17
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Targeting dopamine receptor D3 signalling in inflammation

      editorial

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          During last 15 years dopamine has emerged as a major regulator of inflammation. All five dopamine receptors (DRs, DRD1-DRD5) have been found to be expressed in immune cells where they exert a complex regulation of immunity [1]. Of note, DRs have been found not only in cells of the adaptive immune system, but also in cells belonging to the innate immunity, even including glial cells. The outcome of the dopamine effect in the immune response depends in many factors, including differential expression of DRs in the immune cells present in the inflamed tissue, the local levels of dopamine and the signalling coupled to and the affinity of the different DRs involved. An increasing number of studies analysing human cells in vitro or using in vivo approaches in animal models have been unravelling and deciphering the complexity of dopaminergic regulation of immunity. Integrating the knowledge acquired by these studies, the evidence have indicated that stimulation of low-affinity DRs, for instance DRD1 and DRD2, are coupled to anti-inflammatory mechanisms, thereby dampening inflammation [2, 3]. Conversely, signalling triggered by high-affinity DRs, including DRD3 and DRD5, have been found consistently to promote inflammation [4, 5]. It is noteworthy that tissues containing high-levels of dopamine in steady-state, such as the nigrostriatal pathway or the gut mucosa, undergo a strong decrease of dopamine levels during inflammation [1]. This fact involves a switch in the stimulation of DRs: low-affinity DRs, which display anti-inflammatory properties and are stimulated by high dopamine levels under homeostasis, are not longer stimulated during inflammatory processes. Otherwise, signalling coupled to high-affinity DRs, which seems to be dampened by low-affinity DRs stimulation under homeostasis, becomes dominant when dopamine levels are reduced during inflammation. This idea highlight the relevance of high-affinity DRs favouring the development and progression of inflammatory disorders and makes these receptors key therapeutic targets. Accordingly, DRD3, which display the highest affinity by dopamine, has been strongly involved in favouring inflammation in several experimental systems. In this regard, genetic and pharmacological evidence has indicated that DRD3-signalling constitutes a potent regulator of CD4+ T-cell-mediated responses, including those implicated in Parkinson’s disease [6] and inflammatory colitis [4], two pathologies that involve a reduction of dopamine levels in the inflamed tissue (Figure 1). Mechanistic analyses have revealed that DRD3-signalling in CD4+ T-cells induces suppressor of cytokine signalling 5 in these cells, thus attenuating T-helper 2 (Th2) differentiation and promoting Th1 responses. Moreover, evidence has also indicated that DRD3-signalling favours Th17-immunity under chronic inflammatory conditions [4]. According to the pivotal role of Th1 and Th17 inflammatory responses in the development of Parkinson’s disease, we have recently demonstrated the therapeutic potential of DRD3- antagonism in two different animal models, including 6-hydroxydopamine-induced and 1-methyl-4-phenyl- 1,2,3,6-tetrahydropyridine-induced Parkinson’s disease [7]. In those studies, DRD3-antagonism not only reduced the neurodegenerative and neuroinflammatory process, but also attenuated significantly the motor impairment associated to the loss of dopaminergic neurons. Figure 1 Role of DRD3-signaling in CD4+ T-cells favouring Th1 and Th17 mediated responses in disorders involving reduction of dopamine levels in the target tissue, such as Parkinson’s disease and inflammatory bowel diseases. Importantly, the therapeutic effect observed in Parkinson’s disease models in our studies [7] was exerted by a highly-selective DRD3 antagonist, PG01037, displaying the Ki values 0.70, 93.3 and 375 nM for DRD3, DRD2 and DRD4 respectively. In apparent controversy with our recent findings, pramipexole, a drug described as a DRD3-agonist, has been used for the symptomatic treatment of Parkinson’s disease. However, the range of DRD3-selectivity for this drug is very limited, displaying Ki values of 0.5, 3.3, 3.9 and 3.9 nM for DRD3, DRD2S, DRD2L and DRD4 respectively. Thereby, it is likely that at therapeutic concentrations pramipexole stimulates the dominant effects of DRD2-signalling, abolishing DRD3- mediated effects. Interestingly, the therapeutic effect of DRD3- antagonism seems to be beyond of targeting DRD3 confined to CD4+ T-cells. Our analyses performed in glial cells have suggested that DRD3-deficiency gives an anti-inflammatory behaviour to astrocytes, which results in attenuated microglial activation in a model of Parkinson’s disease [7]. Furthermore, recently we found that DRD3-deficiency or DRD3-antagonism in dendritic cells results in an exacerbated stimulation of CD8+ T-cell-mediated immunity, strengthening the immune response against tumours [8]. Mechanistic analyses indicated that the inhibition of DRD3-signalling in dendritic cells promotes an increase in antigen cross-presentation in class I MHC molecules to CD8+ T-cells, thereby potentiating the development of cytotoxic T-lymphocytes. Thus, emerging evidence indicates DRD3 as a key therapeutic target with a dual potential: whereas DRD3-inhibtion attenuates inflammation in pathologies associated to reduction of dopamine levels and CD4+ T-cell-mediated responses (Figure 1), blocking DRD3-signalling in dendritic cells may improve the outcome of disorders that involve insufficient cytotoxic T-lymphocyte-responses, such as cancer.

          Related collections

          Most cited references7

          • Record: found
          • Abstract: found
          • Article: not found

          Dopamine controls systemic inflammation through inhibition of NLRP3 inflammasome.

          Inflammasomes are involved in diverse inflammatory diseases, so the activation of inflammasomes needs to be tightly controlled to prevent excessive inflammation. However, the endogenous regulatory mechanisms of inflammasome activation are still unclear. Here, we report that the neurotransmitter dopamine (DA) inhibits NLRP3 inflammasome activation via dopamine D1 receptor (DRD1). DRD1 signaling negatively regulates NLRP3 inflammasome via a second messenger cyclic adenosine monophosphate (cAMP), which binds to NLRP3 and promotes its ubiquitination and degradation via the E3 ubiquitin ligase MARCH7. Importantly, in vivo data show that DA and DRD1 signaling prevent NLRP3 inflammasome-dependent inflammation, including neurotoxin-induced neuroinflammation, LPS-induced systemic inflammation, and monosodium urate crystal (MSU)-induced peritoneal inflammation. Taken together, our results reveal an endogenous mechanism of inflammasome regulation and suggest DRD1 as a potential target for the treatment of NLRP3 inflammasome-driven diseases.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Suppression of neuroinflammation by astrocytic dopamine D2 receptors via αB-crystallin.

            Chronic neuroinflammation is a common feature of the ageing brain and some neurodegenerative disorders. However, the molecular and cellular mechanisms underlying the regulation of innate immunity in the central nervous system remain elusive. Here we show that the astrocytic dopamine D2 receptor (DRD2) modulates innate immunity through αB-crystallin (CRYAB), which is known to suppress neuroinflammation. We demonstrate that knockout mice lacking Drd2 showed remarkable inflammatory response in multiple central nervous system regions and increased the vulnerability of nigral dopaminergic neurons to neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced neurotoxicity. Astrocytes null for Drd2 became hyper-responsive to immune stimuli with a marked reduction in the level of CRYAB. Preferential ablation of Drd2 in astrocytes robustly activated astrocytes in the substantia nigra. Gain- or loss-of-function studies showed that CRYAB is critical for DRD2-mediated modulation of innate immune response in astrocytes. Furthermore, treatment of wild-type mice with the selective DRD2 agonist quinpirole increased resistance of the nigral dopaminergic neurons to MPTP through partial suppression of inflammation. Our study indicates that astrocytic DRD2 activation normally suppresses neuroinflammation in the central nervous system through a CRYAB-dependent mechanism, and provides a new strategy for targeting the astrocyte-mediated innate immune response in the central nervous system during ageing and disease.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Dopamine receptor D3 expressed on CD4+ T cells favors neurodegeneration of dopaminergic neurons during Parkinson's disease.

              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.
                Bookmark

                Author and article information

                Journal
                Oncotarget
                Oncotarget
                Oncotarget
                ImpactJ
                Oncotarget
                Impact Journals LLC
                1949-2553
                31 January 2017
                12 January 2017
                : 8
                : 5
                : 7224-7225
                Affiliations
                Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas, Universidad Andres Bello, Santiago, Chile; Laboratorio de Neuroinmunología, Fundación Ciencia & Vida, Ñuñoa, Santiago, Chile
                Author notes
                Correspondence to: Rodrigo Pacheco, rpacheco@ 123456cienciavida.org
                Article
                14601
                10.18632/oncotarget.14601
                5352314
                28086229
                88e57dba-97db-40ee-9947-03a74cec81bb
                Copyright: © 2017 Pacheco

                This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

                History
                : 6 January 2017
                : 10 January 2017
                Categories
                Editorial: Neuroscience

                Oncology & Radiotherapy
                t-cell mediated immunity,dopaminergic system,dopamine receptors,neuroinflammation,inflammatory disorders,neuroscience

                Comments

                Comment on this article