Skip to main content
Erschienen in: Journal of Neuroinflammation 1/2022

Open Access 01.12.2022 | Research

α-Synuclein-containing erythrocytic extracellular vesicles: essential contributors to hyperactivation of monocytes in Parkinson’s disease

verfasst von: Zongran Liu, Robin Barry Chan, Zhijian Cai, Xiaodan Liu, Yufeng Wu, Zhenwei Yu, Tao Feng, Ying Yang, Jing Zhang

Erschienen in: Journal of Neuroinflammation | Ausgabe 1/2022

Abstract

Background

Immune system dysfunction, including higher levels of peripheral monocytes and inflammatory cytokines, is an important feature of Parkinson’s disease (PD) pathogenesis, although the mechanism underlying the process remains to be investigated. In the central nervous system, it is well-known that α-synuclein (α-syn), a key protein involved in PD, activates microglia potently, and it is also reported that α-syn exists in the peripheral system, especially in erythrocytes or red blood cells (RBC) at exceedingly high concentration. The current study focused on the possibility that RBC-derived α-syn mediates the sensitization of peripheral monocytes in PD patients.

Methods

The hyperactivation of monocytes was assessed quantitatively by measuring mRNA levels of typical inflammatory cytokines (including IL-1β, IL-6 and TNF-α) and protein levels of secreted inflammatory cytokines (including pro-inflammatory cytokines: IL-1β, IL-6, TNF-α, IL-8, IFN-γ, IL-2, and IL-12p70 and anti-inflammatory cytokines: IL-4, IL-10, and IL-13). Western blot, nanoparticle tracking analysis and electron microscopy were used to characterize RBC-derived extracellular vesicles (RBC-EVs). Inhibitors of endocytosis and leucine-rich repeat kinase 2 (LRRK2), another key protein involved in PD, were used to investigate how these two factors mediated the process of monocyte sensitization by RBC-EVs.

Results

Increased inflammatory sensitization of monocytes was observed in PD patients and PD model mice. We found that α-syn-containing RBC-EVs isolated from PD model mice or free form oligomeric α-syn induced the inflammatory sensitization of THP-1 cells, and demonstrated that endocytosis was a requirement for this pathophysiological pathway. Furthermore, the hyperactivation of THP-1 cells induced by RBC-EVs was associated with increased LRRK2 production and kinase activity. The phenomenon of inflammatory sensitization of human monocytes and increased LRRK2 were also observed by the treatment of RBC-EVs isolated from PD patients.

Conclusions

Our data provided new insight into how hyperactivation of monocytes occurs in PD patients, and identified the central role played by α-syn-containing RBC-EVs in this process.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12974-022-02413-1.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
PD
Parkinson’s disease
α-syn
α-Synuclein
CNS
Central nervous system
EV
Extracellular vesicle
RBC-EVs
RBC-derived extracellular vesicles
BBB
Blood–brain barrier
LRRK2
Leucine-rich repeat kinase 2

Background

Parkinson’s disease (PD) is a neurodegenerative disease clinically characterized by motor, e.g., resting tremor, bradykinesia, rigidity, and postural instability [1, 2], and non-motor symptoms [3]. The main pathological features of PD include the loss of dopaminergic neurons in the substantia nigra and the presence of Lewy bodies in surviving neurons [3, 4]. It has been well-demonstrated that α-synuclein (α-syn), one of the major components in Lewy bodies, exists not only in the central nervous system (CNS), but is also widespread in blood, especially in erythrocytes or red blood cells (RBCs) [5, 6]. Previously, we have demonstrated that RBCs are capable of secreting α-syn-containing extracellular vesicles (EVs) and these RBC-derived EVs (RBC-EVs) can cross the blood–brain barrier (BBB) and deposit α-syn in astrocytes and microglia [7, 8], resulting in astrocytic dysfunction and microglial activation.
In addition to impairment of motor and non-motor functions and loss of dopamine neurons, inflammation in the CNS is a salient feature of PD pathogenesis [911]. Although the role of neuroinflammation is not clear, microglial activation coupled with increased inflammatory cytokines and infiltration of immune cells in the CNS have been reported in PD [1219]. Besides CNS manifestations, PD patients also frequently experience clinical manifestations that are closely linked to dysfunction of the peripheral immune system [20]. For example, in the serum of PD patients, higher levels of inflammatory cytokines have been observed, indicating the existence of peripheral inflammation [2123]. In addition, the peripheral blood monocytes, enriched in PD patients compared to healthy controls, can secrete more inflammatory cytokines after external stimulation of bacterial endotoxin lipopolysaccharide (LPS), suggesting hyperactivation of the immune system in PD patients [24, 25]. Interestingly, basal and LPS-induced levels of cyto/chemokines in peripheral blood mononuclear cells are significantly correlated with the severity of PD symptoms [26]. Yet, the mechanisms underlying peripheral inflammation remain to be characterized.
Monocytes, a subset of circulating white blood cells, are the precursors of tissue macrophages and dendritic cells (DCs), which are equipped with high phagocytic and antigen-presenting capabilities and take part in the initial innate immune response [27]. It has been shown that oligomeric α-syn can activate microglia with the production of pro-inflammatory mediators [28, 29]. Because heteromeric and oligomeric α-syn are highly abundant in RBCs [3035] and α-syn containing RBC-EVs isolated from PD patients can pass through the BBB and activate microglia [8], it is possible that hyperactivation of monocytes in PD is associated with oligomeric α-syn containing RBC-EVs.
Mutations in the Leucine-rich repeat kinase 2 (LRRK2) gene have been identified as the most common genetic cause for familial PD [36]. Recent studies have discovered that hyperactivated wild-type LRRK2 may also play a role in idiopathic PD, although the mechanism involved is not entirely known [37, 38]. LRRK2 is highly expressed in monocytes, and its kinase activity is associated with immune cell activation [39, 40]. It is likely that the LRRK2 pathway takes part in the hyperactivation of monocytes in PD.
In this study, we tested the hypothesis that α-syn-containing RBC-EVs can induce inflammatory sensitization of monocytes in PD and investigated the molecular mechanisms by which this pathogenic phenomenon occurs.

Methods

Human subjects and clinical sample collection

The study was approved by the Institutional Review Board of Beijing Tiantan Hospital, Capital Medical University. In this study, whole blood samples from 15 patients with PD and 9 age- and sex-matched healthy controls were collected from Tiantan Hospital. All subjects underwent evaluations including medical history, physical and neurological examinations, laboratory tests, and neuropsychological assessments. Briefly, all PD patients met UK PD Society Brain Bank clinical diagnostic criteria for PD. Control subjects were in good health without any signs or symptoms suggesting cognitive impairment or neurological disease. All participants underwent detailed informed consent procedures. Demographic information is listed in Table 1 for all subjects.
Table 1
Characteristics of the clinical cohort of blood samples for RBC-EVs collection and monocyte isolation
Purpose
RBC-EVs collection
Monocyte isolation
Healthy control
PD
Healthy control
PD
Number
3
5
6
10
Sex (female/male)
1/2
2/3
3/3
5/5
Age (mean ± SD)
66 ± 6.24
64 ± 9.51
65.17 ± 4.96
68.1 ± 4.09
UPDRS3 (mean ± SD)
n/a
50.2 ± 7.79
n/a
51.5 ± 7.12

Animals

All animal procedures were approved in accordance with Chinese Guidelines for the ethical review of laboratory animal welfare (LA2020056). PD A53T double transgenic homozygous mice (dbl-PAC-Tg (SNCAA53T); Snca−/−) and SNCA-KO α-syn homozygous knockout mice (B6; 129X1-Sncatm1Rosl/J) were purchased from Jackson laboratory and kept on a 12-h light–dark cycle with ad libitum food and water. Age-matched wild-type mice (WT) were selected as the control group. In this study, 7-month-old, male mice were employed for the isolation of circulating monocytes and RBC-EVs.

RBC-EVs isolation

RBC-EVs were isolated from cultured human and mouse RBCs according to a previous article with minor modifications [8]. Briefly, human and mouse RBCs were separated from whole blood and cultured in RPMI1640 culture medium containing 25 mM HEPES at 37 °C with a humidified atmosphere of 5% CO2 for 48 h. Then, culture medium was collected and centrifuged at 1500×g for 10 min to remove RBCs, followed by filtering the supernatant through 0.22-μm filters. Subsequent filtrate was centrifuged in an ultracentrifuge (Beckmann) at 140,000 × g for 2 h to collect EVs. The collected pellets were washed with ice cold PBS and centrifuged again at 140,000×g for 2 h. Finally, EVs were resuspended and collected using ice cold PBS for further experiments.

Cell culture

THP-1 cells (ATCC #TIB-202) were maintained in RPMI1640 medium with 2 mM L-glutamine and 25 mM HEPES, supplemented with 10% fetal bovine serum.
For immune sensitization experiments, THP-1 cells were cultured with 200 μg RBC-EVs from different sources for 24 h (baseline) and then stimulated with 200 ng/mL bacterial lipopolysaccharide (LPS; Sigma, L2630; ≥ 500,000 Endotoxin Units/mg) for 24 h. THP-1 cells were collected for total RNA extraction and culture media were collected for inflammatory cytokines detection, at baseline and after LPS treatment.
For endocytosis inhibitor experiments, THP-1 cells were cultured with inhibitors for 4 h, followed by the addition of RBC-EVs. After 24-h incubation, 200 ng/mL LPS was applied for THP-1 stimulation for 24 h. Total RNA and culture media were collected for inflammatory cytokines detection.

Monocyte isolation

Circulating monocytes in mice and human blood were isolated by peripheral blood monocytes cells separation medium (Solarbio, P5290, P5230, Beijing, China) and maintained in RPMI1640 medium with 2 mM L-glutamine and 25 mM HEPES, supplemented with 10% fetal bovine serum.
For immune sensitization experiments, circulating monocytes were cultured with 200 μg RBC-EVs from different groups for 24 h (baseline) and then stimulated with 200 ng/mL bacterial LPS for 24 h. Monocytes were collected for total RNA extraction and culture media were collected for inflammatory cytokines detection, at baseline and after LPS treatment.

Electron microscopy

RBC-EVs isolated from different species were dropped onto the grid and then negatively stained with 2% phosphotungstic acid. The morphology of RBC-EVs was acquired by transmission electron microscopy (JEM1400 PLUS).

EVs labelling

RBC-EVs isolated from cultured WT mice were resuspended in 1 mL PBS buffer at particle concentration of 2 × 1010 /mL. Subsequently, 1 mM DiI or DiO stock solution (Coolaber, SL7910, SL7920) was added into the RBC-EV suspension at final concentration of 10 μM to label the RBC-EV for 30 min at room temperature. To remove the excess DiI or DiO dye, the labelled RBC-EV suspension was filtered thrice through Amicon® Ultra-centrifugal filter devices (cutoff MW 30 kDa, Millipore Corporation, Billerica, MA, USA). Finally, the labelled RBC-EVs were collected for further experiments or stored at –80 °C.

Nanoparticle tracking analysis

The particle concentration and size distribution of RBC-EVs were detected by NTA instrument (NS300; Nanosight). RBC-EVs were first diluted by filtered PBS at 1:100 to optimize the number of particles and injected into the sample sink. The movement of each particle was video captured in triplicate of 60 s. Analysis was performed by NTA 3.2 software (Nanosight, Amesbury, UK).

Real-time quantitative PCR

Total RNA was isolated from THP-1 cells and monocytes by TRIZOL™ reagent (Invitrogen, 15,596,018) according to the manufacturer's instruction. Following the extraction of RNA, cDNA was reverse transcribed from 2 mg total RNA by RT Master Mix Kit with gDNase (MCE, HY-K0511). The mRNA level of target gene in acquired cDNA was determined by real-time PCR with PowerUp SYBR® Green Master Mix (applied biosystem, A25742). ACTB was used as the internal reference for normalization. The primers used are presented in Table 2. All results were analyzed using 2−△△Ct and were presented as mean ± SEM.
Table 2
Primers used in qPCR
Gene
Forward primer (5′ to 3′)
Reverse primer (5′ to 3′)
Il1b
TGGACCTTCCAGGATGAGGACA
GTTCATCTCGGAGCCTGTAGTG
Il6
TACCACTTCACAAGTCGGAGGC
CTGCAAGTGCATCATCGTTGTTC
Tnf
GGTGCCTATGTCTCAGCCTCTT
GCCATAGAACTGATGAGAGGGAG
Actb
CATTGCTGACAGGATGCAGAAGG
TGCTGGAAGGTGGACAGTGAGG
IL1b
CCACAGACCTTCCAGGAGAATG
GTGCAGTTCAGTGATCGTACAGG
IL6
AGACAGCCACTCACCTCTTCAG
TTCTGCCAGTGCCTCTTTGCTG
TNF
CTCTTCTGCCTGCTGCACTTTG
ATGGGCTACAGGCTTGTCACTC
LRRK2
AGCAAGGGACAGGCTGAAGTTG
GCAGGCTTTGCGTTGCTTCTCA
ACTB
CACCATTGGCAATGAGCGGTTC
AGGTCTTTGCGGATGTCCACGT

Western blotting

Proteins were extracted from THP-1 cells and monocytes using cell lysis buffer (RIPA, HARVEYBIO, C1503) containing protease inhibitor cocktail (Sigma) and phosphatase inhibitor PhosSTOP™ (Roche, 4,906,837,001). BCA Protein Assay Kit (Applygen, P1511) was applied to determining the protein concentration of RBC-EVs and THP-1 monocytes according to the manufacturer's instruction. A total of 20 μg RBC-EVs or 30 μg THP-1 lysates were loaded to 10% PAGE gels for electrophoresis. Separated proteins were blotted to PVDF membrane (Merck Millipore, 4515). Then the membranes were blocked in TBST buffer (Applygen, B1009) containing 5% BSA (Amresco, A-0332) for 1 h at room temperature and incubated with diluted primary antibody overnight at 4 °C. Antibodies used included Alix (1:1000, Merck, Abc40), TSG101 (1:1000, abcam, ab133586), LRRK2 (1:5000, Abcam, ab133474), p-Rab10 (1:1000, Abcam, ab230261), Rab10 (1:1000, Abcam, ab237703), and β-actin (1:1000, Zsgb-bio, TA-09). The membranes were washed three times by TBST buffer on the second day, followed by incubation with secondary antibody for 1 h at RT. Protein bands were developed by enhanced chemiluminescence reagents (Millipore, WBKLS0100) and detected by Chemi doc™ Imaging System (Bio-Rad, Hercules CA, USA).

Flow cytometry for RBC-EV uptake assay

THP-1 monocytes pre-treated with or without endocytosis inhibitors for 30 min were incubated with 2 × 109 DiO-labelled RBC-EVs for another 4 h. Then, monocytes were collected and washed for analysis using BD FACS Calibur Flow Cytometer (BD Biosciences, Franklin Lakes NJ, USA) to determine the uptake of RBC-EVs in monocytes. Endocytosis inhibitors included Me-β-CD (200 μM, MCE, HY-101461), Amiloride (1 mM, MCE, HY-B0285A), Dynasore (50 μM, Selleck, S8047), and Nocodazole (40 ng/mL, MCE, HY-13520). The inhibition rate was calculated as the following equation:
$$\text{Inhibition rate}\left(\text{\%}\right)\text{=} \, \text{1} - \frac{\text{The number of monocytes containing DiO signal with inhibitors}}{\text{The number of monocytes containing DiO signal with vehicle} \, {\text{control}}}$$

Laser confocal microscopy for RBC-EV endocytosis assay

2 × 109 Dil labelled RBC-EVs were added into THP-1 monocytes pre-treated with or without endocytosis inhibitors for 30 min and incubated for 4 h. The cell nucleus was stained by Hoechst33258 (Solarbio) for 15 min. Images were acquired using Leica TCS SP8 confocal system.

Meso scale discovery multiplexed immunoassays

The detection and quantification for total and aggregated α-syn were based on our previously developed and validated assay [35]. Briefly, antibody against α-syn, MJFR-1 (ab138501, Abcam, only recognizes humanized α-syn) and antibody against conformation specific α-syn filaments, MJFR-14 (ab209538, Abcam, recognizes the structure of α-syn oligomers from both mouse and human species) were, respectively, biotinylated, linker conjugated, and coated onto standard 96-well U-Plex plates (Meso scale discovery). After washing three times with 150 μl Washing Buffer, 50 µl of diluted sample (diluted by Diluent 35 at dilution ratio of 1:5) and calibrator (recombinant α-syn, Sino biological, and oligomeric α-syn, Proteos) were loaded to the immunoassay plate, which was incubated overnight at 4 °C on a shaker at 600 rpm. Next, the plate was washed three times using washing buffer followed by the addition of sulfo-TAG-labelled anti-α-syn antibody (BD42) and incubated at room temperature for 1 h on a shaker at 600 rpm. Finally, 150 μL 2 × read buffer T was added into each well and plates were analyzed in a Quickplex SQ 120 (MSD, USA). Data analysis was performed with the MSD Discovery Workbench 3.0 Data Analysis Toolbox. Total α-syn concentrations were normalized to the total protein levels in RBC-EV, and are shown in unit of pg (total α-syn)/mg (total protein). Oligomeric α-syn concentrations were normalized to the corresponding total α-syn levels, and are shown in units of pg (oligo α-syn)/mg (total protein).
The inflammatory cytokines in culture medium were detected by 10-Vplex plates (containing IFN-γ, IL-10, IL-12p70, IL-13, IL-1β, IL-2, IL-4, IL-6, IL-8, and TNFα, Meso scale discovery) according to the manufacturer's instruction. Plates were analyzed in a Quickplex SQ 120 (MSD, USA). Data analysis was performed with the MSD Discovery Workbench 3.0 Data Analysis Toolbox.

Statistical analysis

Statistical analyses were performed using Prism 9.0 (GraphPad, USA). The statistical significance was assessed by t test or ANOVA analysis, followed with Tukey–Kramer’s post-hoc test, Dunn’s post-hoc test or Bonferroni’s post hoc test for multiple comparisons.

Results

Sensitization of monocytes in PD

To investigate the possibility of hyperactivation of monocytes in PD, we started with examining the expression levels of typical inflammatory cytokines of the monocytes isolated from PD patients and healthy controls at baseline and following 24 h LPS stimulation. The purity of CD14 + monocytes was around 84% (Fig. 1A). The mRNA levels of typical pro-inflammatory cytokines, IL-1β, IL-6 and TNF-α, were significantly higher in monocytes of PD patients than healthy controls (Fig. 1B). The protein levels of released inflammatory cytokines (including pro-inflammatory cytokines: IL-1β, IL-6, TNF-α, IL-8, IFN-γ, IL-2, and IL-12p70 and anti-inflammatory cytokines: IL-4, IL-10, and IL-13) were measured by a commonly utilized MSD platform. In the absence of external stimuli, most of the detected inflammatory cytokine levels, except for IL-8, IL-2, IL-4 and IL-13, were higher in the monocytes of PD patients than healthy controls (Fig. 1C, D). To further evaluate the sensitization of monocytes in PD patients, monocytes were stimulated by LPS; following 24 h stimulation, the mRNA levels of IL1b, IL6 and TNF and the protein levels of pro-inflammatory cytokines, especially IL-1β, IL-6, TNF-α and IL-2 became significantly higher in monocytes of PD patients than healthy controls (Fig. 1B, C), indicating the hyperactivation of monocytes in PD patients. The concentration of inflammatory cytokines in monocytes after LPS stimulation was normalized to the concentration of inflammatory cytokines in monocytes at resting state. As shown in Additional file 1: Fig. S1, LPS stimulation can elicit increased production of pro-inflammatory cytokines (IL-1β, TNF-α and IL-2) in monocytes of PD patients, which further indicated increased immune sensitization in PD. To explore the specific mechanism regulating the hyperactivation of monocytes in PD, we analyzed monocytes obtained from the dbl-PAC-Tg (SNCAA53T) and Snca−/− double transgenic PD A53T model mice (A53T mice) [41, 42]. The purity of CD14 + mouse monocytes was approximately 75% (Fig. 1E). Subsequently, both mRNA and protein expression levels of typical pro-inflammatory cytokines, IL-1β, IL-6, and TNF-α, were examined in monocytes derived from A53T mice and WT mice. At resting state, the production of all three cytokines was higher in monocytes of A53T mice than WT mice (except for protein level of IL-1β). Following 24 h LPS stimulation, significant increases in all three pro-inflammatory cytokines were observed in the monocytes from A53T mice, compared with WT mice (Fig. 1F, G), indicating that monocytes were hyperactivated in A53T mice.

Characterization of RBC-EVs

We previously demonstrated that α-syn containing RBC-EVs isolated from PD patients can pass through the BBB and activate the microglia [8]. To explore potential mechanisms involved in sensitizing monocytes, RBC-EVs were isolated from cultured RBCs of the A53T, SNCA knockout (SNCA-KO) and WT mice and characterized by Western blot, NTA and TEM. As shown in Fig. 2A, the typical markers of EVs, including Alix and TSG101, were enriched in RBC-EVs from A53T, SNCA-KO and WT mice. The size distribution and particle concentration of the RBC-EVs evaluated by NTA were mainly between 70 to 200 nm in diameter (Fig. 2B). Under TEM, RBC-EVs displayed a cup-shaped bilayer membrane structure (Fig. 2C). When the level of total α-syn in EVs was measured with a well-established protocol [35], the concentration of α-syn in WT and SNCA-KO RBC EVs was below detection limit, while the mean concentration of α-syn in A53T RBC-EVs was 487.67 pg/mg. Oligomeric α-syn contained in RBC-EVs was also significantly higher in A53T mice than WT and SNCA-KO mice (A53T: 324.8 pg/mg; WT: 27.35 pg/mg; SNCA-KO: 0 pg/mg).

α-Syn-containing RBC-EVs sensitized monocytes potently

To explore the possibility of immune regulation of A53T RBC-EVs on monocytes, THP-1, a human leukemia monocytic cell line extensively used to study monocyte functions [43], was selected initially as the experimental model. As shown in Fig. 3A, the mRNA levels of IL1b, IL6 and TNF in THP-1 cells treated with RBC-EVs derived from A53T mice were significantly elevated compared to those measured in THP-1 cells treated with vehicle control, while no significant increase existed in THP-1 cells treated with WT RBC-EVs or SNCA KO RBC-EVs. Alterations in the concentration of pro-inflammatory cytokines released in the culture media paralleled the mRNA results (Fig. 3B, blue bars), suggesting that A53T RBC-EVs significantly activated the THP-1 cells. Furthermore, after pre-treatment with RBC-EVs, the THP-1 cells were stimulated for 24 h with LPS to elicit an additional inflammatory response (Fig. 3A–C, red bars). These results suggest that α-syn might be a key mediator in the immune sensitization of monocytes. The hypothesis was further substantiated by exposing THP-1 cells to free oligomeric α-syn (Fig. 4) or monomeric α-syn (Additional file 1: Fig. S2), demonstrating that oligomeric α-syn significantly and dose-dependently hyperactivated monocytes.

Endocytosis required for sensitization of monocytes induced by A53T RBC-EVs

To further probe the mechanisms involved in monocyte sensitization, four types of endocytosis inhibitors, specifically Me-β-CD, Amiloride, Dynasore and Nocodazole [4447], were used to test whether internalization of RBC-EVs is necessary. By flow cytometry, we found that the RBC-EVs entered into THP-1 cells readily but this process was significantly inhibited by a 30-min treatment of four endocytosis inhibitors to different extents, with 15.7%, 7.6%, 34.4% and 33.7% inhibition for Me-β-CD, Amiloride, Dynasore and Nocodazole, respectively (Fig. 5A–F). These results were confirmed by confocal microscopy showing that abundant DiI labelled RBC-EVs can be detected in THP-1 cells treated with blank vehicle, while DiI signal was mostly confined to the plasma membrane in Amiloride treated cells, and barely observed in THP-1 cells treated with Dynasore and Nocodazole. For Me-β-CD treatment group, slight DiI signals can be detected in THP-1 cells (Fig. 5H). Next, we analyzed the effect of the four endocytosis inhibitors on monocyte hyperactivation and found that Dynasore itself significantly induced the production of pro-inflammatory cytokines TNF-α and IL-8 (Additional file 1: Fig. S3), likely attributable to the fact that Dynasore can activate the NF-κb pathway and promote the production of some pro-inflammatory cytokines [44]. Thus, Dynasore was excluded in the next series of experiments. To test the hypothesis that endocytosis of RBC-EVs is required to induce sensitization of monocytes, we pre-treated THP-1 cells with A53T RBC-EVs alone, or co-treated with Nocodazole, Me-β-CD or Amiloride for 24 h, followed by LPS stimulation. Measurement of the cytokines released by THP-1 cells revealed that Nocodazole and Me-β-CD co-treatment significantly inhibited the elevated inflammatory response of THP-1 cells induced by A53T RBC-EVs (Fig. 5I, J). To a lesser extent, Amiloride also had an inhibitory effect on THP-1 sensitization (Additional file 1: Fig. S4). To further investigate the role of α-syn in this process, we examined endocytosed oligomeric α-syn levels in THP-1 cells. As observed in Fig. 5G, around 30 pg/ml oligomeric α-syn was detected in THP-1 cells co-treated with A53T RBC-EVs and vehicle control, while no oligomeric α-syn was detectable in THP-1 cells treated with vehicle control alone, indicating the delivery of α-syn oligomers from RBC-EVs to the THP-1 cells. Compared with A53T RBC-EVs and vehicle control co-treatment group, oligomeric α-syn was hardly detected in THP-1 cells co-treated with A53T RBC-EVs and Nocodazole or with A53T RBC-EVs and Me-β-CD, indicating that Nocodazole and Me-β-CD could largely abolish the delivery of α-syn oligomers into the THP-1 cells. Taken together, these results indicate that α-syn-containing RBC-EVs can sensitize monocytes and this process is mediated by endocytosis.

LRRK2 involved in monocyte sensitization induced by A53T RBC-EVs

Next, we investigated whether LRRK2, a kinase enzyme known to be critical to immune regulation of PD patients, and its substrate Rab10, are involved in monocyte sensitization induced by A53T RBC-EVs. As shown in Fig. 6A, B, the levels of LRRK2 and phosphorylated Rab10 (p-Rab10) were significantly higher in A53T mouse monocytes compared to WT monocytes. THP-1 cells cultured with A53T RBC-EVs, but not WT RBC-EVs, also resulted in increased levels of LRRK2 and p-Rab10 (Fig. 6C–G). An inhibitor of LRRK2, Mli2 [48, 49], was used to determine if LRRK2 activity is essential in hyperactivation of monocytes. As observed in Fig. 6H–J, the treatment of Mli2 blocked increased mRNA levels of IL1b, IL6, and TNF and protein levels of pro-inflammatory cytokines (such as IL-1β, IL-6, IL-8 and TNF-α) in THP-1 cells treated with A53T RBC-EVs at both baseline and LPS stimulated state. These results indicate that hyperactivation of monocytes induced by A53T RBC-EVs is mediated at least in part by LRRK2 kinase activity.

Confirmation of sensitization of monocytes in PD patients

To confirm the significance of the above results in human disease, RBC-EVs were isolated from cultured RBCs from 5 PD patients and 3 neurologically normal control subjects (Table 1 and Fig. 7A–C). Consistent with earlier observations [30, 31, 34, 35], total and oligomeric α-syn contained in RBC-EVs were significantly higher in PD patients compared to healthy controls (HCs) (Fig. 7D, E). In addition, consistent with A53T mouse experiments, THP-1 cells treated with the RBC-EVs derived from PD patients demonstrated significantly elevated mRNA levels of IL1b, IL6 and  TNF (Fig. 7F), and concentration of inflammatory cytokines (Fig. 7G, H) in THP-1 cells at resting state. Following 24 h LPS stimulation, the mRNA and protein levels of inflammatory cytokines further significantly increased in THP-1 cells pre-treated with PD RBC-EVs (Fig. 7F–H). In addition, LRRK2 and p-Rab10 expression were significantly increased in THP-1 cells cultured with PD RBC-EVs, while LRRK2 and p-Rab10 were only slightly increased in THP-1 cells treated with HC RBC-EVs (Fig. 7I–K). These results indicate that RBC-EVs derived from PD patients are able to induce inflammatory sensitization of monocytes, with a process likely involving LRRK2 activation.
To further confirm that PD RBC-EVs can induce the hyperactivation of monocytes, monocytes from healthy controls were isolated and treated with HC RBC-EVs or PD RBC-EVs. As shown in Fig. 8, PD RBC-EVs can significantly increase the mRNA levels of IL1b, IL6 and TNF, and the protein levels of pro-inflammatory cytokines, such as IL-1β, IL-6, TNF-α, and IFN-γ in monocytes. Following 24 h LPS stimulation, the mRNA levels of IL1b, IL6 and TNF and the protein level of pro-inflammatory cytokines, especially IL-1β, IL-6 and TNF-α, became significantly higher in monocytes pre-treated with PD RBC-EVs than HC RBC-EVs.

Discussion

The major observations of the current study centered on hyperactivation of monocytes of PD patients and related mechanisms. In our previous study, we showed that oligomeric α-syn containing RBC-EVs can traffic across the BBB via an adsorptive-mediated transcytosis process and activate microglia in the CNS [8]. Considering the direct exposure of RBC-EVs to circulating monocytes, we hypothesized that RBC-EVs may be involved in the hyperactivation of monocytes in PD patients whose inflammatory markers are commonly elevated in blood [21]. We demonstrated that monocytes obtained from PD patients and A53T mice both exhibited higher production of mRNA of inflammatory cytokine genes and higher levels of secreted cytokines at baseline state compared to controls, and produced significantly more inflammatory cytokines after LPS stimulation, which was consistent with several previous reports [24, 25]. More importantly, RBC-EVs derived from both PD patients and A53T mice, but not healthy controls and SNCA KO mice, were able to induce hyperactivation of THP-1 cells and monocytes in vitro. Moreover, at resting or stimulated state, the alterations of inflammatory cytokines in monocytes treated with PD RBC-EVs were quite consistent with the results in THP-1 cells, indicating that α-syn containing RBC-EVs likely participate in the hyperactivation of monocytes in PD patients.
In whole blood, α-syn species exist either in free forms or in association with EVs. In particular, RBCs can be viewed as a reservoir of α-syn and are able to secrete EVs harboring different structural forms of α-syn [6, 7, 35, 50]. Previously, we have demonstrated that RBC-EVs derived from PD patients and PD mice contain more pathological α-syn and can cross the BBB and deposit α-syn in astrocytes and microglia, resulting in astrocytic dysfunction and microglial activation [7, 8]. To characterize the pathophysiological relevance of α-syn in RBC-EVs-driven hyperactivation of monocytes, the immune regulation of oligomeric α-syn containing RBC-EVs on monocytes was investigated at resting baseline and LPS stimulated states. In the absence of LPS stimulation, A53T RBC-EVs could hyperactivate the resting THP-1 cells, resulting in increased cytokine expression and release. Following LPS stimulation, the production of inflammatory cytokines in THP-1 cells further increased after pre-treatment with α-syn containing A53T RBC-EVs or PD RBC-EVs. These results demonstrate that the oligomeric α-syn containing RBC-EVs are involved in hyperactivation of monocytes in PD. We further directly examined the effects of both monomeric and oligomeric α-syn on monocytes and found that oligomeric α-syn can activate THP-1 cells much more potently than monomeric α-syn. More importantly, A53T RBC-EVs elicited a more potent immune response in monocytes compared to the free form of oligomeric α-syn. By normalizing LPS stimulated to the baseline state levels, the fold change of pro-inflammatory cytokines (e.g., IL-1β, IL-8, and TNF-α) in THP-1 cells pre-treated with A53T RBC-EVs was significantly higher than THP-1 cells pre-treated with free oligomeric α-syn (Additional file 1: Fig. S5). Previous studies reported that free oligomeric α-syn can bind toll-like receptor 2 or 4 (TLR2 or TLR4) on microglia to induce the release of pro-inflammatory cytokines in the CNS [5153], suggesting that a similar mechanism may be potentially involved in the interaction between oligomeric α-syn containing RBC-EVs with monocytes. However, the current study suggests that it is the α-syn containing RBC-EVs, not the free forms of α-syn, that most effectively sensitize monocytes. Thus, α-syn containing RBC-EVs may be an essential factor to the hyperactivation of monocytes in PD.
We tested the hypothesis that RBC-EV-driven immune sensitization involves endocytosis using three different endocytosis inhibitors (Me-β-CD, Nocodazole and Amiloride) and found that Me-β-CD and Nocodazole had potent inhibitory effects. Me-β-CD is a cholesterol depleting agent that can inhibit both clathrin- and caveolae-mediated endocytosis [33, 54, 55], while Nocodazole is a microtubule polymerization blocker that prevents the trafficking of early endosomes to lysosomes [44, 56]. On the other hand, Amiloride, which inhibits macropinocytosis by lowering submembranous pH by interfering Na+/H+ exchange [45], had a weaker inhibitory effect on immune sensitization. Taken together, these data suggest that receptor-mediated endocytosis plays a key role in modulating oligomeric α-syn-containing RBC-EVs sensitization of monocytes and it is likely to involve interaction with receptors that are enriched in clathrin- and/or caveolae coated pits. Of note, our study does not exclude the possibility that α-syn-containing RBC-EVs also interact with a surface receptor, e.g., TLR, to mediate the sensitization of monocytes. This is particularly important to the engagement of surface receptors that are critical to the downstream activation of LRRK2.
Mutations in LRRK2 gene are the most common cause of familial types of PD and polymorphisms in LRRK2 have been shown to modulate risk for sporadic PD [3, 4, 36, 57]. In the periphery, LRRK2 is expressed in myeloid cells, including monocytes and macrophages [58, 59]. Recent works have revealed that PD patients exhibited increased LRRK2 protein expression and kinase activity in hyperactivated immune cells [3740, 60, 61], which is consistent with our observations in both PD patients and A53T mice monocytes. We showed that treatment of THP-1 monocytes with PD RBC-EVs also resulted in increased expression and kinase activity of LRRK2. Furthermore, by inhibiting the kinase activity of LRRK2 with Mli-2 [48, 49], the hyperactivation of monocytes was significantly relieved, indicating the crucial role of LRRK2 in the PD RBC-EVs-driven immune dysregulation process. This data set is consistent with reports, where knocking out LRRK2 or inhibiting the kinase activity of LRRK2 can attenuate the neuroinflammation induced by α-syn [58, 62]. Although the endocytosis of α-syn containing RBC-EVs and increased kinase activity of LRRK2 have been proved to be highly associated with the hyperactivation of PD monocytes, the precise mechanisms involved in the hyperactivation of PD monocytes, including those beyond endocytosis of RBC-EVs and kinase of LRRK2, need to be investigated further.
In the context of neurological disorders, such as PD, it is thought that the initial immune response may be protective, but chronic inflammation can contribute to the onset and progression of disease [63]. In our study, anti-inflammatory cytokines (such as IL-10), were increased along with pro-inflammatory cytokines (such as IL-1β, IL-6, and TNF-α) in PD patients, a phenomenon also reported in other investigations [21, 26, 64]. The underlying mechanism is yet to be defined further, but one potential reason could be a compensatory response of monocytes while stressed. Indeed, this protective mechanism to counter increased peripheral inflammation and promote immune responses to cope with exposure to pathological α-syn (e.g., activation of B-cells, production of IFN-γ and phagocytotic ability of monocytes), has been proposed previously [65, 66]. In addition, of note, in our study, the extent of increase in pro-inflammatory cytokines was much higher than anti-inflammatory cytokines (Figs. 1, 3, 7 and 8).

Conclusions

Our findings demonstrated that RBC-EVs containing pathological oligomeric α-syn causes hyperactivation of circulating monocytes, in a process that requires receptor mediated endocytosis and LRRK2 activation. Our data provides a novel perspective for immune dysregulation in sporadic PD and highlights LRRK2 inhibition in peripheral monocytes as a potential therapeutic target for ameliorating PD pathogenesis.

Acknowledgements

We deeply appreciate the participants for their generous donation of samples.

Declarations

The study was approved by the Institutional Review Board of Beijing Tiantan Hospital, Capital Medical University and all participants underwent detailed informed consent procedures.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Anhänge
Literatur
1.
Zurück zum Zitat Jankovic J. Parkinson’s disease: clinical features and diagnosis. J Neurol Neurosurg Psychiatry. 2008;79:368–76.PubMedCrossRef Jankovic J. Parkinson’s disease: clinical features and diagnosis. J Neurol Neurosurg Psychiatry. 2008;79:368–76.PubMedCrossRef
2.
Zurück zum Zitat Shulman JM, De Jager PL, Feany MB. Parkinson’s disease: genetics and pathogenesis. Annu Rev Pathol. 2011;6:193–222.PubMedCrossRef Shulman JM, De Jager PL, Feany MB. Parkinson’s disease: genetics and pathogenesis. Annu Rev Pathol. 2011;6:193–222.PubMedCrossRef
3.
Zurück zum Zitat Poewe W, Seppi K, Tanner CM, Halliday GM, Brundin P, Volkmann J, Schrag AE, Lang AE. Parkinson disease. Nat Rev Dis Primers. 2017;3:17013.PubMedCrossRef Poewe W, Seppi K, Tanner CM, Halliday GM, Brundin P, Volkmann J, Schrag AE, Lang AE. Parkinson disease. Nat Rev Dis Primers. 2017;3:17013.PubMedCrossRef
4.
Zurück zum Zitat Nussbaum RL, Ellis CE. Alzheimer’s disease and Parkinson’s disease. N Engl J Med. 2003;348:1356–64.PubMedCrossRef Nussbaum RL, Ellis CE. Alzheimer’s disease and Parkinson’s disease. N Engl J Med. 2003;348:1356–64.PubMedCrossRef
5.
Zurück zum Zitat Barbour R, Kling K, Anderson JP, Banducci K, Cole T, Diep L, Fox M, Goldstein JM, Soriano F, Seubert P, Chilcote TJ. Red blood cells are the major source of alpha-synuclein in blood. Neurodegener Dis. 2008;5:55–9.PubMedCrossRef Barbour R, Kling K, Anderson JP, Banducci K, Cole T, Diep L, Fox M, Goldstein JM, Soriano F, Seubert P, Chilcote TJ. Red blood cells are the major source of alpha-synuclein in blood. Neurodegener Dis. 2008;5:55–9.PubMedCrossRef
6.
Zurück zum Zitat Shi M, Zabetian CP, Hancock AM, Ginghina C, Hong Z, Yearout D, Chung KA, Quinn JF, Peskind ER, Galasko D, et al. Significance and confounders of peripheral DJ-1 and alpha-synuclein in Parkinson’s disease. Neurosci Lett. 2010;480:78–82.PubMedPubMedCentralCrossRef Shi M, Zabetian CP, Hancock AM, Ginghina C, Hong Z, Yearout D, Chung KA, Quinn JF, Peskind ER, Galasko D, et al. Significance and confounders of peripheral DJ-1 and alpha-synuclein in Parkinson’s disease. Neurosci Lett. 2010;480:78–82.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Sheng L, Stewart T, Yang D, Thorland E, Soltys D, Aro P, Khrisat T, Xie Z, Li N, Liu Z, et al. Erythrocytic α-synuclein contained in microvesicles regulates astrocytic glutamate homeostasis: a new perspective on Parkinson’s disease pathogenesis. Acta Neuropathol Commun. 2020;8:102.PubMedPubMedCentralCrossRef Sheng L, Stewart T, Yang D, Thorland E, Soltys D, Aro P, Khrisat T, Xie Z, Li N, Liu Z, et al. Erythrocytic α-synuclein contained in microvesicles regulates astrocytic glutamate homeostasis: a new perspective on Parkinson’s disease pathogenesis. Acta Neuropathol Commun. 2020;8:102.PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Matsumoto J, Stewart T, Sheng L, Li N, Bullock K, Song N, Shi M, Banks WA, Zhang J. Transmission of alpha-synuclein-containing erythrocyte-derived extracellular vesicles across the blood-brain barrier via adsorptive mediated transcytosis: another mechanism for initiation and progression of Parkinson’s disease? Acta Neuropathol Commun. 2017;5:71.PubMedPubMedCentralCrossRef Matsumoto J, Stewart T, Sheng L, Li N, Bullock K, Song N, Shi M, Banks WA, Zhang J. Transmission of alpha-synuclein-containing erythrocyte-derived extracellular vesicles across the blood-brain barrier via adsorptive mediated transcytosis: another mechanism for initiation and progression of Parkinson’s disease? Acta Neuropathol Commun. 2017;5:71.PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Blandini F. Neural and immune mechanisms in the pathogenesis of Parkinson’s disease. J Neuroimmune Pharmacol. 2013;8:189–201.PubMedCrossRef Blandini F. Neural and immune mechanisms in the pathogenesis of Parkinson’s disease. J Neuroimmune Pharmacol. 2013;8:189–201.PubMedCrossRef
11.
Zurück zum Zitat Hunot S, Hirsch EC: Neuroinflammatory processes in Parkinson's disease. Ann Neurol 2003, 53 Suppl 3:S49–58; discussion S58–60. Hunot S, Hirsch EC: Neuroinflammatory processes in Parkinson's disease. Ann Neurol 2003, 53 Suppl 3:S49–58; discussion S58–60.
12.
Zurück zum Zitat Barcia C, Ros CM, Annese V, Gómez A, Ros-Bernal F, Aguado-Llera D, Martínez-Pagán ME, de Pablos V, Fernandez-Villalba E, Herrero MT. IFN-γ signaling, with the synergistic contribution of TNF-α, mediates cell specific microglial and astroglial activation in experimental models of Parkinson’s disease. Cell Death Dis. 2012;3:e379.PubMedPubMedCentralCrossRef Barcia C, Ros CM, Annese V, Gómez A, Ros-Bernal F, Aguado-Llera D, Martínez-Pagán ME, de Pablos V, Fernandez-Villalba E, Herrero MT. IFN-γ signaling, with the synergistic contribution of TNF-α, mediates cell specific microglial and astroglial activation in experimental models of Parkinson’s disease. Cell Death Dis. 2012;3:e379.PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat McGeer PL, Itagaki S, Boyes BE, McGeer EG. Reactive microglia are positive for HLA-DR in the substantia nigra of Parkinson’s and Alzheimer’s disease brains. Neurology. 1988;38:1285–91.PubMedCrossRef McGeer PL, Itagaki S, Boyes BE, McGeer EG. Reactive microglia are positive for HLA-DR in the substantia nigra of Parkinson’s and Alzheimer’s disease brains. Neurology. 1988;38:1285–91.PubMedCrossRef
14.
Zurück zum Zitat McGeer PL, Schwab C, Parent A, Doudet D. Presence of reactive microglia in monkey substantia nigra years after 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine administration. Ann Neurol. 2003;54:599–604.PubMedCrossRef McGeer PL, Schwab C, Parent A, Doudet D. Presence of reactive microglia in monkey substantia nigra years after 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine administration. Ann Neurol. 2003;54:599–604.PubMedCrossRef
15.
Zurück zum Zitat Wolf SA, Boddeke HW, Kettenmann H. Microglia in Physiology and Disease. Annu Rev Physiol. 2017;79:619–43.PubMedCrossRef Wolf SA, Boddeke HW, Kettenmann H. Microglia in Physiology and Disease. Annu Rev Physiol. 2017;79:619–43.PubMedCrossRef
16.
Zurück zum Zitat Banks WA, Robinson SM. Minimal penetration of lipopolysaccharide across the murine blood-brain barrier. Brain Behav Immun. 2010;24:102–9.PubMedCrossRef Banks WA, Robinson SM. Minimal penetration of lipopolysaccharide across the murine blood-brain barrier. Brain Behav Immun. 2010;24:102–9.PubMedCrossRef
17.
Zurück zum Zitat Brochard V, Combadière B, Prigent A, Laouar Y, Perrin A, Beray-Berthat V, Bonduelle O, Alvarez-Fischer D, Callebert J, Launay JM, et al. Infiltration of CD4+ lymphocytes into the brain contributes to neurodegeneration in a mouse model of Parkinson disease. J Clin Invest. 2009;119:182–92.PubMed Brochard V, Combadière B, Prigent A, Laouar Y, Perrin A, Beray-Berthat V, Bonduelle O, Alvarez-Fischer D, Callebert J, Launay JM, et al. Infiltration of CD4+ lymphocytes into the brain contributes to neurodegeneration in a mouse model of Parkinson disease. J Clin Invest. 2009;119:182–92.PubMed
18.
Zurück zum Zitat Garretti F, Agalliu D, LindestamArlehamn CS, Sette A, Sulzer D. Autoimmunity in Parkinson’s disease: the role of α-synuclein-specific T cells. Front Immunol. 2019;10:303.PubMedPubMedCentralCrossRef Garretti F, Agalliu D, LindestamArlehamn CS, Sette A, Sulzer D. Autoimmunity in Parkinson’s disease: the role of α-synuclein-specific T cells. Front Immunol. 2019;10:303.PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Campos-Acuña J, Elgueta D, Pacheco R. T-cell-driven inflammation as a mediator of the gut-brain axis involved in Parkinson’s disease. Front Immunol. 2019;10:239.PubMedPubMedCentralCrossRef Campos-Acuña J, Elgueta D, Pacheco R. T-cell-driven inflammation as a mediator of the gut-brain axis involved in Parkinson’s disease. Front Immunol. 2019;10:239.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Tan EK, Chao YX, West A, Chan LL, Poewe W, Jankovic J. Parkinson disease and the immune system - associations, mechanisms and therapeutics. Nat Rev Neurol. 2020;16:303–18.PubMedCrossRef Tan EK, Chao YX, West A, Chan LL, Poewe W, Jankovic J. Parkinson disease and the immune system - associations, mechanisms and therapeutics. Nat Rev Neurol. 2020;16:303–18.PubMedCrossRef
21.
Zurück zum Zitat Qin XY, Zhang SP, Cao C, Loh YP, Cheng Y. Aberrations in peripheral inflammatory cytokine levels in Parkinson disease: a systematic review and meta-analysis. JAMA Neurol. 2016;73:1316–24.PubMedCrossRef Qin XY, Zhang SP, Cao C, Loh YP, Cheng Y. Aberrations in peripheral inflammatory cytokine levels in Parkinson disease: a systematic review and meta-analysis. JAMA Neurol. 2016;73:1316–24.PubMedCrossRef
22.
Zurück zum Zitat Chen H, O’Reilly EJ, Schwarzschild MA, Ascherio A. Peripheral inflammatory biomarkers and risk of Parkinson’s disease. Am J Epidemiol. 2008;167:90–5.PubMedCrossRef Chen H, O’Reilly EJ, Schwarzschild MA, Ascherio A. Peripheral inflammatory biomarkers and risk of Parkinson’s disease. Am J Epidemiol. 2008;167:90–5.PubMedCrossRef
23.
Zurück zum Zitat Mogi M, Togari A, Kondo T, Mizuno Y, Komure O, Kuno S, Ichinose H, Nagatsu T. Caspase activities and tumor necrosis factor receptor R1 (p55) level are elevated in the substantia nigra from parkinsonian brain. J Neural Transm (Vienna). 2000;107:335–41.CrossRef Mogi M, Togari A, Kondo T, Mizuno Y, Komure O, Kuno S, Ichinose H, Nagatsu T. Caspase activities and tumor necrosis factor receptor R1 (p55) level are elevated in the substantia nigra from parkinsonian brain. J Neural Transm (Vienna). 2000;107:335–41.CrossRef
24.
Zurück zum Zitat Grozdanov V, Bousset L, Hoffmeister M, Bliederhaeuser C, Meier C, Madiona K, Pieri L, Kiechle M, McLean PJ, Kassubek J, et al. Increased immune activation by pathologic alpha-synuclein in Parkinson’s disease. Ann Neurol. 2019;86:593–606.PubMedCrossRef Grozdanov V, Bousset L, Hoffmeister M, Bliederhaeuser C, Meier C, Madiona K, Pieri L, Kiechle M, McLean PJ, Kassubek J, et al. Increased immune activation by pathologic alpha-synuclein in Parkinson’s disease. Ann Neurol. 2019;86:593–606.PubMedCrossRef
25.
Zurück zum Zitat Grozdanov V, Bliederhaeuser C, Ruf WP, Roth V, Fundel-Clemens K, Zondler L, Brenner D, Martin-Villalba A, Hengerer B, Kassubek J, et al: Inflammatory dysregulation of blood monocytes in Parkinson's disease patients. Acta Neuropathol 2014, 128:651–663. Grozdanov V, Bliederhaeuser C, Ruf WP, Roth V, Fundel-Clemens K, Zondler L, Brenner D, Martin-Villalba A, Hengerer B, Kassubek J, et al: Inflammatory dysregulation of blood monocytes in Parkinson's disease patients. Acta Neuropathol 2014, 128:651–663.
26.
Zurück zum Zitat Reale M, Iarlori C, Thomas A, Gambi D, Perfetti B, Di Nicola M, Onofrj M. Peripheral cytokines profile in Parkinson’s disease. Brain Behav Immun. 2009;23:55–63.PubMedCrossRef Reale M, Iarlori C, Thomas A, Gambi D, Perfetti B, Di Nicola M, Onofrj M. Peripheral cytokines profile in Parkinson’s disease. Brain Behav Immun. 2009;23:55–63.PubMedCrossRef
28.
Zurück zum Zitat Zhang W, Wang TG, Pei Z, Miller DS, Wu XF, Block ML, Wilson B, Zhang WQ, Zhou Y, Hong JS, Zhang J. Aggregated alpha-synuclein activates microglia: a process leading to disease progression in Parkinson’s disease. FASEB J. 2005;19:533–42.PubMedCrossRef Zhang W, Wang TG, Pei Z, Miller DS, Wu XF, Block ML, Wilson B, Zhang WQ, Zhou Y, Hong JS, Zhang J. Aggregated alpha-synuclein activates microglia: a process leading to disease progression in Parkinson’s disease. FASEB J. 2005;19:533–42.PubMedCrossRef
29.
Zurück zum Zitat Du XY, Xie XX, Liu RT. The role of α-synuclein oligomers in Parkinson’s Disease. Int J Mol Sci. 2020;21:1.CrossRef Du XY, Xie XX, Liu RT. The role of α-synuclein oligomers in Parkinson’s Disease. Int J Mol Sci. 2020;21:1.CrossRef
30.
Zurück zum Zitat Wang X, Yu S, Li F, Feng T. Detection of α-synuclein oligomers in red blood cells as a potential biomarker of Parkinson’s disease. Neurosci Lett. 2015;599:115–9.PubMedCrossRef Wang X, Yu S, Li F, Feng T. Detection of α-synuclein oligomers in red blood cells as a potential biomarker of Parkinson’s disease. Neurosci Lett. 2015;599:115–9.PubMedCrossRef
31.
Zurück zum Zitat Daniele S, Frosini D, Pietrobono D, Petrozzi L, Lo Gerfo A, Baldacci F, Fusi J, Giacomelli C, Siciliano G, Trincavelli ML, et al. α-Synuclein heterocomplexes with β-amyloid are increased in red blood cells of Parkinson’s disease patients and correlate with disease severity. Front Mol Neurosci. 2018;11:53.PubMedPubMedCentralCrossRef Daniele S, Frosini D, Pietrobono D, Petrozzi L, Lo Gerfo A, Baldacci F, Fusi J, Giacomelli C, Siciliano G, Trincavelli ML, et al. α-Synuclein heterocomplexes with β-amyloid are increased in red blood cells of Parkinson’s disease patients and correlate with disease severity. Front Mol Neurosci. 2018;11:53.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Araki K, Sugawara K, Hayakawa EH, Ubukawa K, Kobayashi I, Wakui H, Takahashi N, Sawada K, Mochizuki H, Nunomura W. The localization of α-synuclein in the process of differentiation of human erythroid cells. Int J Hematol. 2018;108:130–8.PubMedCrossRef Araki K, Sugawara K, Hayakawa EH, Ubukawa K, Kobayashi I, Wakui H, Takahashi N, Sawada K, Mochizuki H, Nunomura W. The localization of α-synuclein in the process of differentiation of human erythroid cells. Int J Hematol. 2018;108:130–8.PubMedCrossRef
33.
Zurück zum Zitat Nakai M, Fujita M, Waragai M, Sugama S, Wei J, Akatsu H, Ohtaka-Maruyama C, Okado H, Hashimoto M. Expression of alpha-synuclein, a presynaptic protein implicated in Parkinson’s disease, in erythropoietic lineage. Biochem Biophys Res Commun. 2007;358:104–10.PubMedCrossRef Nakai M, Fujita M, Waragai M, Sugama S, Wei J, Akatsu H, Ohtaka-Maruyama C, Okado H, Hashimoto M. Expression of alpha-synuclein, a presynaptic protein implicated in Parkinson’s disease, in erythropoietic lineage. Biochem Biophys Res Commun. 2007;358:104–10.PubMedCrossRef
34.
Zurück zum Zitat Papagiannakis N, Koros C, Stamelou M, Simitsi AM, Maniati M, Antonelou R, Papadimitriou D, Dermentzaki G, Moraitou M, Michelakakis H, Stefanis L. Alpha-synuclein dimerization in erythrocytes of patients with genetic and non-genetic forms of Parkinson’s Disease. Neurosci Lett. 2018;672:145–9.PubMedCrossRef Papagiannakis N, Koros C, Stamelou M, Simitsi AM, Maniati M, Antonelou R, Papadimitriou D, Dermentzaki G, Moraitou M, Michelakakis H, Stefanis L. Alpha-synuclein dimerization in erythrocytes of patients with genetic and non-genetic forms of Parkinson’s Disease. Neurosci Lett. 2018;672:145–9.PubMedCrossRef
35.
Zurück zum Zitat Tian C, Liu G, Gao L, Soltys D, Pan C, Stewart T, Shi M, Xie Z, Liu N, Feng T, Zhang J. Erythrocytic alpha-Synuclein as a potential biomarker for Parkinson’s disease. Transl Neurodegener. 2019;8:15.PubMedPubMedCentralCrossRef Tian C, Liu G, Gao L, Soltys D, Pan C, Stewart T, Shi M, Xie Z, Liu N, Feng T, Zhang J. Erythrocytic alpha-Synuclein as a potential biomarker for Parkinson’s disease. Transl Neurodegener. 2019;8:15.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Tolosa E, Vila M, Klein C, Rascol O. LRRK2 in Parkinson disease: challenges of clinical trials. Nat Rev Neurol. 2020;16:97–107.PubMedCrossRef Tolosa E, Vila M, Klein C, Rascol O. LRRK2 in Parkinson disease: challenges of clinical trials. Nat Rev Neurol. 2020;16:97–107.PubMedCrossRef
37.
Zurück zum Zitat Bliederhaeuser C, Zondler L, Grozdanov V, Ruf WP, Brenner D, Melrose HL, Bauer P, Ludolph AC, Gillardon F, Kassubek J, et al. LRRK2 contributes to monocyte dysregulation in Parkinson’s disease. Acta Neuropathol Commun. 2016;4:123.PubMedPubMedCentralCrossRef Bliederhaeuser C, Zondler L, Grozdanov V, Ruf WP, Brenner D, Melrose HL, Bauer P, Ludolph AC, Gillardon F, Kassubek J, et al. LRRK2 contributes to monocyte dysregulation in Parkinson’s disease. Acta Neuropathol Commun. 2016;4:123.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Di Maio R, Hoffman EK, Rocha EM, Keeney MT, Sanders LH, De Miranda BR, Zharikov A, Van Laar A, Stepan AF, Lanz TA, et al. LRRK2 activation in idiopathic Parkinson’s disease. Sci Transl Med. 2018;10:eaar5429.PubMedPubMedCentralCrossRef Di Maio R, Hoffman EK, Rocha EM, Keeney MT, Sanders LH, De Miranda BR, Zharikov A, Van Laar A, Stepan AF, Lanz TA, et al. LRRK2 activation in idiopathic Parkinson’s disease. Sci Transl Med. 2018;10:eaar5429.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Raj T, Rothamel K, Mostafavi S, Ye C, Lee MN, Replogle JM, Feng T, Lee M, Asinovski N, Frohlich I, et al. Polarization of the effects of autoimmune and neurodegenerative risk alleles in leukocytes. Science. 2014;344:519–23.PubMedPubMedCentralCrossRef Raj T, Rothamel K, Mostafavi S, Ye C, Lee MN, Replogle JM, Feng T, Lee M, Asinovski N, Frohlich I, et al. Polarization of the effects of autoimmune and neurodegenerative risk alleles in leukocytes. Science. 2014;344:519–23.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Liu Z, Bryant N, Kumaran R, Beilina A, Abeliovich A, Cookson MR, West AB. LRRK2 phosphorylates membrane-bound Rabs and is activated by GTP-bound Rab7L1 to promote recruitment to the trans-Golgi network. Hum Mol Genet. 2018;27:385–95.PubMedCrossRef Liu Z, Bryant N, Kumaran R, Beilina A, Abeliovich A, Cookson MR, West AB. LRRK2 phosphorylates membrane-bound Rabs and is activated by GTP-bound Rab7L1 to promote recruitment to the trans-Golgi network. Hum Mol Genet. 2018;27:385–95.PubMedCrossRef
41.
Zurück zum Zitat Kuo YM, Li Z, Jiao Y, Gaborit N, Pani AK, Orrison BM, Bruneau BG, Giasson BI, Smeyne RJ, Gershon MD, Nussbaum RL. Extensive enteric nervous system abnormalities in mice transgenic for artificial chromosomes containing Parkinson disease-associated alpha-synuclein gene mutations precede central nervous system changes. Hum Mol Genet. 2010;19:1633–50.PubMedPubMedCentralCrossRef Kuo YM, Li Z, Jiao Y, Gaborit N, Pani AK, Orrison BM, Bruneau BG, Giasson BI, Smeyne RJ, Gershon MD, Nussbaum RL. Extensive enteric nervous system abnormalities in mice transgenic for artificial chromosomes containing Parkinson disease-associated alpha-synuclein gene mutations precede central nervous system changes. Hum Mol Genet. 2010;19:1633–50.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Cabin DE, Shimazu K, Murphy D, Cole NB, Gottschalk W, McIlwain KL, Orrison B, Chen A, Ellis CE, Paylor R, et al. Synaptic vesicle depletion correlates with attenuated synaptic responses to prolonged repetitive stimulation in mice lacking alpha-synuclein. J Neurosci. 2002;22:8797–807.PubMedPubMedCentralCrossRef Cabin DE, Shimazu K, Murphy D, Cole NB, Gottschalk W, McIlwain KL, Orrison B, Chen A, Ellis CE, Paylor R, et al. Synaptic vesicle depletion correlates with attenuated synaptic responses to prolonged repetitive stimulation in mice lacking alpha-synuclein. J Neurosci. 2002;22:8797–807.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Chanput W, Mes JJ, Wichers HJ. THP-1 cell line: an in vitro cell model for immune modulation approach. Int Immunopharmacol. 2014;23:37–45.PubMedCrossRef Chanput W, Mes JJ, Wichers HJ. THP-1 cell line: an in vitro cell model for immune modulation approach. Int Immunopharmacol. 2014;23:37–45.PubMedCrossRef
44.
Zurück zum Zitat Ailenberg M, Di Ciano-Oliveira C, Szaszi K, Dan Q, Rozycki M, Kapus A, Rotstein OD. Dynasore enhances the formation of mitochondrial antiviral signalling aggregates and endocytosis-independent NF-kappaB activation. Br J Pharmacol. 2015;172:3748–63.PubMedPubMedCentralCrossRef Ailenberg M, Di Ciano-Oliveira C, Szaszi K, Dan Q, Rozycki M, Kapus A, Rotstein OD. Dynasore enhances the formation of mitochondrial antiviral signalling aggregates and endocytosis-independent NF-kappaB activation. Br J Pharmacol. 2015;172:3748–63.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Koivusalo M, Welch C, Hayashi H, Scott CC, Kim M, Alexander T, Touret N, Hahn KM, Grinstein S. Amiloride inhibits macropinocytosis by lowering submembranous pH and preventing Rac1 and Cdc42 signaling. J Cell Biol. 2010;188:547–63.PubMedPubMedCentralCrossRef Koivusalo M, Welch C, Hayashi H, Scott CC, Kim M, Alexander T, Touret N, Hahn KM, Grinstein S. Amiloride inhibits macropinocytosis by lowering submembranous pH and preventing Rac1 and Cdc42 signaling. J Cell Biol. 2010;188:547–63.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Rosenbaum AI, Zhang G, Warren JD, Maxfield FR. Endocytosis of beta-cyclodextrins is responsible for cholesterol reduction in Niemann-Pick type C mutant cells. Proc Natl Acad Sci U S A. 2010;107:5477–82.PubMedPubMedCentralCrossRef Rosenbaum AI, Zhang G, Warren JD, Maxfield FR. Endocytosis of beta-cyclodextrins is responsible for cholesterol reduction in Niemann-Pick type C mutant cells. Proc Natl Acad Sci U S A. 2010;107:5477–82.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Tacheva-Grigorova SK, Santos AJ, Boucrot E, Kirchhausen T. Clathrin-mediated endocytosis persists during unperturbed mitosis. Cell Rep. 2013;4:659–68.PubMedCrossRef Tacheva-Grigorova SK, Santos AJ, Boucrot E, Kirchhausen T. Clathrin-mediated endocytosis persists during unperturbed mitosis. Cell Rep. 2013;4:659–68.PubMedCrossRef
48.
Zurück zum Zitat Fell MJ, Mirescu C, Basu K, Cheewatrakoolpong B, DeMong DE, Ellis JM, Hyde LA, Lin Y, Markgraf CG, Mei H, et al. MLi-2, a Potent, Selective, and Centrally Active Compound for Exploring the Therapeutic Potential and Safety of LRRK2 Kinase Inhibition. J Pharmacol Exp Ther. 2015;355:397–409.PubMedCrossRef Fell MJ, Mirescu C, Basu K, Cheewatrakoolpong B, DeMong DE, Ellis JM, Hyde LA, Lin Y, Markgraf CG, Mei H, et al. MLi-2, a Potent, Selective, and Centrally Active Compound for Exploring the Therapeutic Potential and Safety of LRRK2 Kinase Inhibition. J Pharmacol Exp Ther. 2015;355:397–409.PubMedCrossRef
49.
Zurück zum Zitat Volpicelli-Daley LA, Abdelmotilib H, Liu Z, Stoyka L, Daher JP, Milnerwood AJ, Unni VK, Hirst WD, Yue Z, Zhao HT, et al. G2019S-LRRK2 Expression Augments α-Synuclein Sequestration into Inclusions in Neurons. J Neurosci. 2016;36:7415–27.PubMedPubMedCentralCrossRef Volpicelli-Daley LA, Abdelmotilib H, Liu Z, Stoyka L, Daher JP, Milnerwood AJ, Unni VK, Hirst WD, Yue Z, Zhao HT, et al. G2019S-LRRK2 Expression Augments α-Synuclein Sequestration into Inclusions in Neurons. J Neurosci. 2016;36:7415–27.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat El-Agnaf OM, Salem SA, Paleologou KE, Curran MD, Gibson MJ, Court JA, Schlossmacher MG, Allsop D. Detection of oligomeric forms of alpha-synuclein protein in human plasma as a potential biomarker for Parkinson’s disease. Faseb j. 2006;20:419–25.PubMedCrossRef El-Agnaf OM, Salem SA, Paleologou KE, Curran MD, Gibson MJ, Court JA, Schlossmacher MG, Allsop D. Detection of oligomeric forms of alpha-synuclein protein in human plasma as a potential biomarker for Parkinson’s disease. Faseb j. 2006;20:419–25.PubMedCrossRef
51.
Zurück zum Zitat Dzamko N, Gysbers A, Perera G, Bahar A, Shankar A, Gao J, Fu Y, Halliday GM. Toll-like receptor 2 is increased in neurons in Parkinson’s disease brain and may contribute to alpha-synuclein pathology. Acta Neuropathol. 2017;133:303–19.PubMedCrossRef Dzamko N, Gysbers A, Perera G, Bahar A, Shankar A, Gao J, Fu Y, Halliday GM. Toll-like receptor 2 is increased in neurons in Parkinson’s disease brain and may contribute to alpha-synuclein pathology. Acta Neuropathol. 2017;133:303–19.PubMedCrossRef
52.
Zurück zum Zitat Harms AS, Kordower JH, Sette A, LindestamArlehamn CS, Sulzer D, Mach RH. Inflammation in Experimental Models of alpha-Synucleinopathies. Mov Disord. 2021;36:37–49.PubMedCrossRef Harms AS, Kordower JH, Sette A, LindestamArlehamn CS, Sulzer D, Mach RH. Inflammation in Experimental Models of alpha-Synucleinopathies. Mov Disord. 2021;36:37–49.PubMedCrossRef
53.
Zurück zum Zitat Collins LM, Toulouse A, Connor TJ, Nolan YM. Contributions of central and systemic inflammation to the pathophysiology of Parkinson’s disease. Neuropharmacology. 2012;62:2154–68.PubMedCrossRef Collins LM, Toulouse A, Connor TJ, Nolan YM. Contributions of central and systemic inflammation to the pathophysiology of Parkinson’s disease. Neuropharmacology. 2012;62:2154–68.PubMedCrossRef
54.
Zurück zum Zitat Rodal SK, Skretting G, Garred O, Vilhardt F, van Deurs B, Sandvig K. Extraction of cholesterol with methyl-beta-cyclodextrin perturbs formation of clathrin-coated endocytic vesicles. Mol Biol Cell. 1999;10:961–74.PubMedPubMedCentralCrossRef Rodal SK, Skretting G, Garred O, Vilhardt F, van Deurs B, Sandvig K. Extraction of cholesterol with methyl-beta-cyclodextrin perturbs formation of clathrin-coated endocytic vesicles. Mol Biol Cell. 1999;10:961–74.PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat Gratton SE, Ropp PA, Pohlhaus PD, Luft JC, Madden VJ, Napier ME, DeSimone JM. The effect of particle design on cellular internalization pathways. Proc Natl Acad Sci USA. 2008;105:11613–8.PubMedPubMedCentralCrossRef Gratton SE, Ropp PA, Pohlhaus PD, Luft JC, Madden VJ, Napier ME, DeSimone JM. The effect of particle design on cellular internalization pathways. Proc Natl Acad Sci USA. 2008;105:11613–8.PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Eguchi K, Taoufiq Z, Thorn-Seshold O, Trauner D, Hasegawa M, Takahashi T. Wild-type monomeric α-synuclein can impair vesicle endocytosis and synaptic fidelity via tubulin polymerization at the calyx of held. J Neurosci. 2017;37:6043–52.PubMedPubMedCentralCrossRef Eguchi K, Taoufiq Z, Thorn-Seshold O, Trauner D, Hasegawa M, Takahashi T. Wild-type monomeric α-synuclein can impair vesicle endocytosis and synaptic fidelity via tubulin polymerization at the calyx of held. J Neurosci. 2017;37:6043–52.PubMedPubMedCentralCrossRef
57.
Zurück zum Zitat Zimprich A, Biskup S, Leitner P, Lichtner P, Farrer M, Lincoln S, Kachergus J, Hulihan M, Uitti RJ, Calne DB, et al. Mutations in LRRK2 cause autosomal-dominant parkinsonism with pleomorphic pathology. Neuron. 2004;44:601–7.PubMedCrossRef Zimprich A, Biskup S, Leitner P, Lichtner P, Farrer M, Lincoln S, Kachergus J, Hulihan M, Uitti RJ, Calne DB, et al. Mutations in LRRK2 cause autosomal-dominant parkinsonism with pleomorphic pathology. Neuron. 2004;44:601–7.PubMedCrossRef
58.
Zurück zum Zitat Daher JP, Volpicelli-Daley LA, Blackburn JP, Moehle MS, West AB. Abrogation of α-synuclein-mediated dopaminergic neurodegeneration in LRRK2-deficient rats. Proc Natl Acad Sci USA. 2014;111:9289–94.PubMedPubMedCentralCrossRef Daher JP, Volpicelli-Daley LA, Blackburn JP, Moehle MS, West AB. Abrogation of α-synuclein-mediated dopaminergic neurodegeneration in LRRK2-deficient rats. Proc Natl Acad Sci USA. 2014;111:9289–94.PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Moehle MS, Webber PJ, Tse T, Sukar N, Standaert DG, DeSilva TM, Cowell RM, West AB. LRRK2 inhibition attenuates microglial inflammatory responses. J Neurosci. 2012;32:1602–11.PubMedPubMedCentralCrossRef Moehle MS, Webber PJ, Tse T, Sukar N, Standaert DG, DeSilva TM, Cowell RM, West AB. LRRK2 inhibition attenuates microglial inflammatory responses. J Neurosci. 2012;32:1602–11.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Cook DA, Kannarkat GT, Cintron AF, Butkovich LM, Fraser KB, Chang J, Grigoryan N, Factor SA, West AB, Boss JM, Tansey MG. LRRK2 levels in immune cells are increased in Parkinson’s disease. NPJ Parkinsons Dis. 2017;3:11.PubMedPubMedCentralCrossRef Cook DA, Kannarkat GT, Cintron AF, Butkovich LM, Fraser KB, Chang J, Grigoryan N, Factor SA, West AB, Boss JM, Tansey MG. LRRK2 levels in immune cells are increased in Parkinson’s disease. NPJ Parkinsons Dis. 2017;3:11.PubMedPubMedCentralCrossRef
61.
Zurück zum Zitat Sabatino JJ Jr, Probstel AK, Zamvil SS. B cells in autoimmune and neurodegenerative central nervous system diseases. Nat Rev Neurosci. 2019;20:728–45.PubMedCrossRef Sabatino JJ Jr, Probstel AK, Zamvil SS. B cells in autoimmune and neurodegenerative central nervous system diseases. Nat Rev Neurosci. 2019;20:728–45.PubMedCrossRef
62.
Zurück zum Zitat Daher JP, Abdelmotilib HA, Hu X, Volpicelli-Daley LA, Moehle MS, Fraser KB, Needle E, Chen Y, Steyn SJ, Galatsis P, et al. Leucine-rich Repeat Kinase 2 (LRRK2) Pharmacological Inhibition Abates α-Synuclein Gene-induced Neurodegeneration. J Biol Chem. 2015;290:19433–44.PubMedPubMedCentralCrossRef Daher JP, Abdelmotilib HA, Hu X, Volpicelli-Daley LA, Moehle MS, Fraser KB, Needle E, Chen Y, Steyn SJ, Galatsis P, et al. Leucine-rich Repeat Kinase 2 (LRRK2) Pharmacological Inhibition Abates α-Synuclein Gene-induced Neurodegeneration. J Biol Chem. 2015;290:19433–44.PubMedPubMedCentralCrossRef
63.
Zurück zum Zitat Passaro AP, Lebos AL, Yao Y, Stice SL. Immune response in neurological pathology: emerging role of central and peripheral immune crosstalk. Front Immunol. 2021;12:676621.PubMedPubMedCentralCrossRef Passaro AP, Lebos AL, Yao Y, Stice SL. Immune response in neurological pathology: emerging role of central and peripheral immune crosstalk. Front Immunol. 2021;12:676621.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Brodacki B, Staszewski J, Toczyłowska B, Kozłowska E, Drela N, Chalimoniuk M, Stepien A. Serum interleukin (IL-2, IL-10, IL-6, IL-4), TNFalpha, and INFgamma concentrations are elevated in patients with atypical and idiopathic parkinsonism. Neurosci Lett. 2008;441:158–62.PubMedCrossRef Brodacki B, Staszewski J, Toczyłowska B, Kozłowska E, Drela N, Chalimoniuk M, Stepien A. Serum interleukin (IL-2, IL-10, IL-6, IL-4), TNFalpha, and INFgamma concentrations are elevated in patients with atypical and idiopathic parkinsonism. Neurosci Lett. 2008;441:158–62.PubMedCrossRef
66.
Zurück zum Zitat Xu W, Roos A, Schlagwein N, Woltman AM, Daha MR, van Kooten C. IL-10-producing macrophages preferentially clear early apoptotic cells. Blood. 2006;107:4930–7.PubMedCrossRef Xu W, Roos A, Schlagwein N, Woltman AM, Daha MR, van Kooten C. IL-10-producing macrophages preferentially clear early apoptotic cells. Blood. 2006;107:4930–7.PubMedCrossRef
Metadaten
Titel
α-Synuclein-containing erythrocytic extracellular vesicles: essential contributors to hyperactivation of monocytes in Parkinson’s disease
verfasst von
Zongran Liu
Robin Barry Chan
Zhijian Cai
Xiaodan Liu
Yufeng Wu
Zhenwei Yu
Tao Feng
Ying Yang
Jing Zhang
Publikationsdatum
01.12.2022
Verlag
BioMed Central
Erschienen in
Journal of Neuroinflammation / Ausgabe 1/2022
Elektronische ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-022-02413-1

Weitere Artikel der Ausgabe 1/2022

Journal of Neuroinflammation 1/2022 Zur Ausgabe

Neu in den Fachgebieten Neurologie und Psychiatrie

Prämenstruelle Beschwerden mit Suizidrisiko assoziiert

04.06.2024 Suizidalität Nachrichten

Manche Frauen, die regelmäßig psychische und körperliche Symptome vor ihrer Menstruation erleben, haben ein deutlich erhöhtes Suizidrisiko. Jüngere Frauen sind besonders gefährdet.

Bei seelischem Stress sind Checkpoint-Hemmer weniger wirksam

03.06.2024 NSCLC Nachrichten

Wie stark Menschen mit fortgeschrittenem NSCLC von einer Therapie mit Immun-Checkpoint-Hemmern profitieren, hängt offenbar auch davon ab, wie sehr die Diagnose ihre psychische Verfassung erschüttert

Demenzkranke durch Antipsychotika vielfach gefährdet

Demenz Nachrichten

Der Einsatz von Antipsychotika gegen psychische und Verhaltenssymptome in Zusammenhang mit Demenzerkrankungen erfordert eine sorgfältige Nutzen-Risiken-Abwägung. Neuen Erkenntnissen zufolge sind auf der Risikoseite weitere schwerwiegende Ereignisse zu berücksichtigen.

Schlaganfall: frühzeitige Blutdrucksenkung im Krankenwagen ohne Nutzen

31.05.2024 Apoplex Nachrichten

Der optimale Ansatz für die Blutdruckkontrolle bei Patientinnen und Patienten mit akutem Schlaganfall ist noch nicht gefunden. Ob sich eine frühzeitige Therapie der Hypertonie noch während des Transports in die Klinik lohnt, hat jetzt eine Studie aus China untersucht.