Parkinson's disease patients have a complex phenotypic and functional Th1 bias: cross-sectional studies of CD4+ Th1/Th2/T17 and Treg in drug-naïve and drug-treated patients

Natasa Kustrimovic, Cristoforo Comi, Luca Magistrelli, Emanuela Rasini, Massimiliano Legnaro, Raffaella Bombelli, Iva Aleksic, Fabio Blandini, Brigida Minafra, Giulio Riboldazzi, Andrea Sturchio, Marco Mauri, Giorgio Bono, Franca Marino, Marco Cosentino, Natasa Kustrimovic, Cristoforo Comi, Luca Magistrelli, Emanuela Rasini, Massimiliano Legnaro, Raffaella Bombelli, Iva Aleksic, Fabio Blandini, Brigida Minafra, Giulio Riboldazzi, Andrea Sturchio, Marco Mauri, Giorgio Bono, Franca Marino, Marco Cosentino

Abstract

Background: Parkinson's disease (PD) affects an estimated 7 to 10 million people worldwide, and only symptomatic treatments are presently available to relieve the consequences of brain dopaminergic neurons loss. Neuronal degeneration in PD is the consequence of neuroinflammation in turn influenced by peripheral adaptive immunity, with CD4+ T lymphocytes playing a key role. CD4+ T cells may however acquire proinflammatory phenotypes, such as T helper (Th) 1 and Th17, as well as anti-inflammatory phenotypes, such as Th2 and the T regulatory (Treg) one, and to what extent the different CD4+ T cell subsets are imbalanced and their functions dysregulated in PD remains largely an unresolved issue.

Methods: We performed two cross-sectional studies in antiparkinson drug-treated and drug-naïve PD patients, and in age- and sex-matched healthy subjects. In the first one, we examined circulating Th1, Th2, Th17, and in the second one circulating Treg. Number and frequency of CD4+ T cell subsets in peripheral blood were assessed by flow cytometry and their functions were studied in ex vivo assays. In both studies, complete clinical assessment, blood count and lineage-specific transcription factors mRNA levels in CD4+ T cells were independently assessed and thereafter compared for their consistency.

Results: PD patients have reduced circulating CD4+ T lymphocytes, due to reduced Th2, Th17, and Treg. Naïve CD4+ T cells from peripheral blood of PD patients preferentially differentiate towards the Th1 lineage. Production of interferon-γ and tumor necrosis factor-α by CD4+ T cells from PD patients is increased and maintained in the presence of homologous Treg. This Th1-biased immune signature occurs in both drug-naïve patients and in patients on dopaminergic drugs, suggesting that current antiparkinson drugs do not affect peripheral adaptive immunity.

Conclusions: The complex phenotypic and functional profile of CD4+ T cell subsets in PD patients strengthen the evidence that peripheral adaptive immunity is involved in PD, and represents a target for the preclinical and clinical assessment of novel immunomodulating therapeutics.

Keywords: CD4+ T lymphocytes; Parkinson’s disease; Th1; Th17; Th2; Treg.

Conflict of interest statement

Ethics approval and consent to participate

The Ethics Committees of Ospedale di Circolo of Varese (I) and Neurological Institute “C. Mondino” of Pavia (I) approved the protocol and all the participants signed a written informed consent before enrollment.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Figures

Fig. 1
Fig. 1
CD4+ T cells in HS and PD patients. Cells are shown as absolute numbers (left panel) and as percentage of total lymphocytes (right panel). Data are medians with 25°–75° percentiles (boxes) and min–max values (whiskers)
Fig. 2
Fig. 2
Th subsets in HS and PD patients. Cells are shown as absolute numbers (upper panels) and as percentage of total lymphocytes (lower panels). Th1/Th2 ratio and Th1/Th17 ratio are also shown (middle panels). Data are medians with 25°–75° percentiles (boxes) and min–max values (whiskers)
Fig. 3
Fig. 3
Transcription factors mRNA levels in CD4+ T cells of HS and PD patients. Data are medians with 25°–75° percentiles (boxes) and min–max values (whiskers)
Fig. 4
Fig. 4
Polarization of naïve CD4+ T cells towards Th1, Th2, and Th17 in HS and PD patients. Data are means ± SD of n = 5–6 subjects tested in triplicate. * = P < 0.05 and ** = P < 0.01 vs HS; # = P < 0.05 and ## = P < 0.01 vs standard conditions (Th0)
Fig. 5
Fig. 5
Treg cells in HS and PD patients. Cells are shown as absolute numbers (upper panels) and as percentage of total CD4+ T cells and of total Treg, respectively (lower panels). Data are medians with 25°–75° percentiles (boxes) and min–max values (whiskers)
Fig. 6
Fig. 6
Treg-induced inhibition of Teff proliferation in HS and PD patients. Teff from HS and PD patients proliferated to a similar extent in the presence of PHA (a) and were concentration-dependently inhibited by Treg (b). Treg inhibition of Teff proliferation was reduced by DA 1 μM in cells from HS and PD-dn patients but not from PD-dt patients (c). Data are means ± SD of n = 9–17 subjects. *P < 0.05 and **P < 0.01 vs respective Teff alone; #P < 0.05 vs respective Teff:Treg 1:1; §P < 0.05 vs both HS and PD-dn
Fig. 7
Fig. 7
Cytokine production by Teff cells in HS and PD patients. Cytokines were measured in supernatants of resting cells (white columns) and of cells stimulated with PHA (black columns). Data are means ± SD of n = 4–9 subjects. *P < 0.05 and **P < 0.01 vs resting cells; # = P < 0.05 and ##P < 0.01 vs HS
Fig. 8
Fig. 8
Teff production of IFN-γ and TNF-α and inhibition by Treg in HS and PD patients. Teff were stimulated with PHA alone (white columns) or in the presence of Treg 1:1 (hatched columns). Data are means ± SD of n = 6–10 subjects. *P < 0.05 and **P < 0.01 vs Teff alone; #P < 0.01 vs HS
Fig. 9
Fig. 9
The Th1 bias in PD. Circulating CD4+ T naive cells in PD patients are reduced by about 30%; however, the proportion of IFN-γ-positive cells is increased (1). Differentiation towards the Th1 lineage is increased (2), while differentiation towards Th17 is impaired (3), and both Th2 and Th17 cells in blood are reduced on average by 20–30% (4). Production of IFN-γ and TNF-α by Th1 cells is strongly increased (5) and not impaired in the presence of Treg, which are also reduced by 30% in the circulation (6). Reduced production of IL-10 by CD4+ effector T cells, which likely contributes to amplify the Th1 bias, is not represented

References

    1. Pringsheim T, Jette N, Frolkis A, Steeves TD. The prevalence of Parkinson’s disease: a systematic review and meta-analysis. Mov Disord. 2014;29:1583. doi: 10.1002/mds.25945.
    1. Tysnes OB, Storstein A. Epidemiology of Parkinson’s disease. J Neural Transm (Vienna) 2017;124:901. doi: 10.1007/s00702-017-1686-y.
    1. Boland DF, Stacy M. The economic and quality of life burden associated with Parkinson's disease: a focus on symptoms. Am J Manag Care. 2012;18(7):S168.
    1. Kowal SL, Dall TM, Chakrabarti R, Storm MV, Jain A. The current and projected economic burden of Parkinson’s disease in the United States. Mov Disord. 2013;28:311. doi: 10.1002/mds.25292.
    1. Johnson SJ, Diener MD, Kaltenboeck A, Birnbaum HG, Siderowf AD. An economic model of Parkinson’s disease: implications for slowing progression in the United States. Mov Disord. 2013;28:319. doi: 10.1002/mds.25328.
    1. Connolly BS, Lang AE. Pharmacological treatment of Parkinson disease: a review. JAMA. 2014;311:1670. doi: 10.1001/jama.2014.3654.
    1. Fox SH. Non-dopaminergic treatments for motor control in Parkinson’s disease. Drugs. 2013;73:1405. doi: 10.1007/s40265-013-0105-4.
    1. Nonnekes J, Timmer MH, de Vries NM, Rascol O, Helmich RC, Bloem BR. Unmasking levodopa resistance in Parkinson’s disease. Mov Disord. 2016;31:1602. doi: 10.1002/mds.26712.
    1. Eriksen JL, Wszolek Z, Petrucelli L. Molecular pathogenesis of Parkinson disease. Arch Neurol. 2005;62:353. doi: 10.1001/archneur.62.3.353.
    1. Schapira AH, Jenner P. Etiology and pathogenesis of Parkinson’s disease. Mov Disord. 2011;26:1049. doi: 10.1002/mds.23732.
    1. Kalia LV, Lang AE. Parkinson’s disease. Lancet. 2015;386:896. doi: 10.1016/S0140-6736(14)61393-3.
    1. Dehay B, Bourdenx M, Gorry P, Przedborski S, Vila M, Hunot S, Singleton A, Olanow CW, Merchant KM, Bezard E, Petsko GA, Meissner WG. Targeting α-synuclein for treatment of Parkinson's disease: mechanistic and therapeutic considerations. Lancet Neurol. 2015;14:855. doi: 10.1016/S1474-4422(15)00006-X.
    1. Przedborski S. Inflammation and Parkinson’s disease pathogenesis. Mov Disord. 2010;25(1):S55. doi: 10.1002/mds.22638.
    1. Mosley RL, Hutter-Saunders JA, Stone DK, Gendelman HE. Inflammation and adaptive immunity in Parkinson’s disease. Cold Spring Harb Perspect Med. 2012;2:a009381. doi: 10.1101/cshperspect.a009381.
    1. Cappellano G, Carecchio M, Fleetwood T, Magistrelli L, Cantello R, Dianzani U, Comi C. Immunity and inflammation in neurodegenerative diseases. Am J Neurodegener Dis. 2013;2:89.
    1. González H, Elgueta D, Montoya A, Pacheco R. Neuroimmune regulation of microglial activity involved in neuroinflammation and neurodegenerative diseases. J Neuroimmunol. 2014;274:1. doi: 10.1016/j.jneuroim.2014.07.012.
    1. McGeer PL, Itagaki S, Akiyama H, McGeer EG. Rate of cell death in parkinsonism indicates active neuropathological process. Ann Neurol. 1988;24:574. doi: 10.1002/ana.410240415.
    1. Brochard V, Combadière B, Prigent A, Laouar Y, Perrin A, Beray-Berthat V, Bonduelle O, Alvarez-Fischer D, Callebert J, Launay JM, Duyckaerts C, Flavell RA, Hirsch EC, Hunot S. Infiltration of CD4+ lymphocytes into the brain contributes to neurodegeneration in a mouse model of Parkinson disease. J Clin Invest. 2009;119:182.
    1. Jiang S, Gao H, Luo Q, Wang P, Yang X. The correlation of lymphocyte subsets, natural killer cell, and Parkinson’s disease: a meta-analysis. Neurol Sci. 2017;38:1373. doi: 10.1007/s10072-017-2988-4.
    1. Zhu J, Yamane H, Paul WE. Differentiation of effector CD4 T cell populations. Annu Rev Immunol. 2010;28:445. doi: 10.1146/annurev-immunol-030409-101212.
    1. O’Shea JJ, Paul WE. Mechanisms underlying lineage commitment and plasticity of helper CD4+ T cells. Science. 2010;327:1098. doi: 10.1126/science.1178334.
    1. González H, Contreras F, Pacheco R. Regulation of the neurodegenerative process associated to Parkinson’s disease by CD4+ T-cells. J NeuroImmune Pharmacol. 2015;10:561. doi: 10.1007/s11481-015-9618-9.
    1. Kustrimovic N, Rasini E, Legnaro M, Bombelli R, Aleksic I, Blandini F, Comi C, Mauri M, Minafra B, Riboldazzi G, Sanchez-Guajardo V, Marino F, Cosentino M. Dopaminergic receptors on CD4+ T naïve and memory lymphocytes correlate with motor impairment in patients with Parkinson’s disease. Sci Rep. 2016;6:33738. doi: 10.1038/srep33738.
    1. Gelb DJ, Oliver E, Gilman S. Diagnostic criteria for Parkinson disease. Arch Neurol. 1999;56:33. doi: 10.1001/archneur.56.1.33.
    1. Goetz CG, Poewe W, Rascol O, Sampaio C, Stebbins GT, Counsell C, Giladi N, Holloway RG, Moore CG, Wenning GK, Yahr MD, Seidl L. 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. 2004;19:1020. doi: 10.1002/mds.20213.
    1. Goetz CG, Tilley BC, Shaftman SR, Stebbins GT, Fahn S, Martinez-Martin P, Poewe W, Sampaio C, Stern MB, Dodel R, Dubois B, Holloway R, Jankovic J, Kulisevsky J, Lang AE, Lees A, Leurgans S, LeWitt PA, Nyenhuis D, Olanow CW, Rascol O, Schrag A, Teresi JA, van Hilten JJ, LaPelle N. Movement Disorder Society UPDRS Revision Task Force. Movement Disorder Society-sponsored revision of the Unifed Parkinson’s disease rating scale (MDS-UPDRS): scale presentation and clinimetric testing results. Mov Disord. 2008;23:2129. doi: 10.1002/mds.22340.
    1. Tomlinson CL, Stowe R, Patel S, Rick C, Gray R, Clarke CE. Systematic review of levodopa dose equivalency reporting in Parkinson’s disease. Mov Disord. 2010;25:2649. doi: 10.1002/mds.23429.
    1. Miyara M, Yoshioka Y, Kitoh A, Shima T, Wing K, Niwa A, Parizot C, Taflin C, Heike T, Valeyre D, Mathian A, Nakahata T, Yamaguchi T, Nomura T, Ono M, Amoura Z, Gorochov G, Sakaguchi S. Functional delineation and differentiation dynamics of human CD4+ T cells expressing the FoxP3 transcription factor. Immunity. 2009;30:899. doi: 10.1016/j.immuni.2009.03.019.
    1. Messi M, Giacchetto I, Nagata K, Lanzavecchia A, Natoli G, Sallusto F. Memory and flexibility of cytokine gene expression as separable properties of human T(H)1 and T(H)2 lymphocytes. Nat Immunol. 2003;4:78. doi: 10.1038/ni872.
    1. Acosta-Rodriguez EV, Napolitani G, Lanzavecchia A, Sallusto F. Interleukins 1beta and 6 but not transforming growth factor-beta are essential for the differentiation of interleukin 17-producing human T helper cells. Nat Immunol. 2007;8:942. doi: 10.1038/ni1496.
    1. Laurence A, Tato CM, Davidson TS, Kanno Y, Chen Z, Yao Z, Blank RB, Meylan F, Siegel R, Hennighausen L, Shevach EM, O’shea JJ. Interleukin-2 signaling via STAT5 constrains T helper 17 cell generation. Immunity. 2007;26:371. doi: 10.1016/j.immuni.2007.02.009.
    1. Olson KE, Gendelman HE. Immunomodulation as a neuroprotective and therapeutic strategy for Parkinson's disease. Curr Opin Pharmacol. 2016;26:87. doi: 10.1016/j.coph.2015.10.006.
    1. von Euler Chelpin M, Vorup-Jensen T. Targets and mechanisms in prevention of Parkinson’s disease through immunomodulatory treatments. Scand J Immunol. 2017;85:321. doi: 10.1111/sji.12542.
    1. Baba Y, Kuroiwa A, Uitti RJ, Wszolek ZK, Yamada T. Alterations of T-lymphocyte populations in Parkinson disease. Parkinsonism Relat Disord. 2005;11:493. doi: 10.1016/j.parkreldis.2005.07.005.
    1. Bas J, Calopa M, Mestre M, Molleví DG, Cutillas B, Ambrosio S, Buendia E. Lymphocyte populations in Parkinson’s disease and in rat models of parkinsonism. J Neuroimmunol. 2001;113:146. doi: 10.1016/S0165-5728(00)00422-7.
    1. Stevens CH, Rowe D, Morel-Kopp MC, Orr C, Russell T, Ranola M, Ward C, Halliday GM. Reduced T helper and B lymphocytes in Parkinson's disease. J Neuroimmunol. 2012;252:95. doi: 10.1016/j.jneuroim.2012.07.015.
    1. van den Broek T, JAM B, van Wijk F. The full spectrum of human naive T cells. Nat Rev Immunol. 2018;18(6):363–373. doi: 10.1038/s41577-018-0001-y.
    1. Trinchieri G. Interleukin-10 production by effector T cells: Th1 cells show self control. J Exp Med. 2007;204:239. doi: 10.1084/jem.20070104.
    1. Oestreich KJ, Weinmann AS. Transcriptional mechanisms that regulate T helper 1 cell differentiation. Curr Opin Immunol. 2012;24:191. doi: 10.1016/j.coi.2011.12.004.
    1. Chen Z, Laurence A, O'Shea JJ. Signal transduction pathways and transcriptional regulation in the control of Th17 differentiation. Semin Immunol. 2007;19:400. doi: 10.1016/j.smim.2007.10.015.
    1. Ho IC, Tai TS, Pai SY. GATA3 and the T-cell lineage: essential functions before and after T-helper-2-cell differentiation. Nat Rev Immunol. 2009;9:125. doi: 10.1038/nri2476.
    1. Maier E, Duschl A, Horejs-Hoeck J. STAT6-dependent and -independent mechanisms in Th2 polarization. Eur J Immunol. 2012;42:2827. doi: 10.1002/eji.201242433.
    1. Lu L, Barbi J, Pan F. The regulation of immune tolerance by FOXP3. Nat Rev Immunol. 2017;17:703. doi: 10.1038/nri.2017.75.
    1. Won HY, Hwang ES. Transcriptional modulation of regulatory T cell development by novel regulators NR4As. Arch Pharm Res. 2016;39:1530. doi: 10.1007/s12272-016-0803-z.
    1. 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. J Immunol. 2013;190:5048. doi: 10.4049/jimmunol.1203121.
    1. Olson KE, Kosloski-Bilek LM, Anderson KM, Diggs BJ, Clark BE, Gledhill JM, Jr, Shandler SJ, Mosley RL, Gendelman HE. Selective VIP receptor agonists facilitate immune transformation for dopaminergic neuroprotection in MPTP-intoxicated mice. J Neurosci. 2015;35:16463. doi: 10.1523/JNEUROSCI.2131-15.2015.
    1. Saunders JA, Estes KA, Kosloski LM, Allen HE, Dempsey KM, Torres-Russotto DR, Meza JL, Santamaria PM, Bertoni JM, Murman DL, Ali HH, Standaert DG, Mosley RL, Gendelman HE. CD4+ regulatory and effector/memory T cell subsets profile motor dysfunction in Parkinson's disease. J NeuroImmune Pharmacol. 2012;7:927. doi: 10.1007/s11481-012-9402-z.
    1. Liu Z, Huang Y, Cao BB, Qiu YH, Peng YP. Th17 cells induce dopaminergic neuronal death via LFA-1/ICAM-1 interaction in a mouse model of Parkinson’s disease. Mol Neurobiol. 2017;54:7762. doi: 10.1007/s12035-016-0249-9.
    1. Cosentino M, Fietta AM, Ferrari M, Rasini E, Bombelli R, Carcano E, Saporiti F, Meloni F, Marino F, Lecchini S. Human CD4+CD25+ regulatory T cells selectively express tyrosine hydroxylase and contain endogenous catecholamines subserving an autocrine/paracrine inhibitory functional loop. Blood. 2007;109:632. doi: 10.1182/blood-2006-01-028423.
    1. Reynolds AD, Stone DK, Mosley RL, Gendelman HE. Nitrated {alpha}-synuclein-induced alterations in microglial immunity are regulated by CD4+ T cell subsets. J Immunol. 2009;182:4137. doi: 10.4049/jimmunol.0803982.
    1. Fung TC, Olson CA, Hsiao EY. Interactions between the microbiota, immune and nervous systems in health and disease. Nat Neurosci. 2017;20:145. doi: 10.1038/nn.4476.
    1. Scheperjans F, Aho V, Pereira PA, Koskinen K, Paulin L, Pekkonen E, Haapaniemi E, Kaakkola S, Eerola-Rautio J, Pohja M, Kinnunen E, Murros K, Auvinen P. Gut microbiota are related to Parkinson’s disease and clinical phenotype. Mov Disord. 2015;30:350. doi: 10.1002/mds.26069.
    1. Larsen JM. The immune response to Prevotella bacteria in chronic inflammatory disease. Immunology. 2017;151:363. doi: 10.1111/imm.12760.
    1. de Azevedo MS, Innocentin S, Dorella FA, Rocha CS, Mariat D, Pontes DS, Miyoshi A, Azevedo V, Langella P, Chatel JM. Immunotherapy of allergic diseases using probiotics or recombinant probiotics. J Appl Microbiol. 2013;115:319. doi: 10.1111/jam.12174.
    1. Klingelhoefer L, Reichmann H. Pathogenesis of Parkinson disease--the gut-brain axis and environmental factors. Nat Rev Neurol. 2015;11:625. doi: 10.1038/nrneurol.2015.197.
    1. Sulzer D, Alcalay RN, Garretti F, Cote L, Kanter E, Agin-Liebes J, Liong C, McMurtrey C, Hildebrand WH, Mao X, Dawson VL, Dawson TM, Oseroff C, Pham J, Sidney J, Dillon MB, Carpenter C, Weiskopf D, Phillips E, Mallal S, Peters B, Frazier A, Lindestam Arlehamn CS, Sette A. T cells from patients with Parkinson's disease recognize α-synuclein peptides. Nature. 2017;546:656. doi: 10.1038/nature22815.
    1. Lin CH, Lin JW, Liu YC, Chang CH, Wu RM. Risk of Parkinson’s disease following severe constipation: a nationwide population-based cohort study. Parkinsonism Relat Disord. 2014;20:1371. doi: 10.1016/j.parkreldis.2014.09.026.
    1. Khalif IL, Quigley EM, Konovitch EA, Maximova ID. Alterations in the colonic flora and intestinal permeability and evidence of immune activation in chronic constipation. Dig Liver Dis. 2005;37:838. doi: 10.1016/j.dld.2005.06.008.
    1. Mittal S, Bjørnevik K, Im DS, Flierl A, Dong X, Locascio JJ, Abo KM, Long E, Jin M, Xu B, Xiang YK, Rochet JC, Engeland A, Rizzu P, Heutink P, Bartels T, Selkoe DJ, Caldarone BJ, Glicksman MA, Khurana V, Schüle B, Park DS, Riise T, Scherzer CR. β2-Adrenoreceptor is a regulator of the α-synuclein gene driving risk of Parkinson’s disease. Science. 2017;357:891. doi: 10.1126/science.aaf3934.
    1. Elenkov IJ, Wilder RL, Chrousos GP, Vizi ES. The sympathetic nerve—an integrative interface between two supersystems: the brain and the immune system. Pharmacol Rev. 2000;52:595.
    1. Marino F, Cosentino M. Adrenergic modulation of immune cells: an update. Amino Acids. 2013;45:55–71. doi: 10.1007/s00726-011-1186-6.
    1. Scanzano A, Cosentino M. Adrenergic regulation of innate immunity: a review. Front Pharmacol. 2015;6:171. doi: 10.3389/fphar.2015.00171.
    1. Guereschi MG, Araujo LP, Maricato JT, Takenaka MC, Nascimento VM, Vivanco BC, Reis VO, Keller AC, Brum PC, Basso AS. Beta2-adrenergic receptor signaling in CD4+ Foxp3+ regulatory T cells enhances their suppressive function in a PKA-dependent manner. Eur J Immunol. 2013;43:1001. doi: 10.1002/eji.201243005.

Source: PubMed

3
S'abonner