Dopaminergic Receptors on CD4+ T Naive and Memory Lymphocytes Correlate with Motor Impairment in Patients with Parkinson's Disease

Natasa Kustrimovic, Emanuela Rasini, Massimiliano Legnaro, Raffaella Bombelli, Iva Aleksic, Fabio Blandini, Cristoforo Comi, Marco Mauri, Brigida Minafra, Giulio Riboldazzi, Vanesa Sanchez-Guajardo, Franca Marino, Marco Cosentino, Natasa Kustrimovic, Emanuela Rasini, Massimiliano Legnaro, Raffaella Bombelli, Iva Aleksic, Fabio Blandini, Cristoforo Comi, Marco Mauri, Brigida Minafra, Giulio Riboldazzi, Vanesa Sanchez-Guajardo, Franca Marino, Marco Cosentino

Abstract

Parkinson's disease (PD) is characterized by loss of dopaminergic neurons in substantia nigra pars compacta, α-synuclein (α-syn)-rich intraneuronal inclusions (Lewy bodies), and microglial activation. Emerging evidence suggests that CD4+ T lymphocytes contribute to neuroinflammation in PD. Since the mainstay of PD treatment is dopaminergic substitution therapy and dopamine is an established transmitter connecting nervous and immune systems, we examined CD4+ T naive and memory lymphocytes in PD patients and in healthy subjects (HS), with specific regard to dopaminergic receptor (DR) expression. In addition, the in vitro effects of α-syn were assessed on CD4+ T naive and memory cells. Results showed extensive association between DR expression in T lymphocytes and motor dysfunction, as assessed by UPDRS Part III score. In total and CD4+ T naive cells expression of D1-like DR decrease, while in T memory cells D2-like DR increase with increasing score. In vitro, α-syn increased CD4+ T memory cells, possibly to a different extent in PD patients and in HS, and affected DR expression with cell subset-specific patterns. The present results support the involvement of peripheral adaptive immunity in PD, and may contribute to develop novel immunotherapies for PD, as well as to better use of current dopaminergic antiparkinson drugs.

Figures

Figure 1. CD4+ T naive and memory…
Figure 1. CD4+ T naive and memory cells in HS and PD patients.
Cells are shown as absolute numbers (panels a,b) and as percentage of total CD4+ T cells (c,d). Data are shown as medians with 25°–75° percentiles (boxes) and min-max values (whiskers). Comparisons are shown between HS and PD patients as a whole (a,c) and between drug naive (PD-dn) and drug treated (PD-dt) patients (b,d). Differences were analyzed by means of two-tailed Student’s t test or by Mann-Whitney test, as appropriate. P values less than 0.05 are indicated in the graphs.
Figure 2. DR expression on CD4+ T…
Figure 2. DR expression on CD4+ T cells from HS and from PD patients.
DR expression is shown as mRNA levels (panels a,b) and as protein expression on the membranes of CD4+ T cells, expressed as absolute numbers of DR+ cells (c,d) and as percentage of total CD4+ T cells (e,f). Comparisons are shown between HS and PD patients as a whole (a,c,e) and between drug naive (PD-dn) and drug treated (PD-dt) patients (b,d,f). Data are shown as medians with 25°–75° percentiles (boxes) and min-max values (whiskers). Differences were analyzed by means of two-tailed Student’s t test or by Mann-Whitney test, as appropriate. P values less than 0.05 are indicated in the graphs.
Figure 3. DR expression on CD4+ T…
Figure 3. DR expression on CD4+ T naive cells from HS and from PD patients.
DR+ cells are shown as absolute numbers (panels a,b) and as percentage of total CD4+ cells (c,d). Data are shown as medians with 25°–75° percentiles (boxes) and min-max values (whiskers). Comparisons are shown between HS and PD patients as a whole (a,c) and between drug naive (PD-dn) and drug treated (PD-dt) patients (b,d). Differences were analyzed by means of two-tailed Student’s t test or by Mann-Whitney test, as appropriate. P values less than 0.05 are indicated in the graphs.
Figure 4. DR expression on CD4+ T…
Figure 4. DR expression on CD4+ T cells and UPDRS-III score.
DR expression is shown as mRNA levels (panel a) and as protein expression on the membranes of CD4+ T cells, expressed as absolute numbers of DR+ cells (b) and as percentage of total CD4+ T cells (c) Data are medians with 25°–75° percentiles (boxes) and min-max values (whiskers). Differences in DR expression between HS and PD patients were analyzed by parametric ANOVA or Kruskal-Wallis nonparametric ANOVA, as appropriate, with either Holm-Sidak’s or Dunn’s adjustments for multiple comparisons, where * = P < 0.05 and ** = P < 0.01. Trend analysis in PD patients was performed by ANOVA post test for linear trend.
Figure 5. DR expression in CD4+ T…
Figure 5. DR expression in CD4+ T naïve, TCM and TEM and UPDRS-III score.
DR expression is shown as protein expression on the membranes of CD4+ T naïve (left), TCM (center) and TEM (right) cells, expressed as absolute numbers of DR+ cells. Data are medians with 25°–75° percentiles (boxes) and min-max values (whiskers). Differences between DR levels in HS and in PD patients were analyzed by parametric ANOVA or Kruskal-Wallis nonparametric ANOVA, with either Holm-Sidak’s or Dunn’s adjustments for multiple comparisons, where * = P < 0.05 and ** = P < 0.01. Trend analysis in PD patients was performed by ANOVA post test for linear trend.
Figure 6. Effect of TTd, monomeric, and…
Figure 6. Effect of TTd, monomeric, and fibrillar α-syn on the frequency of CD4+ T naive and memory subsets.
Panel (a) Effects of TTd (left), monomeric (middle), and fibrillar α-syn (right) in cells from HS (open columns) and PD patients (hatched columns). Data are expressed as percentage variation with respect to control conditions (without TTd or α-syn), and are means ± SEM of n = 6–8 separate experiments each performed in duplicate. * = P < 0.05 and ** = P < 0.01 vs. control conditions, and # = P < 0.01 vs. HS. Panel (b) monomeric (hatched columns), and fibrillar α-syn (shaded columns) on DR expression in T naive (left), TCM (center) and TEM cells (right) from 5 HS. Data are means ± SEM. * = P < 0.05 and ** = P < 0.01 vs control (open columns); # = P < 0.01 vs monomeric α-syn.

References

    1. Berg D. et al.. Time to redefine PD? Introductory statement of the MDS task force on the definition of Parkinson’s disease. Mov. Disord . 29, 454–462 (2014).
    1. Obeso J. A., Rodriguez-Oroz M. C., Stamelou M., Bhatia K. P. & Burn D. J. The expanding universe of disorders of the basal ganglia. Lancet . 384, 523–531 (2014).
    1. Pringsheim T., Jette N., Frolkis A. & Steeves T. D. The prevalence of Parkinson’s disease: a systematic review and meta-analysis. Mov. Disord . 29, 1583–1590 (2014).
    1. Cappellano G. et al.. Immunity and inflammation in neurodegenerative diseases. Am. J. Neurodegener. Dis . 2, 89–107 (2013)
    1. Przedborski S. Inflammation and Parkinson’s disease pathogenesis. Mov. Disord. 25, S55–57 (2010).
    1. Mosley R. L., Hutter-Saunders J. A., Stone D. K. & Gendelman H. E. Inflammation and adaptive immunity in Parkinson’s disease. Cold. Spring. Harb. Perspect. Med . 2, a009381 (2012).
    1. González H., Elgueta D., Montoya A. & Pacheco R. Neuroimmune regulation of microglial activity involved in neuroinflammation and neurodegenerative diseases. J. Neuroimmunol. 274, 1–13 (2014).
    1. Baba Y., Kuroiwa A., Uitti R. J., Wszolek Z. K. & Yamada T. Alterations of T-lymphocyte populations in Parkinson disease. Parkinsonism Relat. Disord. 11, 493–498 (2005).
    1. Bas J. et al.. Lymphocyte populations in Parkinson’s disease and in rat models of parkinsonism. J Neuroimmunol . 113, 146–152 (2001).
    1. Stevens C. H. et al.. Reduced T helper and B lymphocytes in Parkinson’s disease. J. Neuroimmunol. 252, 95–99 (2012).
    1. Brochard V. et al.. Infiltration of CD4+ lymphocytes into the brain contributes to neurodegeneration in a mouse model of Parkinson disease. J. Clin. Invest . 119, 182–192 (2009).
    1. Saunders J. A. et al.. CD4+ regulatory and effector/memory T cell subsets profile motor dysfunction in Parkinson’s disease. J. Neuroimmune Pharmacol . 7, 927–938 (2012).
    1. Connolly B.S. & Lang A.E. Pharmacological treatment of Parkinson disease: a review. JAMA 311, 1670–1683 (2014).
    1. Basu S. & Dasgupta P. S. Dopamine, a neurotransmitter, influences the immune system. J Neuroimmunol . 102, 113–124 (2000).
    1. Sarkar C., Basu B., Chakroborty D., Dasgupta P. S. & Basu S. The immunoregulatory role of dopamine: an update. Brain Behav. Immun. 24, 525–528 (2010).
    1. Levite M. Dopamine in the immune system: dopamine receptors in immune cells, potent effects, endogenous production and involvement in immune and neuropsychiatric diseases. (ed. Levite M. ) In Nerve-driven-immunity – Neurotransmitters and neuropeptides in the immune system 1–45 (Springer-Verlag, 2012).
    1. Cosentino M. et al.. Endogenous catecholamine synthesis, metabolism, storage and uptake in human neutrophils. Life Sci . 64, 975–981 (1999).
    1. Marino F. et al.. Endogenous catecholamine synthesis, metabolism storage, and uptake in human peripheral blood mononuclear cells. Exp. Hematol. 27, 489–495 (1999).
    1. Cosentino M. et al.. HPLC-ED measurement of endogenous catecholamines in human immune cells and hematopoietic cell lines. Life Sci . 68, 283–295 (2000).
    1. Cosentino M. et al.. Stimulation with phytohaemagglutinin induces the synthesis of catecholamines in human peripheral blood mononuclear cells:role of protein kinase C and contribution of intracellular calcium. J. Neuroimmunol . 125, 125–133 (2002a).
    1. Cosentino M. et al.. Catecholamine production and tyrosine hydroxylase expression in peripheral blood mononuclear cells from multiple sclerosis patients: effect of cell stimulation and possible relevance for activation-induced apoptosis. J. Neuroimmunol. 133, 233–240 (2002b).
    1. Cosentino M. et al.. Interferon-gamma and interferon-beta affect endogenous catecholamines in human peripheral blood mononuclear cells: implications for multiple sclerosis. J. Neuroimmunol. 162, 112–121 (2005).
    1. Cosentino M. et al.. Human CD4+ CD25+ regulatory T cells selectively express tyrosine hydroxylase and contain endogenous catecholamines subserving an autocrine/paracrine inhibitory functional loop. Blood . 109, 632–642 (2007).
    1. Nakano K. et al.. Dopamine released by dendritic cells polarizes Th2 differentiation. Int. Immunol. 21, 645–654 (2009).
    1. Schetz J. A. Dopamine receptors, introduction. IUPHAR/BPS Guide to PHARMACOLOGY Aviable at: . (Accessed on 31/01/2014) (2009).
    1. Beaulieu J. M. & Gainetdinov R. R. The physiology, signalling, and pharmacology of dopamine receptors. Pharmacol. Rev. 63, 182–217 (2011).
    1. Brito-Melo G. E. et al.. Increase in dopaminergic, but not serotoninergic, receptors in T-cells as a marker for schizophrenia severity. J. Psychiatr. Res. 46, 738–742 (2012).
    1. Kustrimovic N., Rasini E., Legnaro M., Marino F. & Cosentino M. Expression of dopaminergic receptors on human CD4+ T lymphocytes: flow cytometric analysis of naive and memory subsets and relevance for the neuroimmunology of neurodegenerative disease. J. Neuroimmune Pharmacol . 9, 302–312 (2014).
    1. González H. et al.. Dopamine receptor D3 expressed on CD4+ T cells favors neurodegeneration of dopaminergic neurons during Parkinson’s disease. J. Immunol . 190, 5048–5056 (2013).
    1. Harms A. S. et al.. MHCII is required for α-synuclein-induced activation of microglia, CD4 T cell proliferation, and dopaminergic neurodegeneration. J. Neurosci. 33, 9592–9600 (2013).
    1. Reynolds A. D., Stone D. K., Mosley R. L. & Gendelman H. E. Nitrated {alpha}-synuclein-induced alterations in microglial immunity are regulated by CD4+ T cell subsets. J. Immunol. 182, 4137–4149 (2009).
    1. Sanchez-Guajardo V., Tentillier N. & Romero-Ramos M. The relation between α-synuclein and microglia in Parkinson’s disease: Recent developments. Neuroscience . 302, 47–58 (2015).
    1. Linton P. J. & Dorshkind K. Age-related changes in lymphocyte development and function. Nat. Immunol. 5, 133–139 (2004).
    1. Lazuardi L. et al.. Age-related loss of naïve T cells and dysregulation of T-cell/B-cell interactions in human lymph nodes. Immunology . 114, 37–43 (2005).
    1. Leddy A. L., Crowner B. E. & Earhart G. M. Functional gait assessment and balance evaluation system test: reliability, validity, sensitivity, and specificity for identifying individuals with Parkinson disease who fall. Phys. Ther. 91, 102–113 (2011).
    1. Lanzavecchia A. & Sallusto F. Dynamics of T lymphocyte responses: intermediates, effectors, and memory cells. Science . 290, 92–97 (2000).
    1. Sallusto F., Geginat J. & Lanzavecchia A. Central memory and effector memory T cell subsets: function, generation, and maintenance. Annu. Rev. Immunol. 22, 745–763 (2004).
    1. Saha B., Mondal A. C., Basu S. & Dasgupta P. S. Circulating dopamine level, in lung carcinoma patients, inhibits proliferation and cytotoxicity of CD4+ and CD8+ T cells by D1 dopamine receptors: an in vitro analysis. Int. Immunopharmacol. 1, 1363–1374 (2001).
    1. Levite M. et al.. Dopamine interacts directly with its D3 and D2 receptors on normal human T cells, and activates β-integrin function. Eur. J. Immunol. 31, 3504–3512 (2001).
    1. Sarkar C. et al.. Cutting Edge: Stimulation of dopamine D4 receptors induce T cell quiescence by up-regulating Kruppel-like factor-2 expression through inhibition of ERK1/ERK2 phosphorylation. J. Immunol. 177, 7525–7529 (2006).
    1. Wang S. et al.. α-Synuclein, a chemoattractant, directs microglial migration via H2O2-dependent Lyn phosphorylation. Proc. Natl. Acad. Sci. USA 112, E1926–1935 (2015).
    1. Shi M. et al.. Plasma exosomal α-synuclein is likely CNS-derived and increased in Parkinson’s disease. Acta Neuropathol . 128, 639–650 (2014).
    1. Appel. S. H., Beers D. R. & Henkel J. S. T cell-microglial dialogue in Parkinson’s disease and amyotrophic lateral sclerosis: are we listening? Trends Immunol . 31, 7–17 (2010).
    1. Hutter-Saunders J. A., Mosley R. L. & Gendelman H. E. Pathways towards an effective immunotherapy for Parkinson’s disease. Expert. Rev. Neurother . 11, 1703–1715 (2011).
    1. Conway K. A., Harper J. D. & Lansbury P. T. Accelerated in vitro fibril formation by a mutant α-synuclein linked to early-onset Parkinson disease. Nat. Med . 4, 1318–1320 (1998).
    1. Narhi L. et al.. Both familial Parkinson’s disease mutations accelerate α-synuclein aggregation. J. Biol. Chem. 274, 9843–9846 (1999).
    1. Yanamandra K. et al.. α-Synuclein Reactive Antibodies as Diagnostic Biomarkers in Blood Sera of Parkinson’s Disease Patients. PLOS . 6, 18513 (2011).
    1. Smith L. M., Schiess M. C., Coffey M. P., Klaver A. C. & Loeffler D. A. α-Synuclein and anti-α-synuclein antibodies in Parkinson’s disease, atypical Parkinson syndromes, REM sleep behavior disorder, and healthy controls. PLoS One . 7, e52285 (2012).
    1. Besong-Agbo D. et al.. Naturally occurring α-synuclein autoantibody levels are lower in patients with Parkinson disease. Neurology . 80, 169–75 (2013).
    1. Cosentino M. & Marino F. Adrenergic and dopaminergic modulation of immunity in multiple sclerosis: teaching old drugs new tricks? J. Neuroimmune Pharmacol . 8, 163–179 (2013).
    1. Gelb D. J., Oliver E. & Gilman S. Diagnostic criteria for Parkinson disease. Arch. Neurol. 56, 33–39 (1999).
    1. Goetz C. G. et al.. Movement Disorder Society Task Force on Rating Scales for Parkinson’s Disease. Movement Disorder Society Task Force report on the Hoehn and Yahr staging scale: status and recommendations. Mov. Disord . 19, 1020–1028 (2004).
    1. Goetz C. G. et al.. Movement Disorder Society-Sponsored Revision of the Unifed Parkinson’s Disease Rating Scale (MDS-UPDRS): Scale Presentation and Clinimetric Testing Results. Mov. Disord . 23, 2129–2170 (2008).
    1. Tomlinson C. L. et al.. Systematic review of levodopa dose equivalency reporting in Parkinson’s disease. Mov. Disord. 25, 2649–2653 (2010).
    1. Nielsen S. B. et al.. Wild type and A30P mutant alpha-synuclein form different fibril structures. PloS one . 8, e67713 (2013).
    1. Cosentino M. et al.. Dopaminergic modulation of CD4+ CD25(high) regulatory T lymphocytes in multiple sclerosis patients during interferon-β therapy. Neuroimmunomodulation . 19, 283–292 (2012).

Source: PubMed

3
Abonnere