2(high) phenotype in CMV-positive PD patients than controls, suggesting the potential involvement of CMV in exacerbating PD.
Collapse
Affiliation(s)
- David Goldeck
- Department of Internal Medicine II, Centre for Medical Research, University of Tübingen, Tübingen Aging and Tumour Immunology Group, Waldhörnlestr. 22, 72072 Tübingen, Germany.
| | - Walter Maetzler
- Department of Neurodegeneration, Hertie Institute for Clinical Brain Research, University of Tübingen, Hoppe Seyler-Str. 3, 72076 Tübingen, Germany; German Center of Neurodegenerative Diseases, University of Tübingen, Germany
| | - Daniela Berg
- Department of Neurodegeneration, Hertie Institute for Clinical Brain Research, University of Tübingen, Hoppe Seyler-Str. 3, 72076 Tübingen, Germany; German Center of Neurodegenerative Diseases, University of Tübingen, Germany
| | - Lilly Oettinger
- Department of Internal Medicine II, Centre for Medical Research, University of Tübingen, Tübingen Aging and Tumour Immunology Group, Waldhörnlestr. 22, 72072 Tübingen, Germany
| | - Graham Pawelec
- Department of Internal Medicine II, Centre for Medical Research, University of Tübingen, Tübingen Aging and Tumour Immunology Group, Waldhörnlestr. 22, 72072 Tübingen, Germany; School of Science and Technology, Nottingham Trent University, Nottingham, UK
| |
Collapse
|
78
|
Rangel-Barajas C, Coronel I, Florán B. Dopamine Receptors and Neurodegeneration. Aging Dis 2015; 6:349-68. [PMID: 26425390 DOI: 10.14336/ad.2015.0330] [Citation(s) in RCA: 149] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Accepted: 03/30/2015] [Indexed: 01/19/2023] Open
Abstract
Dopamine (DA) is one of the major neurotransmitters and participates in a number of functions such as motor coordination, emotions, memory, reward mechanism, neuroendocrine regulation etc. DA exerts its effects through five DA receptors that are subdivided in 2 families: D1-like DA receptors (D1 and D5) and the D2-like (D2, D3 and D4). All DA receptors are widely expressed in the central nervous system (CNS) and play an important role in not only in physiological conditions but also pathological scenarios. Abnormalities in the DAergic system and its receptors in the basal ganglia structures are the basis Parkinson's disease (PD), however DA also participates in other neurodegenerative disorders such as Huntington disease (HD) and multiple sclerosis (MS). Under pathological conditions reorganization of DAergic system has been observed and most of the times, those changes occur as a mechanism of compensation, but in some cases contributes to worsening the alterations. Here we review the changes that occur on DA transmission and DA receptors (DARs) at both levels expression and signals transduction pathways as a result of neurotoxicity, inflammation and in neurodegenerative processes. The better understanding of the role of DA receptors in neuropathological conditions is crucial for development of novel therapeutic approaches to treat alterations related to neurodegenerative diseases.
Collapse
Affiliation(s)
- Claudia Rangel-Barajas
- 1Department of Psychological and Brain Sciences Program in Neurosciences, Indiana University Bloomington, Bloomington, IN 47405, USA ; 2Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Israel Coronel
- 3Health Sciences Faculty, Anahuac University, Mexico Norte, State of Mexico, Mexico
| | - Benjamín Florán
- 4Department of Physiology, Biophysics and Neurosciences CINVESTAV-IPN, Mexico
| |
Collapse
|
79
|
Cosentino M, Ferrari M, Kustrimovic N, Rasini E, Marino F. Influence of dopamine receptor gene polymorphisms on circulating T lymphocytes: A pilot study in healthy subjects. Hum Immunol 2015; 76:747-52. [PMID: 26429319 DOI: 10.1016/j.humimm.2015.09.032] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 08/24/2015] [Accepted: 09/27/2015] [Indexed: 01/11/2023]
Abstract
Dopamine is a key transmitter in the neuroimmune network, acting through five dopaminergic receptors (DR): the D1-like D1 and D5 and the D2-like D2, D3 and D4. Several DR gene variants exist and may affect DR expression and activity. We assessed total lymphocytes, CD3+, CD4+ and CD8+ T lymphocytes in peripheral blood of healthy subjects and their association with selected DR gene variants (DRD1 rs4532 and rs686, DRD5 rs6283, DRD2 rs1800497 and rs6277, DRD3 rs6280 and rs1800828, DRD4 rs747302 and 7 48-base pair VNTR). DRD1 rs4532 and rs686 and DRD5 rs6283 were associated with total lymphocytes, and with CD3+ and CD4+ (but not CD8+) T lymphocytes, while none of the D2-like DR gene variants showed any association with lymphocyte counts. An arbitrary score based on the activity of D1-like vs D2-like DR correlated with total lymphocytes, CD3+ and CD4+ T cells (but not with CD8+ T cells). The association between D1-like DR gene variants and lymphocyte count, and in particular with CD4+ (but not CD8+) T lymphocytes, may imply a functional prevalence of D1-like over D2-like DR in CD4+ T cells. This is the first study showing an influence of DR gene polymorphisms on lymphocyte count, and in particular on CD4+ T cells. Future studies should address the possible association between DR gene variants and the immune function in health and disease. The relevance of these findings for the immune effects of dopaminergic agents should be also carefully examined.
Collapse
Affiliation(s)
- Marco Cosentino
- Center for Research in Medical Pharmacology, University of Insubria, Varese, Italy.
| | - Marco Ferrari
- Center for Research in Medical Pharmacology, University of Insubria, Varese, Italy
| | - Natasa Kustrimovic
- Center for Research in Medical Pharmacology, University of Insubria, Varese, Italy
| | - Emanuela Rasini
- Center for Research in Medical Pharmacology, University of Insubria, Varese, Italy
| | - Franca Marino
- Center for Research in Medical Pharmacology, University of Insubria, Varese, Italy
| |
Collapse
|
80
|
Abstract
The variety of physiological functions controlled by dopamine in the brain and periphery is mediated by the D1, D2, D3, D4 and D5 dopamine GPCRs. Drugs acting on dopamine receptors are significant tools for the management of several neuropsychiatric disorders including schizophrenia, bipolar disorder, depression and Parkinson's disease. Recent investigations of dopamine receptor signalling have shown that dopamine receptors, apart from their canonical action on cAMP-mediated signalling, can regulate a myriad of cellular responses to fine-tune the expression of dopamine-associated behaviours and functions. Such signalling mechanisms may involve alternate G protein coupling or non-G protein mechanisms involving ion channels, receptor tyrosine kinases or proteins such as β-arrestins that are classically involved in GPCR desensitization. Another level of complexity is the growing appreciation of the physiological roles played by dopamine receptor heteromers. Applications of new in vivo techniques have significantly furthered the understanding of the physiological functions played by dopamine receptors. Here we provide an update of the current knowledge regarding the complex biology, signalling, physiology and pharmacology of dopamine receptors.
Collapse
|
81
|
Wu SF, Xu G, Stanley D, Huang J, Ye GY. Dopamine modulates hemocyte phagocytosis via a D1-like receptor in the rice stem borer, Chilo suppressalis. Sci Rep 2015; 5:12247. [PMID: 26179416 PMCID: PMC4503959 DOI: 10.1038/srep12247] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 06/19/2015] [Indexed: 12/14/2022] Open
Abstract
Dopamine (DA) is a signal moiety bridging the nervous and immune systems. DA dysregulation is linked to serious human diseases, including addiction, schizophrenia, and Parkinson's disease. However, DA actions in the immune system remain incompletely understood. In this study, we found that DA modulates insect hemocyte phagocytosis using hemocytes prepared from the rice stem borer (RSB), Chilo suppressalis. We investigated whether insect hemocytes are capable of de novo DA production. Here we show that exposing hemocytes to lipopolysaccharide (LPS) led to induction of DA-generating enzymes. Exogenous DA induced rapid phosphorylation of extracellular signal-regulated kinase (ERK) in naïve hemocytes. Activation of ERK was inhibited by preincubating with a DOP1 receptor antagonist. Thus, DA signaling via the DOP1 receptor may contribute to early hemocyte activation. DA synthesized and released from hemocytes may act in an autocrine mechanism to stimulate or maintain phagocytic activity. Consistent with this hypothesis, we found that inhibition of DA synthesis with α-methyl-DL-tyrosine methyl ester hydrochloride or blockage of DOP1 receptor with antagonist SCH23390 impaired hemocyte phagocytosis. Topical DA application also significantly decreased RSB mortality following challenge with the insect pathogenic fungus, Beauveria bassiana. We infer that a DA-dependent signaling system operates in hemocytes to mediate phagocytotic functions.
Collapse
Affiliation(s)
- Shun-Fan Wu
- 1] State Key Laboratory of Rice Biology &Key Laboratory of Agricultural Entomology of Ministry of Agriculture, Institute of Insect Sciences, Zhejiang University, Hangzhou 310058, China [2] College of Plant Protection, Nanjing Agricultural University, Nanjing 210095, China; State &Local Joint Engineering Research Center of Green Pesticide Invention and Application
| | - Gang Xu
- State Key Laboratory of Rice Biology &Key Laboratory of Agricultural Entomology of Ministry of Agriculture, Institute of Insect Sciences, Zhejiang University, Hangzhou 310058, China
| | - David Stanley
- USDA - Agricultural Research Service, BCIRL, 1503 S. Providence Road, Columbia MO 65203 USA
| | - Jia Huang
- State Key Laboratory of Rice Biology &Key Laboratory of Agricultural Entomology of Ministry of Agriculture, Institute of Insect Sciences, Zhejiang University, Hangzhou 310058, China
| | - Gong-Yin Ye
- State Key Laboratory of Rice Biology &Key Laboratory of Agricultural Entomology of Ministry of Agriculture, Institute of Insect Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
82
|
Adaptive Immunity in Schizophrenia: Functional Implications of T Cells in the Etiology, Course and Treatment. J Neuroimmune Pharmacol 2015; 10:610-9. [PMID: 26162591 DOI: 10.1007/s11481-015-9626-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 07/03/2015] [Indexed: 12/21/2022]
Abstract
Schizophrenia is a severe and highly complex neurodevelopmental disorder with an unknown etiopathology. Recently, immunopathogenesis has emerged as one of the most compelling etiological models of schizophrenia. Over the past few years considerable research has been devoted to the role of innate immune responses in schizophrenia. The findings of such studies have helped to conceptualize schizophrenia as a chronic low-grade inflammatory disorder. Although the contribution of adaptive immune responses has also been emphasized, however, the precise role of T cells in the underlying neurobiological pathways of schizophrenia is yet to be ascertained comprehensively. T cells have the ability to infiltrate brain and mediate neuro-immune cross-talk. Conversely, the central nervous system and the neurotransmitters are capable of regulating the immune system. Neurotransmitter like dopamine, implicated widely in schizophrenia risk and progression can modulate the proliferation, trafficking and functions of T cells. Within brain, T cells activate microglia, induce production of pro-inflammatory cytokines as well as reactive oxygen species and subsequently lead to neuroinflammation. Importantly, such processes contribute to neuronal injury/death and are gradually being implicated as mediators of neuroprogressive changes in schizophrenia. Antipsychotic drugs, commonly used to treat schizophrenia are also known to affect adaptive immune system; interfere with the differentiation and functions of T cells. This understanding suggests a pivotal role of T cells in the etiology, course and treatment of schizophrenia and forms the basis of this review.
Collapse
|
83
|
Franz D, Contreras F, González H, Prado C, Elgueta D, Figueroa C, Pacheco R. Dopamine receptors D3 and D5 regulate CD4(+)T-cell activation and differentiation by modulating ERK activation and cAMP production. J Neuroimmunol 2015; 284:18-29. [PMID: 26025054 DOI: 10.1016/j.jneuroim.2015.05.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 04/29/2015] [Accepted: 05/01/2015] [Indexed: 01/11/2023]
Abstract
Dopamine receptors have been described in T-cells, however their signalling pathways coupled remain unknown. Since cAMP and ERKs play key roles regulating T-cell physiology, we aim to determine whether cAMP and ERK1/2-phosphorylation are modulated by dopamine receptor 3 (D3R) and D5R, and how this modulation affects CD4(+) T-cell activation and differentiation. Our pharmacologic and genetic evidence shows that D3R-stimulation reduced cAMP levels and ERK2-phosphorylation, consequently increasing CD4(+) T-cell activation and Th1-differentiation, respectively. Moreover, D5R expression reinforced TCR-triggered ERK1/2-phosphorylation and T-cell activation. In conclusion, these findings demonstrate how D3R and D5R modulate key signalling pathways affecting CD4(+) T-cell activation and Th1-differentiation.
Collapse
Affiliation(s)
- Dafne Franz
- Laboratorio de Neuroinmunología, Fundación Ciencia & Vida, Ñuñoa, 7780272 Santiago, Chile
| | - Francisco Contreras
- Laboratorio de Neuroinmunología, Fundación Ciencia & Vida, Ñuñoa, 7780272 Santiago, Chile
| | - Hugo González
- Laboratorio de Neuroinmunología, Fundación Ciencia & Vida, Ñuñoa, 7780272 Santiago, Chile
| | - Carolina Prado
- Laboratorio de Neuroinmunología, Fundación Ciencia & Vida, Ñuñoa, 7780272 Santiago, Chile
| | - Daniela Elgueta
- Laboratorio de Neuroinmunología, Fundación Ciencia & Vida, Ñuñoa, 7780272 Santiago, Chile; Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas, Universidad Andres Bello, 8370146 Santiago, Chile
| | - Claudio Figueroa
- Departamento de Ciencias Biológicas y Químicas, Facultad de Ciencia, Universidad San Sebastián, Providencia, 7510157 Santiago, Chile
| | - Rodrigo Pacheco
- Laboratorio de Neuroinmunología, Fundación Ciencia & Vida, Ñuñoa, 7780272 Santiago, Chile; Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas, Universidad Andres Bello, 8370146 Santiago, Chile.
| |
Collapse
|
84
|
Mbongue JC, Nicholas DA, Zhang K, Kim NS, Hamilton BN, Larios M, Zhang G, Umezawa K, Firek AF, Langridge WHR. Induction of indoleamine 2, 3-dioxygenase in human dendritic cells by a cholera toxin B subunit-proinsulin vaccine. PLoS One 2015; 10:e0118562. [PMID: 25714914 PMCID: PMC4340906 DOI: 10.1371/journal.pone.0118562] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 01/20/2015] [Indexed: 12/28/2022] Open
Abstract
Dendritic cells (DC) interact with naïve T cells to regulate the delicate balance between immunity and tolerance required to maintain immunological homeostasis. In this study, immature human dendritic cells (iDC) were inoculated with a chimeric fusion protein vaccine containing the pancreatic β-cell auto-antigen proinsulin linked to a mucosal adjuvant the cholera toxin B subunit (CTB-INS). Proteomic analysis of vaccine inoculated DCs revealed strong up-regulation of the tryptophan catabolic enzyme indoleamine 2, 3-dioxygenase (IDO1). Increased biosynthesis of the immunosuppressive enzyme was detected in DCs inoculated with the CTB-INS fusion protein but not in DCs inoculated with proinsulin, CTB, or an unlinked combination of the two proteins. Immunoblot and PCR analyses of vaccine treated DCs detected IDO1mRNA by 3 hours and IDO1 protein synthesis by 6 hours after vaccine inoculation. Determination of IDO1 activity in vaccinated DCs by measurement of tryptophan degradation products (kynurenines) showed increased tryptophan cleavage into N-formyl kynurenine. Vaccination did not interfere with monocytes differentiation into DC, suggesting the vaccine can function safely in the human immune system. Treatment of vaccinated DCs with pharmacological NF-κB inhibitors ACHP or DHMEQ significantly inhibited IDO1 biosynthesis, suggesting a role for NF-κB signaling in vaccine up-regulation of dendritic cell IDO1. Heat map analysis of the proteomic data revealed an overall down-regulation of vaccinated DC functions, suggesting vaccine suppression of DC maturation. Together, our experimental data indicate that CTB-INS vaccine induction of IDO1 biosynthesis in human DCs may result in the inhibition of DC maturation generating a durable state of immunological tolerance. Understanding how CTB-INS modulates IDO1 activity in human DCs will facilitate vaccine efficacy and safety, moving this immunosuppressive strategy closer to clinical applications for prevention of type 1 diabetes autoimmunity.
Collapse
Affiliation(s)
- Jacques C. Mbongue
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
- Loma Linda University School of Medicine, Department of Basic Sciences, Division of Physiology, Loma Linda, CA, United States of America
| | - Dequina A. Nicholas
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
- Mass Spectrometer Core Facility, Department of Biochemistry, Loma Linda University School of Medicine, Loma Linda University School of Medicine, Department of Basic Sciences, Loma Linda, CA, United States of America
| | - Kangling Zhang
- Mass Spectrometer Core Facility, Department of Biochemistry, Loma Linda University School of Medicine, Loma Linda University School of Medicine, Department of Basic Sciences, Loma Linda, CA, United States of America
- Department of Pharmacology and Toxicology, School of Medicine, University of Texas Medical Branch, Galveston, TX, United States of America
| | - Nan-Sun Kim
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
- Mass Spectrometer Core Facility, Department of Biochemistry, Loma Linda University School of Medicine, Loma Linda University School of Medicine, Department of Basic Sciences, Loma Linda, CA, United States of America
- Department of Molecular Biology, Chonbuk National University, Jeon-Ju, Republic of Korea
| | - Brittany N. Hamilton
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
- Loma Linda University School of Medicine, Department of Basic Sciences, Division of Microbiology and Molecular Genetics, Loma Linda, CA, United States of America
| | - Marco Larios
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
| | - Guangyu Zhang
- Mass Spectrometer Core Facility, Department of Biochemistry, Loma Linda University School of Medicine, Loma Linda University School of Medicine, Department of Basic Sciences, Loma Linda, CA, United States of America
| | - Kazuo Umezawa
- Aichi Medical University, School of Medicine, Department of Molecular Target Medicine Screening, Nagakute, Aichi, Japan
| | - Anthony F. Firek
- Endocrinology Section, JL Pettis Memorial VA Medical Center, Loma Linda, CA, United States of America
| | - William H. R. Langridge
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
- Mass Spectrometer Core Facility, Department of Biochemistry, Loma Linda University School of Medicine, Loma Linda University School of Medicine, Department of Basic Sciences, Loma Linda, CA, United States of America
- * E-mail:
| |
Collapse
|
85
|
Alpaerts K, Buckinx R, Adriaensen D, Van Nassauw L, Timmermans JP. Identification and Putative Roles of Distinct Subtypes of Intestinal Dendritic Cells in Neuroimmune Communication: What can be Learned from Other Organ Systems? Anat Rec (Hoboken) 2015; 298:903-16. [DOI: 10.1002/ar.23106] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Revised: 09/13/2014] [Accepted: 11/08/2014] [Indexed: 12/13/2022]
Affiliation(s)
- Katrien Alpaerts
- Laboratory of Cell biology and Histology; Department of Veterinary Sciences; University of Antwerp; Antwerp Belgium
| | - Roeland Buckinx
- Laboratory of Cell biology and Histology; Department of Veterinary Sciences; University of Antwerp; Antwerp Belgium
| | - Dirk Adriaensen
- Laboratory of Cell biology and Histology; Department of Veterinary Sciences; University of Antwerp; Antwerp Belgium
| | - Luc Van Nassauw
- Laboratory of Human Anatomy and Embryology; Faculty of Medicine and Health Sciences; University of Antwerp; Antwerp Belgium
| | - Jean-Pierre Timmermans
- Laboratory of Cell biology and Histology; Department of Veterinary Sciences; University of Antwerp; Antwerp Belgium
| |
Collapse
|
86
|
Cytokine Serum Levels as Potential Biological Markers for the Psychopathology in Schizophrenia. ACTA ACUST UNITED AC 2014. [DOI: 10.1155/2014/493505] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
We discuss the role of immune system disturbance in schizophrenia and especially changes of serum levels of cytokines in patients with schizophrenia. The cytokines are essential to wide range of functions related to the defense of the organisms from infectious and environmental dangers. However it is not known whether cytokines influence the presentation of psychotic symptoms. Identification of changes in the serum level of certain cytokines and their correlation with distinct psychopathological symptoms may facilitate the identification of subgroups of patients who are likely to benefit from immunotherapy or anti-inflammatory therapy. Such patients may benefit from tailored immunotherapy designed for modulation of abnormal cytokine levels related to specific positive or negative symptoms of schizophrenia.
Collapse
|
87
|
González H, Pacheco R. T-cell-mediated regulation of neuroinflammation involved in neurodegenerative diseases. J Neuroinflammation 2014; 11:201. [PMID: 25441979 PMCID: PMC4258012 DOI: 10.1186/s12974-014-0201-8] [Citation(s) in RCA: 164] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 11/12/2014] [Indexed: 12/25/2022] Open
Abstract
Neuroinflammation is involved in several neurodegenerative disorders and emerging evidence indicates that it constitutes a critical process that is required for the progression of neurodegeneration. Microglial activation constitutes a central event in neuroinflammation. Furthermore, microglia can not only be activated with an inflammatory and neurotoxic phenotype (M1-like phenotype), but they also can acquire a neurosupportive functional phenotype (M2-like phenotype) characterised by the production of anti-inflammatory mediators and neurotrophic factors. Importantly, during the past decade, several studies have shown that CD4+ T-cells infiltrate the central nervous system (CNS) in many neurodegenerative disorders, in which their participation has a critical influence on the outcome of microglial activation and consequent neurodegeneration. In this review, we focus on the analysis of the interplay of the different sub-populations of CD4+ T-cells infiltrating the CNS and how they participate in regulating the outcome of neuroinflammation and neurodegeneration in the context of Parkinson’s disease, Alzheimer’s disease, amyotrophic lateral sclerosis and multiple sclerosis. In this regard, encephalitogenic inflammatory CD4+ T-cells, such as Th1, Th17, GM-CSF-producer CD4+ T-cells and γδT-cells, strongly contribute to chronic neuroinflammation, thus perpetuating neurodegenerative processes. In contrast, encephalitogenic or meningeal Tregs and Th2 cells decrease inflammatory functions in microglial cells and promote a neurosupportive microenvironment. Moreover, whereas some neurodegenerative disorders such as multiple sclerosis, Parkinson’s disease and Alzheimer’s disease involve the participation of inflammatory CD4+ T-cells 'naturally', the physiopathology of other neurodegenerative diseases, such as amyotrophic lateral sclerosis, is associated with the participation of anti-inflammatory CD4+ T-cells that delay the neurodegenerative process. Thus, current evidence supports the hypothesis that the involvement of CD4+ T-cells against CNS antigens constitutes a key component in regulating the progression of the neurodegenerative process.
Collapse
|
88
|
Debnath M, Berk M. Th17 pathway-mediated immunopathogenesis of schizophrenia: mechanisms and implications. Schizophr Bull 2014; 40:1412-21. [PMID: 24711545 PMCID: PMC4193719 DOI: 10.1093/schbul/sbu049] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Schizophrenia is a highly complex and severe neuropsychiatric disorder with an unknown etiopathology. Evidence for a dysregulated immune system in both the risk for and progression of schizophrenia has recently been overwhelming. Importantly, chronic low-grade inflammation both in the periphery and central nervous system has been shown to contribute predominantly to the pathogenesis of schizophrenia in a subset of individuals. Inflammation in the central nervous system is mediated by a range of proinflammatory cytokines, resident immune cells such as microglia, and brain infiltrating peripheral immunocompetent cells, such as T lymphocytes. Recently, Th17 cells, a subset of T helper cells have emerged as crucial players in mucosal defense against infections. It is linked to atopic, inflammatory, and autoimmune disorders. The risk factors/mechanisms leading to low-grade inflammation in schizophrenia are diverse and include infectious agents, stress, trauma, environmental toxins, genetic vulnerability, physical inactivity, obesity, poor diet, and sleep disruption. Herein, we propose that fetal programming of cellular immune components driven by intrauterine adversity can lead to the generation of long-lasting effector/memory Th17 cells. Th17 cells can disrupt the blood-brain barrier, infiltrate the central nervous system, and, along with other cytokines and microglia, lead to neuroprogression through neuroinflammation in schizophrenia.
Collapse
Affiliation(s)
- Monojit Debnath
- Department of Human Genetics, National Institute of Mental Health & Neurosciences, Bangalore, Karnataka, India;
| | - Michael Berk
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, Victoria, Australia;,Department of Psychiatry, Florey Institute of Neuroscience and Mental Health, Orygen Youth Health Research Centre, University of Melbourne, Parkville, Australia
| |
Collapse
|
89
|
Ferreira TB, Barros PO, Teixeira B, Cassano T, Centurião N, Kasahara TM, Hygino J, Vasconcelos CCF, Filho HA, Alvarenga R, Wing AC, Andrade RM, Andrade AF, Bento CAM. Dopamine favors expansion of glucocorticoid-resistant IL-17-producing T cells in multiple sclerosis. Brain Behav Immun 2014; 41:182-90. [PMID: 24882215 DOI: 10.1016/j.bbi.2014.05.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2014] [Revised: 05/14/2014] [Accepted: 05/21/2014] [Indexed: 01/10/2023] Open
Abstract
Dopamine (DA) is a neurotransmitter produced mainly in the central nervous system (CNS) that has immunomodulatory actions on T cells. As the multiple sclerosis (MS) has long been regarded as an autoimmune disease of CNS mediated by T cells, the objective of this study was to evaluate the impact of DA on in vitro functional status of T cells from relapsing-remitting (RR)-MS patients. Peripheral T-cells from RR-MS patients were activated by mitogens and cell proliferation and cytokine production were assayed by [(3)H]-thymidine uptake and ELISA, respectively. Our results demonstrated that DA enhanced in vitro T cell proliferation and Th17-related cytokines in MS-derived cell cultures. In addition, this catecholamine reduced Treg-related cytokines (IL-10 and TGF-β) release by activated CD4(+) T cells. These DA-induced effects on T cells were mainly dependent on IL-6 production by both polyclonally-activated CD4(+) T cells and LPS-stimulated monocytes. Furthermore, the production of IL-17 and IL-6 by MS-derived T cells was directly related with neurological disability (EDSS score), and the release of these cytokines was less sensitive to glucocorticoid inhibition in MS patients than in control group, mainly after DA addition. In conclusion, our data suggest that DA amplifies glucocorticoid-resistant Th17 phenotype in MS patients, and this phenomenon could be, at least in part, due to its ability to induce IL-6 production by monocytes and CD4(+) T cells.
Collapse
Affiliation(s)
- Thais B Ferreira
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Priscila O Barros
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bruna Teixeira
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tatiane Cassano
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Newton Centurião
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Taissa M Kasahara
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Joana Hygino
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Helcio Alvarenga Filho
- Post-graduate Program in Neurology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Regina Alvarenga
- Post-graduate Program in Neurology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Cristina Wing
- Post-graduate Program in Neurology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Regis M Andrade
- Department of General Medicine, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Arnaldo F Andrade
- Department of Microbiology, Immunology and Parasitology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cleonice A M Bento
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil; Post-graduate Program in Neurology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
90
|
Sundman E, Olofsson PS. Neural control of the immune system. ADVANCES IN PHYSIOLOGY EDUCATION 2014; 38:135-139. [PMID: 25039084 PMCID: PMC4056170 DOI: 10.1152/advan.00094.2013] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 03/25/2014] [Indexed: 06/02/2023]
Abstract
Neural reflexes support homeostasis by modulating the function of organ systems. Recent advances in neuroscience and immunology have revealed that neural reflexes also regulate the immune system. Activation of the vagus nerve modulates leukocyte cytokine production and alleviates experimental shock and autoimmune disease, and recent data have suggested that vagus nerve stimulation can improve symptoms in human rheumatoid arthritis. These discoveries have generated an increased interest in bioelectronic medicine, i.e., therapeutic delivery of electrical impulses that activate nerves to regulate immune system function. Here, we discuss the physiology and potential therapeutic implications of neural immune control.
Collapse
|
91
|
Abstract
The initiation and perpetuation of autoimmunity recognize numerous checkpoints, from the genomic susceptibility to the breakdown of tolerance. This latter phenomenon includes the loss of B cell anergy and T regulatory cell failure, as well as the production of autoantibodies and autoreactive T cells. These mechanisms ultimately lead to tissue injury via different mechanisms that span from the production of proinflammatory cytokines to the chemotaxis of immune cells to the target sites. The pathways to autoimmunity have been widely investigated over the past year and resulted in a number of articles in peer-reviewed journals that has increased by nearly 10 % compared to 2011. We herein follow on the attempt to provide a brief discussion of the majority of articles on autoimmune diseases that were published in the major immunology journals in the previous solar year. The selection is necessarily arbitrary and may thus not be seen as comprehensive but reflects current research trends. Indeed, 2012 articles were mostly dedicated to define new and old mechanisms with potential therapeutic implications in autoimmunity in general, though based on specific clinical conditions or animal models. As paradigmatic examples, the environmental influence on autoimmunity, Th17 changes modulating the autoimmune response, serum autoantibodies and B cell changes as biomarkers and therapeutic targets were major issues addressed by experimental articles in 2012. Further, a growing number of studies investigated the sex bias of autoimmunity and supported different working hypotheses to explain the female predominance, including sex chromosome changes and reproductive life factors. In conclusion, the resulting scenario illustrates that common factors may underlie different autoimmune diseases and this is well represented by the observed alterations in interferon-α and TGFβ or by the shared signaling pathways.
Collapse
Affiliation(s)
- Carlo Selmi
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy,
| |
Collapse
|
92
|
Romero-Ramos M, von Euler Chelpin M, Sanchez-Guajardo V. Vaccination strategies for Parkinson disease: induction of a swift attack or raising tolerance? Hum Vaccin Immunother 2014; 10:852-67. [PMID: 24670306 DOI: 10.4161/hv.28578] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Parkinson disease is the second most common neurodegenerative disease in the world, but there is currently no available cure for it. Current treatments only alleviate some of the symptoms for a few years, but they become ineffective in the long run and do not stop the disease. Therefore it is of outmost importance to develop therapeutic strategies that can prevent, stop, or cure Parkinson disease. A very promising target for these therapies is the peripheral immune system due to its probable involvement in the disease and its potential as a tool to modulate neuroinflammation. But for such strategies to be successful, we need to understand the particular state of the peripheral immune system during Parkinson disease in order to avoid its weaknesses. In this review we examine the available data regarding how dopamine regulates the peripheral immune system and how this regulation is affected in Parkinson disease; the specific cytokine profiles observed during disease progression and the alterations documented to date in patients' peripheral blood mononuclear cells. We also review the different strategies used in Parkinson disease animal models to modulate the adaptive immune response to salvage dopaminergic neurons from cell death. After analyzing the evidence, we hypothesize the need to prime the immune system to restore natural tolerance against α-synuclein in Parkinson disease, including at the same time B and T cells, so that T cells can reprogram microglia activation to a beneficial pattern and B cell/IgG can help neurons cope with the pathological forms of α-synuclein.
Collapse
Affiliation(s)
- Marina Romero-Ramos
- CNS disease modeling group; Department of Biomedicine; Aarhus University; Aarhus, Denmark; NEURODIN; Department of Biomedicine; Aarhus University; Aarhus, Denmark
| | - Marianne von Euler Chelpin
- CNS disease modeling group; Department of Biomedicine; Aarhus University; Aarhus, Denmark; NEURODIN; Department of Biomedicine; Aarhus University; Aarhus, Denmark; Neuroimmunology of Degenerative Diseases group; Department of Biomedicine; Aarhus University; Aarhus, Denmark
| | - Vanesa Sanchez-Guajardo
- NEURODIN; Department of Biomedicine; Aarhus University; Aarhus, Denmark; Neuroimmunology of Degenerative Diseases group; Department of Biomedicine; Aarhus University; Aarhus, Denmark
| |
Collapse
|
93
|
Pacheco R, Contreras F, Zouali M. The dopaminergic system in autoimmune diseases. Front Immunol 2014; 5:117. [PMID: 24711809 PMCID: PMC3968755 DOI: 10.3389/fimmu.2014.00117] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 03/05/2014] [Indexed: 01/02/2023] Open
Abstract
Bidirectional interactions between the immune and the nervous systems are of considerable interest both for deciphering their functioning and for designing novel therapeutic strategies. The past decade has brought a burst of insights into the molecular mechanisms involved in neuroimmune communications mediated by dopamine. Studies of dendritic cells (DCs) revealed that they express the whole machinery to synthesize and store dopamine, which may act in an autocrine manner to stimulate dopamine receptors (DARs). Depending on specific DARs stimulated on DCs and T cells, dopamine may differentially favor CD4+ T cell differentiation into Th1 or Th17 inflammatory cells. Regulatory T cells can also release high amounts of dopamine that acts in an autocrine DAR-mediated manner to inhibit their suppressive activity. These dopaminergic regulations could represent a driving force during autoimmunity. Indeed, dopamine levels are altered in the brain of mouse models of multiple sclerosis (MS) and lupus, and in inflamed tissues of patients with inflammatory bowel diseases or rheumatoid arthritis (RA). The distorted expression of DARs in peripheral lymphocytes of lupus and MS patients also supports the importance of dopaminergic regulations in autoimmunity. Moreover, dopamine analogs had beneficial therapeutic effects in animal models, and in patients with lupus or RA. We propose models that may underlie key roles of dopamine and its receptors in autoimmune diseases.
Collapse
Affiliation(s)
- Rodrigo Pacheco
- Laboratory of Neuroimmunology, Fundación Ciencia & Vida , Santiago , Chile ; Programa de Biomedicina, Universidad San Sebastián , Santiago , Chile
| | - Francisco Contreras
- Laboratory of Neuroimmunology, Fundación Ciencia & Vida , Santiago , Chile ; Universidad Andrés Bello, Facultad de Ciencias Biológicas , Santiago , Chile
| | - Moncef Zouali
- INSERM UMR 1132 , Paris , France ; University Paris Diderot , Paris , France
| |
Collapse
|
94
|
Gong S, Li J, Ma L, Li K, Zhang L, Wang G, Liu Y, Ji X, Liu X, Chen P, Ouyang R, Zhang S, Zhou Z, Wang CY, Xiang X, Yang Y. Blockade of dopamine D1-like receptor signalling protects mice against OVA-induced acute asthma by inhibiting B-cell activating transcription factor signalling and Th17 function. FEBS J 2013; 280:6262-73. [PMID: 24112622 DOI: 10.1111/febs.12549] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Revised: 09/07/2013] [Accepted: 09/24/2013] [Indexed: 01/21/2023]
Abstract
Previous studies have consistently demonstrated that dopamine D1-like receptor (D1-like-R) signalling is implicated in the pathogenesis of experimental autoimmune encephalomyelitis and type I diabetes. Given that allergic asthma shares certain disease aetiology similarities with autoimmune diseases, we conducted studies in OVA-induced mice aiming to address the impact of D1-like-R signalling on the pathogenesis of allergic asthma. It was noted that blockade of D1-like-R signalling provided protection for mice against OVA-induced acute asthma. Particularly, treatment of OVA-induced mice with SCH23390, a D1-like-R antagonist, significantly attenuated inflammatory infiltration in the airways along with repressed goblet cell hyperplasia and mucus production, as well as airway resistance. By contrast, administration of SKF83959, a D1-like-R agonist, displayed the opposite effect. Blockade of D1-like-R signalling impaired Th17 function, as manifested by a significant reduction of Th17 cells in the spleen and bronchoalveolar lavage fluid. Mechanistic studies revealed that D1-like-R signalling enhances B-cell activating transcription factor activity, which then transcribes the expression of RORγt, a Th17 transcription factor; accordingly, D1-like-R signalling regulates Th17 differentiation to promote the development of allergic asthma. Taken together, the data obtained in the present suggest that blockade of D1-like-R signalling could be an effective therapeutic strategy for the prevention and treatment of allergic asthma in clinical practice.
Collapse
Affiliation(s)
- Subo Gong
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, China; Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Yin W, Ouyang S, Li Y, Xiao B, Yang H. Immature dendritic cell-derived exosomes: a promise subcellular vaccine for autoimmunity. Inflammation 2013; 36:232-40. [PMID: 22956173 DOI: 10.1007/s10753-012-9539-1] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Exosomes, 60-90-nm-sized vesicles, are produced by a large number of cell types, including tumor cells, neurons, astrocytes, hemocytes, intestinal epithelial cells, and so on. Dendritic cell (DC), the most potent professional antigen-presenting cell in the immune system, produces exosomes in the course of maturation. Mature DCs produce exosomes with the ability to elicit potent immunoactivation, resulting in tumor eradication and bacterial or virus elimination. Given the notion that exosomes are stable and easy to be modified artificially, autologous mature DC-derived exosomes have been vaccinated into patients with malignant diseases. In clinical trials utilizing exosomes as therapeutic approaches, researchers observed considerable curative effect with little side effect. However, immature or suppressive DC-derived exosomes harbor anti-inflammatory properties distinct from mature DC-derived exosomes. In murine models of autoimmune disease and transplantation, immature DC-derived exosomes reduced T cell-dependent immunoactivation, relieved clinical manifestation of autoimmune disease, and prolonged survival time of transplantation. Although the exact mechanism of how immature DC-derived exosomes function in vivo is still unclear, and there are no clinical trials regarding application of exosome vaccine into patients with autoimmune disease, we will analyze the promise of immature DC-derived exosomes as a subcellular vaccine in autoimmunity in this review.
Collapse
Affiliation(s)
- Weifan Yin
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China, 410008
| | | | | | | | | |
Collapse
|
96
|
Ciaramella A, Salani F, Bizzoni F, Pontieri FE, Stefani A, Pierantozzi M, Assogna F, Caltagirone C, Spalletta G, Bossù P. Blood dendritic cell frequency declines in idiopathic Parkinson's disease and is associated with motor symptom severity. PLoS One 2013; 8:e65352. [PMID: 23776473 PMCID: PMC3679103 DOI: 10.1371/journal.pone.0065352] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 04/25/2013] [Indexed: 12/13/2022] Open
Abstract
The role of inflammation in Parkinson’s Disease (PD) is well appreciated, but its underlying mechanisms are still unclear. Our objective was to determine whether dendritic cells (DC), a unique type of migratory immune cells that regulate immunological response and inflammation have an impact on PD. In a case-control study including 80 PD patients and 80 age- and gender-matched healthy control subjects, the two main blood subsets of plasmacytoid and myeloid DC were defined by flow cytometry analysis. Clinical evaluation of subjects consisting of cognition and depression assessment was performed using the Mini Mental State Examination and the Beck Depression Inventory. The severity of motor symptoms was measured using the Unified Parkinson’s Disease Rating Scale-Part III. Comparison between patient and control DC measures and their relationships with clinical assessments were evaluated.The following main results were obtained: 1) the level of circulating DC (mainly the myeloid subset) was significantly reduced in PD patients in comparison with healthy controls; 2) after controlling for depressive and cognitive characteristics, the frequency of myeloid DC was confirmed as one of the independent determinants of PD; 3) the number of both myeloid and plasmacytoid DC was negatively associated with motor symptom severity. Overall, the decline of blood DC, perhaps due to the recruitment of immune cells to the site of disease-specific lesions, can be considered a clue of the immune alteration that characterizes PD, suggesting innovative exploitations of DC monitoring as a clinically significant tool for PD treatment. Indeed, this study suggests that reduced peripheral blood DC are a pathologically-relevant factor of PD and also displays the urgency to better understand DC role in PD for unraveling the immune system contribution to disease progression and thus favoring the development of innovative therapies ideally based on immunomodulation.
Collapse
Affiliation(s)
- Antonio Ciaramella
- Clinical and Behavioral Neurology, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Francesca Salani
- Clinical and Behavioral Neurology, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Federica Bizzoni
- Clinical and Behavioral Neurology, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Francesco E. Pontieri
- Department of Neurology and Psychiatry, University “Sapienza”, Movement Disorder Unit, Sant’Andrea Hospital, Rome, Italy
| | | | | | - Francesca Assogna
- Clinical and Behavioral Neurology, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Carlo Caltagirone
- Clinical and Behavioral Neurology, IRCCS Fondazione Santa Lucia, Rome, Italy
| | | | - Paola Bossù
- Clinical and Behavioral Neurology, IRCCS Fondazione Santa Lucia, Rome, Italy
- * E-mail:
| |
Collapse
|
97
|
Straub RH, Bijlsma JWJ, Masi A, Cutolo M. Role of neuroendocrine and neuroimmune mechanisms in chronic inflammatory rheumatic diseases--the 10-year update. Semin Arthritis Rheum 2013; 43:392-404. [PMID: 23731531 DOI: 10.1016/j.semarthrit.2013.04.008] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2013] [Revised: 04/04/2013] [Accepted: 04/13/2013] [Indexed: 12/20/2022]
Abstract
BACKGROUND Neuroendocrine immunology in musculoskeletal diseases is an emerging scientific field. It deals with the aspects of efferent neuronal and neurohormonal bearing on the peripheral immune and musculoskeletal systems. This review aims to add new information that appeared since 2001. SEARCH STRATEGY The following PubMed search sentence was used to find a total of 15,462 references between 2001 and March 2013: "(rheum* OR SLE OR vasculitis) AND (nerve OR hormone OR neurotransmitter OR neuropeptide OR steroid)." In a continuous process, year by year, this search strategy yielded relevant papers that were screened and collected in a database, which build the platform of this review. RESULTS The main findings are the anti-inflammatory role of androgens, the loss of androgens (androgen drain), the bimodal role of estrogens (support B cells and inhibit macrophages and T cells), increased conversion of androgens to estrogens in inflammation (androgen drain), disturbances of the gonadal axis, inadequate amount of HPA axis hormones relative to inflammation (disproportion principle), biologics partly improve neuroendocrine axes, anti-corticotropin-releasing hormone therapies improve inflammation (antalarmin), bimodal role of the sympathetic nervous system (proinflammatory early, anti-inflammatory late-most probably due to catecholamine-producing local cells), anti-inflammatory role of alpha melanocyte-stimulating hormone, vasoactive intestinal peptide, and the Vagus nerve via α7 nicotinergic receptors. Circadian rhythms of hypothalamic origin are responsible for circadian rhythms of symptoms (neuroimmune link revealed). Important new pain-sensitizing immunological pathways were found in the last decade. CONCLUSIONS The last decade brought much new information that gave birth to the first therapies of chronic inflammatory diseases on the basis of neuroendocrine immune targets. In addition, a new theory linked evolutionary medicine, neuroendocrine regulation of distribution of energy-rich fuels, and volume regulation that can explain many disease sequelae in patients with chronic inflammatory diseases.
Collapse
Affiliation(s)
- Rainer H Straub
- Laboratory of Experimental Rheumatology and Neuroendocrino-Immunology, Division of Rheumatology, Department of Internal Medicine I, University Hospital, Regensburg, Germany.
| | | | | | | |
Collapse
|
98
|
González H, Contreras F, Prado C, Elgueta D, Franz D, Bernales S, Pacheco R. Dopamine receptor D3 expressed on CD4+ T cells favors neurodegeneration of dopaminergic neurons during Parkinson's disease. THE JOURNAL OF IMMUNOLOGY 2013; 190:5048-56. [PMID: 23589621 DOI: 10.4049/jimmunol.1203121] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Emerging evidence has demonstrated that CD4(+) T cells infiltrate into the substantia nigra (SN) in Parkinson's disease (PD) patients and in animal models of PD. SN-infiltrated CD4(+) T cells bearing inflammatory phenotypes promote microglial activation and strongly contribute to neurodegeneration of dopaminergic neurons. Importantly, altered expression of dopamine receptor D3 (D3R) in PBLs from PD patients has been correlated with disease severity. Moreover, pharmacological evidence has suggested that D3R is involved in IFN-γ production by human CD4(+) T cells. In this study, we examined the role of D3R expressed on CD4(+) T cells in neurodegeneration of dopaminergic neurons in the SN using a mouse model of PD. Our results show that D3R-deficient mice are strongly protected against loss of dopaminergic neurons and microglial activation during 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD. Notably, D3R-deficient mice become susceptible to MPTP-induced neurodegeneration and microglial activation upon transfer of wild-type (WT) CD4(+) T cells. Furthermore, RAG1 knockout mice, which are devoid of T cells and are resistant to MPTP-induced neurodegeneration, become susceptible to MPTP-induced loss of dopaminergic neurons when reconstituted with WT CD4(+) T cells but not when transferred with D3R-deficient CD4(+) T cells. In agreement, experiments analyzing activation and differentiation of CD4(+) T cells revealed that D3R favors both T cell activation and acquisition of the Th1 inflammatory phenotype. These findings indicate that D3R expressed on CD4(+) T cells plays a fundamental role in the physiopathology of MPTP-induced PD in a mouse model.
Collapse
Affiliation(s)
- Hugo González
- Laboratorio de Neuroinmunología, Fundación Ciencia y Vida, Ñuñoa 7780272, Santiago, Chile
| | | | | | | | | | | | | |
Collapse
|