Skip to main content

High-salt diet does not boost neuroinflammation and neurodegeneration in a model of α-synucleinopathy

Abstract

Aim

Pre-clinical studies in models of multiple sclerosis and other inflammatory disorders suggest that high-salt diet may induce activation of the immune system and potentiate inflammation. However, high-salt diet constitutes a common non-pharmacological intervention to treat autonomic problems in synucleinopathies such as Parkinson’s disease and multiple system atrophy. Since neuroinflammation plays an important pathogenic role in these neurodegenerative disorders, we asked here whether high-salt diet may aggravate the disease phenotype in a transgenic model of multiple system atrophy.

Methods

Nine-month-old PLP-hαSyn and matched wildtype mice received normal or high-salt diet for a period of 3 months. Behavioral, histological, and molecular analyses were performed to evaluate the effect of high-salt diet on motor decline, neuroinflammation, neurodegeneration, and α-synuclein accumulation in these mice.

Results

Brain subregion-specific molecular and histological analyses showed no deleterious effects of high-salt diet on the level of microglial activation. Moreover, neuroinflammation-related cytokines and chemokines, T cell recruitment or astrogliosis were unaffected by high-salt diet exposure. Behavioral testing showed no effect of diet on motor decline. High-salt diet was not related to the deterioration of neurodegeneration or α-synuclein accumulation in PLP-hαSyn mice.

Conclusions

Here, we demonstrate that high-salt diet does not aggravate neuroinflammation and neurodegeneration in PLP-hαSyn mice. Our findings discard a deleterious pro-neuroinflammatory effect of high-salt diet in multiple system atrophy.

Introduction

High-salt diet (HSD) has been associated with chronic inflammation [1], neuroinflammation [2, 3], and autoimmune diseases [4, 5]. In this regard, high-salt intake has been shown to affect immune cells and induce the differentiation of T helper (Th)17 cells with pathogenic phenotype [6, 7] which play an important role in the induction of neuroinflammation, CNS autoimmunity, and neurovascular and cognitive dysfunction [8,9,10,11]. The induction of pathogenic Th17 cells and their infiltration in the CNS by HSD has shown to potentiate neuroinflammation in experimental autoimmune encephalomyelitis (EAE), an animal model which mimics many features of multiple sclerosis (MS) [6, 9, 12, 13]. In this mouse model, HSD accelerates disease onset, augments its severity, and enhances blood-brain barrier (BBB) disruption and brain pathology [6, 7, 14, 15]. Moreover, in experimental models high salt also promotes pro-inflammatory phenotype of myeloid cells by enhancing cytokine production and polarization towards M1 phenotype, leading to an overall imbalance of immune homeostasis [14, 16, 17]. In accordance with these findings, a recent study demonstrated that increased sodium intake is associated with clinical disease exacerbation, augmented relapse risk, and increased magnetic resonance imaging (MRI) activity in MS patients [4]. However, larger clinical studies recently failed to show an association between salt intake and higher MS disease risk, progression, or activity [18, 19], thus the possible deleterious effect of HSD in MS is still under discussion.

Synucleinopathies such as Parkinson’s disease (PD) and multiple system atrophy (MSA) constitute neurodegenerative disorders characterized by autonomic failure, motor impairment and the abnormal accumulation of α-synuclein (α-syn) in the cytoplasm of either neurons (Lewy bodies, characteristics of PD) or in oligodendroglial cytoplasmic inclusions (GCIs, characteristics of MSA) [20,21,22,23]. The accumulation of α-syn in MSA brains is associated with microglial activation and neuroinflammation [24,25,26], demyelination [27, 28], and neurodegeneration [29]. MSA is subdivided into two motor subtypes depending on the main brain areas affected by the pathology, the parkinsonian variant (MSA-P) characterized by striatonigral degeneration (SND) and the cerebellar variant (MSA-C) characterized by olivopontocerebellar atrophy (OPCA), but autonomic failure is present in both variants [30,31,32]. At present, there are no disease-modifying therapies to stop disease progression and only mitigation of some symptoms is feasible [33]. In this regard, increased fluid and salt intake is often recommended by physicians to alleviate neurogenic orthostatic hypotension [34].

The association of HSD with pro-inflammatory disorders together with the experimental evidence showing its deleterious effects in different in vitro and in vivo models questions the use of HSD in MSA and PD patients. The fact that neuroinflammation and the immune system, especially microglial cells, play an important role in MSA and PD pathogenesis [24,25,26, 35,36,37] and the recent evidences of a possible involvement of Th17 cells in PD [38,39,40,41,42,43] support these concerns. In order to evaluate the pathological consequences of HSD in α-synucleinopathies we have analyzed its effects in the PLP-hαSyn transgenic mice model of MSA. Here, we show that high dietary intake of salt does not accelerate disease progression nor increase neuroinflammation, microglial activation, or neurodegeneration in MSA mice, suggesting that HSD may not have a pro-neuroinflammatory effect in this particular α-synucleinopathy.

Material and methods

Animals and treatments

Clinical and pathological features of MSA are recapitulated in the PLP-hαSyn mouse model. These mice overexpress human wildtype α-syn in oligodendrocytes under the myelin proteolipid protein (PLP) promoter [44] leading to the formation of GCI-like structures, autonomic failure, progressive SND, and motor impairment [44,45,46,47,48,49,50,51,52,53,54]. SND in the PLP-hαSyn mice is characterized by a reduction in the number of dopaminergic neurons in the substantia nigra pars compacta (SNc) [55] followed by a reduction of the density of dopaminergic terminals and medium spiny neurons (MSNs) in the striatum linked to significant motor decline [51]. Similar to the human pathology, PLP-hαSyn mice develop progressive microglial activation initially triggered by α-syn pathology [51, 53]. Our group has also previously shown that stressors relevant to human MSA, e.g., mitochondrial dysfunction [52] or proteolysis disruption [56], can aggravate the pathology in PLP-hαSyn mice towards full-blown MSA with spreading of GCI, SND, OPCA, and strong microglial activation. In summary, the PLP-hαSyn mouse model provides an important and relevant pre-clinical tool to study disease mechanisms of MSA progression downstream of the accumulation of α-syn in oligodendrocytes.

PLP-hαSyn and C57BL/6 N wildtype animals were kept under temperature-controlled pathogen-free conditions on a light/dark 12 h cycle. Nine-month-old PLP-hαSyn and wildtype animals were both randomized in 2 groups, one fed with normal food pellets (0.19% sodium; SSNIFF Spezialdiäten GmbH) and tap water, another fed with HSD (4% NaCl; SSNIFF Spezialdiäten GmbH) and tap water containing 1% NaCl as previously described [2, 6, 7]. After 3 months of treatment, the animals were sacrificed and brains were collected. Bodyweight of all animals was measured weekly throughout the treatment period (Additional file 1: Figure S1). Although PLP-hαSyn mice presented lower body weight compared to healthy control animals, no differences due to diet were observed within the 2 animal groups (Additional file 2: Figure S1). All the experiments were performed according to the ethical guidelines of the EU (Directive 2010/63/EU for animal experiments) and the Austrian Federal Ministry of Science and Research (permission BMFWF-66.011/0018-WF/v/3b/2015). All analyses were done by a researcher who was blinded to the treatment of the animals.

Stride length analysis

Stride length analysis was performed with DigiGait™ Imaging System (Mouse Specifics Inc.) as previously described [51, 56]. Briefly, mice were placed onto a transparent treadmill belt and gait of each mouse was recorded with a video camera placed below the belt. Stride length was analyzed with DigiGait Software 9.0 (Mouse Specific, USA) and expressed in cm.

Tissue processing and histology

Animals were perfused intracardially with phosphate-buffered saline (PBS, pH 7.4, Sigma) under deep thiopental anesthesia and brains were extracted. Hemibrains were post-fixed overnight in 4% paraformaldehyde (pH 7.4, Sigma) at 4 °C and then cryoprotected in 30% sucrose (in PBS). Finally, the brains were frozen using 2-Bodyweight (Sigma) and stored at − 80 °C. The brains were cut in 40 μm thick coronal sections using a freezing microtome (Leica) and stored free-floating in a cryoprotectant buffer at − 20 °C.

Immunohistological analyses

Free-floating sections were immunostained following standard protocols. Microglial activation was evaluated by immunofluorescence using antibodies against IBA1 (1:600, WAKO) and CD68 (1:200, R&D). In order to evaluate the level of SND, SNc sections were stained with anti-tyrosine hydroxylase (TH) antibody (1:1000, Millipore) and the number of dopaminergic (TH+) neurons was analyzed by stereological counting. Striatal sections were stained with anti-DARPP32 antibody (BD Bioscience; 1:2000) and the number medium spiny neurons (MSNs; DARPP32+) was quantified. OPCA in the cerebellum was evaluated by DARPP32 immunostaining of Purkinje cells (DARPP32+). To analyze the number of GCIs, representative sections including SNc, cerebellar white matter (CBWM), and motor cortex (M2) were stained with anti-phosphorylated α-syn antibody (pS129; 1:1000, Abcam). For immunofluorescence, suitable secondary anti-IgG antibodies conjugated with Alexa 488 or Alexa 594 (Life Technologies) were applied and coverslipped with mounting medium Fluromount-G (Southern Biotech). For immunohistochemistry, sections were incubated with biotinylated secondary antibodies followed by Vectastain ABC reagent (Vector Laboratories) and 3,3′-diaminobenzidine (Sigma) to visualize the binding sites. Stained sections were mounted on slides, dehydrated, and coverslipped with Entellan (Merck).

Image analyses

Neuroanatomy was assessed using a Mouse Brain Atlas. For microglial activation assessment, images were acquired with a fluorescence microscope (Leica DMI4000) and the positive area for IBA1 or CD68 was estimated using ImageJ (National Institutes of Health). Results are presented as percentage of IBA1 or CD68 area per section total area. Stereological analysis was performed using the Nikon E-800 microscope equipped with Nikon digital camera DXM1200 and Stereoinvestigator software (Microbrightfield Europe e.K) as described previously [56]. The number of TH+ neurons in the SNc and DARPP-32+ neurons in the striatum was measured by applying the optical fractionator workflow [51]. The density of GCIs and Purkinje cells (DARPP32+) were assessed with meander scan and is expressed in GCI/mm2 and DARPP32+ neurons/mm2 respectively.

RNA extraction and quantitative RT-qPCR

For molecular analyses, hemibrains were quickly dissected in the forebrain, midbrain, cerebellum, and brainstem, frozen in liquid nitrogen and stored at − 80 °C. RNA was extracted using TRIzol reagent (Life technologies) according to the manufacturer’s instructions. Tissue was homogenized with ULTRA-TURRAX T-8 basic tissueruptor (IKA) in the presence of TRIzol. RNA samples (3 μg) were retrotranscribed to cDNA using High-Capacity cDNA Reverse Transcription Kit (Applied-Biosystems). Real time PCR was performed in a 7500 Real-Time PCR Systems (Applied-Biosystems) using TaqMan™ Universal PCR Master Mix (Applied-Biosystems). Gapdh mRNA levels were estimated to normalize for mRNA input amounts. TaqMan probe sequences are available upon request. mRNA levels were obtained using the 2−ΔΔCt method and expressed as fold-change relative to the wildtype normal diet control group [57].

Cytokine/chemokine levels

Fresh frozen forebrain, midbrain, cerebellum, and brainstem were homogenized in Triton-X (TX) extraction buffer (50 mM Tris-base pH 7.6, 150 mM NaCl, 1% Triton-X-100, 2 mM EDTA) containing protease and phosphatase inhibitors. The lysates were centrifuged (16,000×g for 10 min at 4 °C) to remove debris and the supernatant was collected and stored at − 80 °C. Protein concentrations were determined with BCA Protein Assay Kit (Sigma). ProcartaPlex® Multiplex Immunoassay system (eBioscience, Waltham, MA USA) was used to simultaneously measure the concentration of different cytokines and chemokines. The same protein amount was loaded for all samples. Duplicates were performed per each sample and mean values were calculated for subsequent statistical analysis. Data are presented as pg cytokine/chemokine per mg total protein.

Dot blot analysis of soluble α-syn

Lysates obtained previously were ultra-centrifuged (100,000×g for 60 min at 4 °C) and the supernatant was collected and stored at − 80 °C. Equal amounts of protein (5 μg) per sample were spotted onto nitrocellulose membranes (GE Healthcare) and air-dried for 30 min. Membranes were incubated overnight at 4 °C in blocking buffer (PBS, pH 7.6, 0.1% Tween 20, 5% non-fat dry milk) with primary antibody against human α-syn (4B12; 1:1000, Genetex). Signal detection was performed using HRP-conjugated secondary antibodies and WesternBright Quantum kit (Advansta). Images were acquired using the Fusion FX system for western blot and gel imaging and quantified with FUSION CAPT V16.09b software (Vilber Lourmat).

Statistical analyses

All statistical analyses were conducted using the software Graph-Pad Prism 7 (Graphpad Software). The mean ± S.E.M was used to present the results. Two-way analysis of variance (ANOVA) with post hoc Bonferroni test was used to compare the groups if not indicated otherwise. A p value < 0.05 was considered statistically significant.

Results

High-salt diet causes partial upregulation of genes linked to microglial and astroglial activation without changes at protein level in PLP-hαSyn brains

To assess the effect of HSD on microglia we performed histological and molecular analyses for two different markers of microglial activation, IBA1 and CD68 [58, 59]. The increase of IBA1 and CD68 levels has been associated with α-syn accumulation and neurodegeneration in PD and MSA animal models [51, 53, 60,61,62,63,64,65]. In agreement with previous data [51], significant microglial activation was observed in PLP-hαSyn mouse brains compared to healthy controls (Fig. 1). Gene expression analysis showed upregulation of Cd68mRNA in the forebrain, midbrain and cerebellum of PLP-hαSyn mice (Fig. 1a). A significant upregulation of Cd68mRNA was also observed in the HSD PLP-hαSyn group compared to PLP-hαSyn mice fed with normal diet (Fig. 1a). However, immunohistological analysis only showed a significant increase of CD68 in PLP-hαSyn mice compared to healthy control animals with no specific effect of diet (Fig. 1b, c). Higher levels of CD68 were observed by immunofluorescence in striatum, substantia nigra (SN), pontine nuclei (PN), and cerebellar white matter (CBWM) of PLP-hαSyn animals compared to wildtypes with no effect of diet either in PLP-hαSyn or healthy control mice, keeping both high-salt groups similar levels to their normal diet groups (Fig. 1b, c). Similar results were obtained with IBA1 (Fig. 1d–f). A significant upregulation of Iba1mRNA was observed in the midbrain and cerebellum of PLP-hαSyn mice compared to wildtype animals (Fig. 1d). Immmunohistological analyses showed a significant increase of IBA1 levels in the SN, PN, and cerebellum of transgenic vs control mice (Fig. 1e, f). No effects of the diet were observed either in PLP-hαSyn or in healthy control animals discarding a specific effect of diet on microglial activation (Fig. 1e, f).

Fig. 1
figure 1

High-salt diet does not affect microglial activation in MSA mice. a Relative gene expression (mRNA levels) of the microglial activation marker Cd68 in different brain regions. The data is expressed in fold change relative to WT mice fed with a normal diet. b Representative immunofluorescence (IF) images of the substantia nigra (SN) sections stained against CD68 (in red). Scale bar, 150 μm. c CD68 positive (CD68+) area in different brain regions (STR: striatum; SN; CBWM: cerebellar white matter; PN: Pontine nuclei) determined by ImageJ analysis and expressed as the % of the total area. d, Relative gene expression of the microglial activation marker Iba1 in different brain regions. The data is expressed in fold change relative to WT mice fed with a normal diet. e Representative IF images of SN sections stained against IBA1 (in green). Scale bar, 150 μm. f IBA1 positive (IBA1+) area in different brain regions. WT, wildtype healthy control animals; TG, PLP-hαSyn mice. White bars: WT mice; gray bars: WT mice fed with HSD (TG + HSD); black bars: TG mice; red bars: TG mice fed with HSD (TG + HSD). Error bars indicate SEM. Two-way ANOVA: *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001 (Bonferroni’s test)

In order to assess the role of astroglia in the inflammatory response observed in PLP-hαSyn mice, we analyzed in the brain subregions the expression levels of Gfap (glial fibrillary acidic protein), a marker of astrogliosis. Significant upregulation of Gfap mRNA was only observed in the cerebellum of PLP-hαSyn with no effect of diet, discarding a general involvement of astroglia in neuroinflammation (Additional file 2: Figure S2).

High-salt diet does not interfere with the neuroinflammatory signaling in the PLP-hαSyn brain

To further characterize the subregion-specific effect of HSD on neuroinflammation in PLP-α-syn mice, we evaluated separately the levels of 36 cytokines and chemokines in forebrain, midbrain, cerebellum, and brainstem by using a multi-analyte detection system (Fig. 2a, b). Heatmap portraying the overall changes of cytokines/chemokines in PLP-hαSyn and control mice showed different profiles between genotypes but no effect of diet (Fig.2a). The analysis revealed a significant increase of CCL3, CCL4, and CCL5 chemokines in PLP-hαSyn mouse brains compared to wildtype animals but no effect of salt (Fig. 2a-b and Additional file 3: Figure S3). The brain concentration of the remaining analytes showed no significant effect of genotype or diet (Additional file 4: Tables S1–S4).

Fig. 2
figure 2

High-salt diet does not affect neuroinflammation in MSA mice. a Heat map comparing the log2 fold change in cytokine and chemokine expression in different brain regions of MSA mice fed with/without high-salt diet and control mice fed with high-salt, as referred to control mice fed with a normal diet. b Protein levels of CCL3, CCL4 and CCL5 chemokines in the midbrain. Protein levels are expressed in pg of protein of interest per mg of total proteins. c Relative gene expression of T-cell markers Cd4 and Cd8 in the midbrain. The data is expressed in fold change relative to WT mice fed with a normal diet. White bars: WT mice; gray bars: WT mice fed with HSD (TG + HSD); black bars: TG mice; red bars: TG mice fed with HSD (TG + HSD). Error bars indicate SEM. Two-way ANOVA: *p < 0.05, **p < 0.01, ****p < 0.0001 (Bonferroni’s test)

We also evaluated changes in the adaptive immune system by analyzing the gene expression levels of two markers of T lymphocytes, Cd4, and Cd8. RT-qPCR analysis showed no changes between animal groups (Fig. 2c and Additional file 3: Figure S3), excluding an effect of the synucleinopathy or diet on T cell recruitment and discarding the involvement of the adaptive immune system in neuroinflammation in PLP-hαSyn mice (Fig. 2c and Additional file 3: Figure S3).

High-salt diet causes no deterioration of motor impairment, neurodegeneration, and myelin dysfunction in PLP-hαSyn mice

In order to evaluate the effect of diet on the gait impairment, we performed Digigait behavioral test. PLP-hαSyn mice showed a significant reduction of stride length compared to healthy control mice with no effect of diet on motor impairment (Fig. 3a). To assess the consequences of HSD on SND, the number of dopaminergic (TH+) neurons in the SNc and the number of MSNs (DARPP-32+) in the striatum were quantified in control and PLP-hαSyn mice. As previously described [51], stereological counting showed significant loss of TH+ and DARPP-32+ neurons in SNc and striatum respectively of PLP-hαSyn mice compared to wildtype animals (Fig. 3b–d). We did not detect any effect of diet neither in control nor in PLP-hαSyn mice (Fig. 3b–d). OPCA was evaluated by the stereological counting of Purkinje cells (DARPP32+) in the cerebellar cortex of PLP-hαSyn and control animals (Fig. 3e). No changes in the number of Purkinje neurons in the cerebellar cortex were observed between animal groups (Fig. 3e), discarding an expansion of the pathology after high-salt exposure similar to the ones observed previously after oxidative stress insult or proteasome inhibition [52, 56]. We also performed a subregion-specific evaluation of the effect of diet on myelin (Fig. 3f and Additional file 3: Figure S3). Gene expression analysis showed significant downregulation of Mbp mRNA (myelin basic protein) in PLP-hαSyn mice compared to healthy controls in most brain subregions (Fig. 3f and Additional file 3: Figure S3). However, we did not observe diet-associated differences neither in control nor in PLP-hαSyn animals, thus excluding a possible role of HSD in motor impairment, neurodegeneration, or myelination dysfunction (Fig. 3 and Additional file 3: Figure S3).

Fig. 3
figure 3

High-salt diet has no further harmful effect on motor impairment, neurodegeneration, and myelin dysfunction. a Gait analysis focused on stride length expressed in centimeter. b Stereological counting of the total number of dopaminergic (TH+) neurons in the entire substantia nigra pars compacta (SNc). c Representative images of SN sections stained against TH. Scale bar, 400 μm. d Stereological counting of the total number of medium spiny (DARPP32+) neurons in the entire STR. e Stereological counting of Purkinje (DARPP32+) neurons in the cerebellar cortex (CB). f Relative gene expression of Mbp in the midbrain. Error bars indicate SEM. Two-way ANOVA: *p < 0.05, **p < 0.01, ***p < 0.001 (Bonferroni’s test)

High-salt diet does not affect α-syn pathology in PLP-hαSyn mice

To fully evaluate the effect of HSD in the PLP-hαSyn mouse model, we assessed α-syn accumulation by histological and molecular analyses. In order to do that, representative brain sections of SNc, CBWM, and motor cortex (M2) were stained with anti-phosphorylated (p-S129) α-syn antibody and the density of GCIs was quantified (Fig. 4a, b). HSD showed no effect on GCI number in any of the regions analyzed (Fig. 4a, b). We also performed subregion-specific dot blot analysis of soluble α-syn (Fig. 4c, d). Significant higher levels of soluble α-syn were found in all brain subregions of PLP-hαSyn mice compared to wildtype animals, but no effect of diet was observed (Fig. 4c, d). Therefore, an effect of diet on α-syn accumulation was discarded.

Fig. 4
figure 4

High-salt diet does not interfere with α-syn accumulation in MSA mice. a Representative images of SN sections stained against phosphorylated α-syn. Red arrows indicate individual GCI. Scale bar, 25 μm. b GCI density in SNc, CBWM, and motor cortex (M2) of PLP-hαSyn mice determined by stereological counting of brain sections stained against phosphorylated α-syn and expressed in GCI/mm2. c Representative images of DOT BLOT analysis for soluble human α-synuclein levels in the midbrain. d Quantification of soluble human α-synuclein levels in different brain areas. The data is shown in arbitrary units (a.u.). Error bars indicate SEM. Two-way ANOVA): ***p < 0.001, ****p < 0.0001 (Bonferroni’s test)

Discussion

Recently, several publications have associated HSD with activation of the immune system and neuroinflammation in experimental models of different neurological disorders, including MS [2,3,4,5,6,7, 11, 14, 16, 17, 66]. Despite the possible deleterious effect of salt on neuroinflammation, a high dietary intake of salt constitutes one of the most recommended non-pharmacological approaches to treat autonomic symptoms in MSA and PD patients [34]. Since microglial activation and neuroinflammation constitute two of the main pathological features of MSA and PD [24,25,26, 35,36,37], the use of HSD could aggravate CNS pathology by enhancing microglial activation, neuroinflammation, and the infiltration of peripheral immune cells.

In order to evaluate the effect of HSD on CNS pathology in α-synucleinopathies, PLP-hαSyn and wildtype animals were both fed with food pellets containing 0.19% (control diet) or 4% NaCl (high-salt diet). HSD consisting 4% NaCl constitutes an increase of about 8–19 times depending on the salt content in normal diet food pellets, which usually range between 0.19 and 0.4% depending on the study. For the human general population, the level of sodium intake recommended in major dietary guidelines ranges from 1200 to 2300 mg per day [67,68,69,70]. However, in MSA and PD, physicians often prescribe increased salt intake to around 10 g of salt per day [71,72,73], being 4–7 times higher than dietary recommendation for the general population. Thus, the experimental approach used in the present study is comparable to the spectrum of human salt consumption and may reflect changes due to HSD similar, or even stronger, than those expected in MSA and PD patients treated with salt. The deleterious effect of HSD in the CNS has also been described in rodents after exposure to 8% NaCl food pellets [11, 74]. However, this may not reflect physiological and clinically relevant conditions since it represents an increase of dietary salt intake of about 16–40 times compared to the normal diet.

Here, we demonstrate that HSD does not affect the activation of microglial cells in PLP-hαSyn mice. Previous results from our group have shown a progressive increase in microglial activation with disease progression in this animal model [51] that can also be augmented in the presence of second deleterious stimuli such as mitochondrial dysfunction or proteasome impairment [52, 56]. In contrast, HSD did not increase microglial activation in the present study according to different molecular and histological analyses. We have also previously shown that the total number of microglial/macrophage cells (IBA1+) does not change in the CNS of MSA mice with disease progression and instead only an increase in their activation state is observed [51]. Therefore, the infiltration of peripheral macrophages in PLP-hαSyn mouse brains appears to be unlikely. Our results differ from experimental studies of inflammatory disorders associated with CNS infiltration of peripheral and monocyte-derived macrophages, where HSD increases activation and polarization towards an M1 phenotype [14, 16, 17] (Fig. 5). The difference between those studies and ours may reflect distinct cell-specific responses of peripheral macrophages and microglial cells to HSD due to their different developmental origin and activation patterns [75,76,77,78]. However, further analyses are required to fully understand these differences.

Fig. 5
figure 5

Pathophysiological features of MSA and EAE models and potential pathogenic effect of a high-salt diet. a Schematic overview of the central nervous system in healthy conditions. b In MSA, α-syn accumulates in the cytoplasm of oligodendrocytes inducing oligodendroglial dysfunction. Soluble α-syn oligomeric species spread through the brain parenchyma and trigger microglial activation and neuroinflammation. All these events lead to demyelination and neurodegeneration. High-salt diet exposure has no effect on the CNS of PLP-hαSyn mice possibly due to the absence of peripheral immune cell involvement in MSA brain pathology. c EAE mice model mimics many features of MS, such as blood-brain barrier (BBB) disruption, demyelinating lesions associated with infiltrating T cells, macrophages, and B cells, microglial activation, neuroinflammation, and neurodegeneration. In EAE mice, high-salt diet increases activation and infiltration of T cells and peripheral macrophages in the CNS accelerating disease onset, augmenting its severity and enhancing blood-brain barrier disruption and brain pathology

We also show that HSD does not interfere with neuroinflammation, astroglial activation, or T cell CNS infiltration in PLP-hαSyn mice. The absence of changes in different markers of T lymphocytes (CD4 and CD8) and their associated proinflammatory cytokines and chemokines (i.e., IFN-γ, IL-17, IL-12, IL-23) [13, 79,80,81,82,83] in the brain of MSA mice compared to wildtype animals suggest that these cells are not critical in the neuroinflammatory process observed in PLP-hαSyn mice. Moreover, human studies have shown no clear evidence of T cell involvement in MSA so far. The absence of a higher activation of the immune system in PLP-hαSyn mice could explain the differences with HSD studies in EAE models of MS where activation and infiltration of T cells and peripheral macrophages in the CNS accelerate disease onset, augment its severity, and enhance brain pathology [6,7,8,9,10, 12, 14, 15] (Fig. 5). However, the results obtained from our study cannot be extrapolated to PD, where an association between T cells and neurodegeneration has been recently suggested [38,39,40,41,42,43]. Further studies including pre-clinical models of PD are needed to clarify the effect of HSD on underlying neuropathology.

HSD did not affect neurodegeneration or demyelination in MSA mice. Although PLP-hαSyn animals develop SND and myelin dysfunction characterized respectively by a significant loss of dopaminergic neurons in the SNc and MSNs in the striatum and a downregulation of the Mbp gene in several brain subregions, no changes were observed after HSD exposure in these mice. These findings again differ from the studies in EAE models of MS where HSD has led to enhanced demyelination [14, 15]. In agreement with our data, a large clinical study has recently revealed that salt intake does not influence MS disease course or activity thus indicating that EAE models, which are induced by active immunization with myelin components or by passive transfer of autoreactive T cells, do not necessarily reflect the human disease [18, 84]. In contrast to the MSA model used in our study, where neuroinflammation is a secondary event induced by the aggregation of α-syn in oligodendrocytes [51, 53], neuroinflammation is a primary event induced by a peripheral immune response in the EAE models used for the studies on salt intake [9, 12, 13, 84]. Thus, the differences in the effects of diet on the neuropathology of MSA and MS animal models could also be explained by the absence of neuroinflammatory changes induced by HSD in PLP-hαSyn mice (Fig. 5).

Previous results from our group have shown that a combination of synucleinopathy with second hit stimuli such as oxidative stress or proteasome inhibition can aggravate the pathology in PLP-hαSyn mice towards full-blown MSA with strong microglial activation and spreading of SND, OPCA, and GCIs [52, 56]. However, PLP-hαSyn mice showed no changes in α-syn accumulation after HSD exposure by neither molecular nor histological analyses, therefore excluding an effect of diet on any of the synucleinopathy features.

In contrast to our observations, two recent studies by Faraco et al. have shown that HSD induces cognitive impairment in wildtype animals [11, 74]. Faraco et al. observed significant deleterious effects in wildtype mice after 12 weeks of HSD with 4% NaCl food pellets, a similar protocol to the one used in our study. However, we did not find differences between wildtype animals fed with normal or HSD. These differences may be explained by the use of different mouse substrains. The PLP-hαSyn and control animals used in our study were C57BL/6 N. Unfortunately, Faraco et al. do not mention which specific C57BL/6 substrain was used in their study. It has been shown that C57BL/6 substrains present behavioral [85] and genetic differences—including immune function —[86]. Moreover, other groups have shown that HSD has either no effect or even ameliorates symptoms in animal models of other inflammatory disorders [87, 88]. Thus, the differences between our data and previous HSD studies, and in particular data in EAE mice, may be explained by the use of different mouse strains or by the different experimental approaches used to boost the inflammatory process, as previously discussed.

Despite the publications supporting the deleterious effect of HSD in experimental models of MS, two large clinical studies failed to show an association between salt intake and higher MS disease risk, progression, or activity [18, 19]. Moreover, although sodium intake differs between East Asian, American, and European population [89], MSA cohort study groups from Japan, Europe, and the USA show a similar median survival [90,91,92], supporting the current findings and suggesting that salt consumption differences may not affect disease progression in MSA.

Several limitations of the current study need to be acknowledged. Despite not having observed an effect of diet in the CNS of MSA mice, we cannot exclude an effect in peripheral organs. Moreover, in the present study, we have not evaluated the effect of HSD on cardiovascular regulation or pathology. Among the different autonomic cardiovascular problems present in MSA patients, the presence of orthostatic hypotension (OH) constitutes one of the major criteria for diagnosis [93]. Data from our group show cardiovascular defects in PLP-hαSyn mice [50], but it is not possible to address the issue of OH in a mouse model. In this regard, we cannot exclude a role of high sodium intake on neuropathology linked to OH in MSA patients. However, the analysis of the effect of diet in both the peripheral immune system and the different cardiovascular features of PLP-hαSyn mice is beyond the scope of the present study, where the main objective was to study the effect of HSD in the neuroinflammatory process underlying MSA pathology in the CNS.

Conclusions

Here, we demonstrate that HSD does not interfere with microglial activation, neuroinflammation, motor function, neurodegeneration, and α-syn accumulation in the PLP-hαSyn mouse model of MSA, making deleterious effects of HSD on brain pathology and its progression unlikely. Our findings contrast with experimental data obtained in EAE models of MS and do not support a pro-neuroinflammatory effect of the current clinical practice of a high-salt diet for the treatment of autonomic failure in MSA.

Availability of data and materials

The data that support the findings of this study are available from the corresponding author upon reasonable request.

Abbreviations

CBWM:

Cerebellar white matter

CNS:

Central nervous system

EAE:

Experimental autoimmune encephalomyelitis

GCIs:

Glial cytoplasmic inclusions

HSD:

High-salt diet

MS:

Multiple sclerosis

MSA:

Multiple system atrophy

MSNs:

Medium spiny neurons

nOH:

Neurogenic orthostatic hypotension

OPCA:

Olivopontocerebellar atrophy

PD:

Parkinson’s disease

PN:

Pontine nuclei

SN:

Substantia nigra

SNc:

Substantia nigra pars compacta

SND:

Striatonigral degeneration

TH:

Tyrosine hydroxylase

α-syn:

Alpha-synuclein

References

  1. Sundstrom B, Johansson I, Rantapaa-Dahlqvist S. Interaction between dietary sodium and smoking increases the risk for rheumatoid arthritis: results from a nested case-control study. Rheumatology (Oxford). 2015;54(3):487–93.

    Article  CAS  Google Scholar 

  2. Gilman TL, Mitchell NC, Daws LC, Toney GM. Neuroinflammation contributes to high salt intake-augmented neuronal activation and active coping responses to acute stress. Int J Neuropsychopharmacol. 2019;22(2):137–42.

    Article  CAS  PubMed  Google Scholar 

  3. Janakiraman M, Krishnamoorthy G. Emerging role of diet and microbiota interactions in Neuroinflammation. Front Immunol. 2018;9:2067.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Farez MF, Fiol MP, Gaitan MI, Quintana FJ, Correale J. Sodium intake is associated with increased disease activity in multiple sclerosis. J Neurol Neurosurg Psychiatry. 2015;86(1):26–31.

    Article  PubMed  Google Scholar 

  5. Hucke S, Wiendl H, Klotz L. Implications of dietary salt intake for multiple sclerosis pathogenesis. Mult Scler. 2016;22(2):133–9.

    Article  CAS  PubMed  Google Scholar 

  6. Kleinewietfeld M, Manzel A, Titze J, Kvakan H, Yosef N, Linker RA, et al. Sodium chloride drives autoimmune disease by the induction of pathogenic TH17 cells. Nature. 2013;496(7446):518–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Wu C, Yosef N, Thalhamer T, Zhu C, Xiao S, Kishi Y, et al. Induction of pathogenic TH17 cells by inducible salt-sensing kinase SGK1. Nature. 2013;496(7446):513–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Codarri L, Gyulveszi G, Tosevski V, Hesske L, Fontana A, Magnenat L, et al. RORgammat drives production of the cytokine GM-CSF in helper T cells, which is essential for the effector phase of autoimmune neuroinflammation. Nat Immunol. 2011;12(6):560–7.

    Article  CAS  PubMed  Google Scholar 

  9. Kebir H, Kreymborg K, Ifergan I, Dodelet-Devillers A, Cayrol R, Bernard M, et al. Human TH17 lymphocytes promote blood-brain barrier disruption and central nervous system inflammation. Nat Med. 2007;13(10):1173–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Reboldi A, Coisne C, Baumjohann D, Benvenuto F, Bottinelli D, Lira S, et al. C-C chemokine receptor 6-regulated entry of TH-17 cells into the CNS through the choroid plexus is required for the initiation of EAE. Nat Immunol. 2009;10(5):514–23.

    Article  CAS  PubMed  Google Scholar 

  11. Faraco G, Brea D, Garcia-Bonilla L, Wang G, Racchumi G, Chang H, et al. Dietary salt promotes neurovascular and cognitive dysfunction through a gut-initiated TH17 response. Nat Neurosci. 2018;21(2):240–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Gold R, Linington C, Lassmann H. Understanding pathogenesis and therapy of multiple sclerosis via animal models: 70 years of merits and culprits in experimental autoimmune encephalomyelitis research. Brain J Neurol. 2006;129(Pt 8):1953–71.

    Article  Google Scholar 

  13. Segal BM. Experimental autoimmune encephalomyelitis: cytokines, effector T cells, and antigen-presenting cells in a prototypical Th1-mediated autoimmune disease. Curr Allergy Asthma Rep. 2003;3(1):86–93.

    Article  PubMed  Google Scholar 

  14. Hucke S, Eschborn M, Liebmann M, Herold M, Freise N, Engbers A, et al. Sodium chloride promotes pro-inflammatory macrophage polarization thereby aggravating CNS autoimmunity. J Autoimmun. 2016;67:90–101.

    Article  CAS  PubMed  Google Scholar 

  15. Krementsov DN, Case LK, Hickey WF, Teuscher C. Exacerbation of autoimmune neuroinflammation by dietary sodium is genetically controlled and sex specific. FASEB J. 2015;29(8):3446–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Zhang WC, Zheng XJ, Du LJ, Sun JY, Shen ZX, Shi C, et al. High salt primes a specific activation state of macrophages, M (Na). Cell Res. 2015;25(8):893–910.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Binger KJ, Gebhardt M, Heinig M, Rintisch C, Schroeder A, Neuhofer W, et al. High salt reduces the activation of IL-4- and IL-13-stimulated macrophages. J Clin Invest. 2015;125(11):4223–38.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Fitzgerald KC, Munger KL, Hartung HP, Freedman MS, Montalban X, Edan G, et al. Sodium intake and multiple sclerosis activity and progression in BENEFIT. Ann Neurol. 2017;82(1):20–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Cortese M, Yuan C, Chitnis T, Ascherio A, Munger KL. No association between dietary sodium intake and the risk of multiple sclerosis. Neurology. 2017;89(13):1322–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Fanciulli A, Wenning GK. Multiple-system atrophy. N Engl J Med. 2015;372(3):249–63.

    Article  PubMed  CAS  Google Scholar 

  21. Spillantini MG, Schmidt ML, Lee VM, Trojanowski JQ, Jakes R, Goedert M. Alpha-synuclein in Lewy bodies. Nature. 1997;388(6645):839–40.

    Article  CAS  PubMed  Google Scholar 

  22. Trojanowski JQ, Lee VM. Parkinson's disease and related alpha-synucleinopathies are brain amyloidoses. Ann N Y Acad Sci. 2003;991:107–10.

    Article  CAS  PubMed  Google Scholar 

  23. Tu PH, Galvin JE, Baba M, Giasson B, Tomita T, Leight S, et al. Glial cytoplasmic inclusions in white matter oligodendrocytes of multiple system atrophy brains contain insoluble alpha-synuclein. Ann Neurol. 1998;44(3):415–22.

    Article  CAS  PubMed  Google Scholar 

  24. Gerhard A, Banati RB, Goerres GB, Cagnin A, Myers R, Gunn RN, et al. [11C](R)-PK11195 PET imaging of microglial activation in multiple system atrophy. Neurology. 2003;61(5):686–9.

    Article  CAS  PubMed  Google Scholar 

  25. Ishizawa K, Komori T, Sasaki S, Arai N, Mizutani T, Hirose T. Microglial activation parallels system degeneration in multiple system atrophy. J Neuropathol Exp Neurol. 2004;63(1):43–52.

    Article  PubMed  Google Scholar 

  26. Vieira BD, Radford RA, Chung RS, Guillemin GJ, Pountney DL. Neuroinflammation in multiple system atrophy: response to and cause of alpha-Synuclein aggregation. Front Cell Neurosci. 2015;9:437.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Don AS, Hsiao JH, Bleasel JM, Couttas TA, Halliday GM, Kim WS. Altered lipid levels provide evidence for myelin dysfunction in multiple system atrophy. Acta Neuropathol Commun. 2014;2:150.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Ishizawa K, Komori T, Arai N, Mizutani T, Hirose T. Glial cytoplasmic inclusions and tissue injury in multiple system atrophy: a quantitative study in white matter (olivopontocerebellar system) and gray matter (nigrostriatal system). Neuropathology. 2008;28(3):249–57.

    Article  PubMed  Google Scholar 

  29. Stefanova N, Wenning GK. Review: multiple system atrophy: emerging targets for interventional therapies. Neuropathol Appl Neurobiol. 2016;42(1):20–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Ahmed Z, Asi YT, Sailer A, Lees AJ, Houlden H, Revesz T, et al. The neuropathology, pathophysiology and genetics of multiple system atrophy. Neuropathol Appl Neurobiol. 2012;38(1):4–24.

    Article  CAS  PubMed  Google Scholar 

  31. Ozawa T, Paviour D, Quinn NP, Josephs KA, Sangha H, Kilford L, et al. The spectrum of pathological involvement of the striatonigral and olivopontocerebellar systems in multiple system atrophy: clinicopathological correlations. Brain. 2004;127(Pt 12):2657–71.

    Article  PubMed  Google Scholar 

  32. Yoshida M. Multiple system atrophy: alpha-synuclein and neuronal degeneration. Neuropathology. 2007;27(5):484–93.

    Article  PubMed  Google Scholar 

  33. Eschlbock S, Krismer F, Wenning GK. Interventional trials in atypical parkinsonism. Parkinsonism Relat Disord. 2016;22(Suppl 1):S82–92.

    Article  PubMed  Google Scholar 

  34. Metzler M, Duerr S, Granata R, Krismer F, Robertson D, Wenning GK. Neurogenic orthostatic hypotension: pathophysiology, evaluation, and management. J Neurol. 2013;260(9):2212–9.

    Article  PubMed  Google Scholar 

  35. Bartels AL, Willemsen AT, Doorduin J, de Vries EF, Dierckx RA, Leenders KL. [11C]-PK11195 PET: quantification of neuroinflammation and a monitor of anti-inflammatory treatment in Parkinson's disease? Parkinsonism Relat Disord. 2010;16(1):57–9.

    Article  CAS  PubMed  Google Scholar 

  36. Gerhard A, Pavese N, Hotton G, Turkheimer F, Es M, Hammers A, et al. In vivo imaging of microglial activation with [11C](R)-PK11195 PET in idiopathic Parkinson's disease. Neurobiol Dis. 2006;21(2):404–12.

    Article  CAS  PubMed  Google Scholar 

  37. 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(8):1285–91.

    Article  CAS  PubMed  Google Scholar 

  38. Brochard V, Combadiere B, Prigent A, Laouar Y, Perrin A, Beray-Berthat V, et al. Infiltration of CD4+ lymphocytes into the brain contributes to neurodegeneration in a mouse model of Parkinson disease. J Clin Invest. 2009;119(1):182–92.

    CAS  PubMed  Google Scholar 

  39. Stevens CH, Rowe D, Morel-Kopp MC, Orr C, Russell T, Ranola M, et al. Reduced T helper and B lymphocytes in Parkinson's disease. J Neuroimmunol. 2012;252(1–2):95–9.

    Article  CAS  PubMed  Google Scholar 

  40. Bas J, Calopa M, Mestre M, Mollevi DG, Cutillas B, Ambrosio S, et al. Lymphocyte populations in Parkinson's disease and in rat models of parkinsonism. J Neuroimmunol. 2001;113(1):146–52.

    Article  CAS  PubMed  Google Scholar 

  41. Baba Y, Kuroiwa A, Uitti RJ, Wszolek ZK, Yamada T. Alterations of T-lymphocyte populations in Parkinson disease. Parkinsonism Relat Disord. 2005;11(8):493–8.

    Article  PubMed  Google Scholar 

  42. Sulzer D, Alcalay RN, Garretti F, Cote L, Kanter E, Agin-Liebes J, et al. T cells from patients with Parkinson's disease recognize alpha-synuclein peptides. Nature. 2017;546(7660):656–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Sommer A, Maxreiter F, Krach F, Fadler T, Grosch J, Maroni M, et al. Th17 lymphocytes induce neuronal cell death in a human iPSC-based model of Parkinson's disease. Cell Stem Cell. 2018;23(1):123–31 e6.

    Article  CAS  PubMed  Google Scholar 

  44. Kahle PJ, Neumann M, Ozmen L, Muller V, Jacobsen H, Spooren W, et al. Hyperphosphorylation and insolubility of alpha-synuclein in transgenic mouse oligodendrocytes. EMBO Rep. 2002;3(6):583–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Boudes M, Uvin P, Pinto S, Voets T, Fowler CJ, Wenning GK, et al. Bladder dysfunction in a transgenic mouse model of multiple system atrophy. Mov Disord. 2013;28(3):347–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Fernagut PO, Meissner WG, Biran M, Fantin M, Bassil F, Franconi JM, et al. Age-related motor dysfunction and neuropathology in a transgenic mouse model of multiple system atrophy. Synapse. 2014;68(3):98–106.

    Article  CAS  PubMed  Google Scholar 

  47. Flabeau O, Meissner WG, Ozier A, Berger P, Tison F, Fernagut PO. Breathing variability and brainstem serotonergic loss in a genetic model of multiple system atrophy. Mov Disord. 2014;29(3):388–95.

    Article  CAS  PubMed  Google Scholar 

  48. Hartner L, Keil TW, Kreuzer M, Fritz EM, Wenning GK, Stefanova N, et al. Distinct parameters in the EEG of the PLP alpha-SYN mouse model for multiple system atrophy reinforce face validity. Front Behav Neurosci. 2016;10:252.

    PubMed  Google Scholar 

  49. Krismer F, Wenning GK, Li Y, Poewe W, Stefanova N. Intact olfaction in a mouse model of multiple system atrophy. PLoS One. 2013;8(5):e64625.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Kuzdas D, Stemberger S, Gaburro S, Stefanova N, Singewald N, Wenning GK. Oligodendroglial alpha-synucleinopathy and MSA-like cardiovascular autonomic failure: experimental evidence. Exp Neurol. 2013;247:531–6.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Refolo V, Bez F, Polissidis A, Kuzdas-Wood D, Sturm E, Kamaratou M, et al. Progressive striatonigral degeneration in a transgenic mouse model of multiple system atrophy: translational implications for interventional therapies. Acta Neuropathol Commun. 2018;6(1):2.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Stefanova N, Reindl M, Neumann M, Haass C, Poewe W, Kahle PJ, et al. Oxidative stress in transgenic mice with oligodendroglial alpha-synuclein overexpression replicates the characteristic neuropathology of multiple system atrophy. Am J Pathol. 2005;166(3):869–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Stefanova N, Reindl M, Neumann M, Kahle PJ, Poewe W, Wenning GK. Microglial activation mediates neurodegeneration related to oligodendroglial alpha-synucleinopathy: implications for multiple system atrophy. Mov Disord. 2007;22(15):2196–203.

    Article  PubMed  Google Scholar 

  54. Stemberger S, Poewe W, Wenning GK, Stefanova N. Targeted overexpression of human alpha-synuclein in oligodendroglia induces lesions linked to MSA-like progressive autonomic failure. Exp Neurol. 2010;224(2):459–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Bassil F, Fernagut PO, Bezard E, Pruvost A, Leste-Lasserre T, Hoang QQ, et al. Reducing C-terminal truncation mitigates synucleinopathy and neurodegeneration in a transgenic model of multiple system atrophy. Proc Natl Acad Sci U S A. 2016;113(34):9593–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Stefanova N, Kaufmann WA, Humpel C, Poewe W, Wenning GK. Systemic proteasome inhibition triggers neurodegeneration in a transgenic mouse model expressing human alpha-synuclein under oligodendrocyte promoter: implications for multiple system atrophy. Acta Neuropathol. 2012;124(1):51–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) method. Methods. 2001;25(4):402–8.

    Article  CAS  PubMed  Google Scholar 

  58. Ito D, Imai Y, Ohsawa K, Nakajima K, Fukuuchi Y, Kohsaka S. Microglia-specific localisation of a novel calcium binding protein, Iba1. Brain Res Mol Brain Res. 1998;57(1):1–9.

    Article  CAS  PubMed  Google Scholar 

  59. Zotova E, Bharambe V, Cheaveau M, Morgan W, Holmes C, Harris S, et al. Inflammatory components in human Alzheimer's disease and after active amyloid-beta42 immunization. Brain. 2013;136(Pt 9):2677–96.

    Article  PubMed  Google Scholar 

  60. Austin SA, Floden AM, Murphy EJ, Combs CK. Alpha-synuclein expression modulates microglial activation phenotype. J Neurosci. 2006;26(41):10558–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Doorn KJ, Moors T, Drukarch B, van de Berg W, Lucassen PJ, van Dam AM. Microglial phenotypes and toll-like receptor 2 in the substantia nigra and hippocampus of incidental Lewy body disease cases and Parkinson's disease patients. Acta Neuropathol Commun. 2014;2:90.

    PubMed  PubMed Central  Google Scholar 

  62. Fellner L, Irschick R, Schanda K, Reindl M, Klimaschewski L, Poewe W, et al. Toll-like receptor 4 is required for alpha-synuclein dependent activation of microglia and astroglia. Glia. 2013;61(3):349–60.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Sanchez-Guajardo V, Barnum CJ, Tansey MG, Romero-Ramos M. Neuroimmunological processes in Parkinson's disease and their relation to alpha-synuclein: microglia as the referee between neuronal processes and peripheral immunity. ASN Neuro. 2013;5(2):113–39.

    Article  CAS  PubMed  Google Scholar 

  64. Theodore S, Cao S, McLean PJ, Standaert DG. Targeted overexpression of human alpha-synuclein triggers microglial activation and an adaptive immune response in a mouse model of Parkinson disease. J Neuropathol Exp Neurol. 2008;67(12):1149–58.

    Article  CAS  PubMed  Google Scholar 

  65. Zhang W, Wang T, Pei Z, Miller DS, Wu X, Block ML, et al. Aggregated alpha-synuclein activates microglia: a process leading to disease progression in Parkinson's disease. FASEB J. 2005;19(6):533–42.

    Article  CAS  PubMed  Google Scholar 

  66. Zhang T, Fang S, Wan C, Kong Q, Wang G, Wang S, et al. Excess salt exacerbates blood-brain barrier disruption via a p38/MAPK/SGK1-dependent pathway in permanent cerebral ischemia. Sci Rep. 2015;5:16548.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Guideline: Sodium Intake for Adults and Children. WHO Guidelines Approved by the Guidelines Review Committee. Geneva; 2012.

  68. Whelton PK, Appel LJ, Sacco RL, Anderson CA, Antman EM, Campbell N, et al. Sodium, blood pressure, and cardiovascular disease: further evidence supporting the American Heart Association sodium reduction recommendations. Circulation. 2012;126(24):2880–9.

    Article  CAS  PubMed  Google Scholar 

  69. Application of lower sodium intake recommendations to adults--United States, 1999-2006. MMWR Morb Mortal Wkly Rep. 2009;58(11):281–3.

  70. Dietary Guidelines for Americans, 2010. Agriculture USDo, Services USDoHaH. Washington; 2011.

  71. Figueroa JJ, Basford JR, Low PA. Preventing and treating orthostatic hypotension: as easy as a, B, C. Cleve Clin J Med. 2010;77(5):298–306.

    Article  PubMed  PubMed Central  Google Scholar 

  72. El-Sayed H, Hainsworth R. Salt supplement increases plasma volume and orthostatic tolerance in patients with unexplained syncope. Heart. 1996;75(2):134–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Gilhus NE, Barnes MP, Brainin M, Lahrmann H, Cortelli P, Hilz M, Mathias CJ, Struhal W, Tassinari M. Orthostatic Hypotension. Eur Handb Neurol Manag. 2010.

  74. Faraco G, Hochrainer K, Segarra SG, Schaeffer S, Santisteban MM, Menon A, et al. Dietary salt promotes cognitive impairment through tau phosphorylation. Nature. 2019;574(7780):686–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Ginhoux F, Greter M, Leboeuf M, Nandi S, See P, Gokhan S, et al. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science. 2010;330(6005):841–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. London A, Cohen M, Schwartz M. Microglia and monocyte-derived macrophages: functionally distinct populations that act in concert in CNS plasticity and repair. Front Cell Neurosci. 2013;7:34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Li Q, Barres BA. Microglia and macrophages in brain homeostasis and disease. Nat Rev Immunol. 2018;18(4):225–42.

    Article  CAS  PubMed  Google Scholar 

  78. Yamasaki R, Lu H, Butovsky O, Ohno N, Rietsch AM, Cialic R, et al. Differential roles of microglia and monocytes in the inflamed central nervous system. J Exp Med. 2014;211(8):1533–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Kroenke MA, Segal BM. IL-23 modulated myelin-specific T cells induce EAE via an IFNgamma driven, IL-17 independent pathway. Brain Behav Immun. 2011;25(5):932–7.

    Article  CAS  PubMed  Google Scholar 

  80. Liu J, Lin F, Strainic MG, An F, Miller RH, Altuntas CZ, et al. IFN-gamma and IL-17 production in experimental autoimmune encephalomyelitis depends on local APC-T cell complement production. J Immunol. 2008;180(9):5882–9.

    Article  CAS  PubMed  Google Scholar 

  81. Cua DJ, Sherlock J, Chen Y, Murphy CA, Joyce B, Seymour B, et al. Interleukin-23 rather than interleukin-12 is the critical cytokine for autoimmune inflammation of the brain. Nature. 2003;421(6924):744–8.

    Article  CAS  PubMed  Google Scholar 

  82. McGeachy MJ, Chen Y, Tato CM, Laurence A, Joyce-Shaikh B, Blumenschein WM, et al. The interleukin 23 receptor is essential for the terminal differentiation of interleukin 17-producing effector T helper cells in vivo. Nat Immunol. 2009;10(3):314–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Voskuhl RR, Martin R, Bergman C, Dalal M, Ruddle NH, McFarland HF. T helper 1 (Th1) functional phenotype of human myelin basic protein-specific T lymphocytes. Autoimmunity. 1993;15(2):137–43.

    Article  CAS  PubMed  Google Scholar 

  84. Lassmann H, Bradl M. Multiple sclerosis: experimental models and reality. Acta Neuropathol. 2017;133(2):223–44.

    Article  CAS  PubMed  Google Scholar 

  85. Bryant CD, Zhang NN, Sokoloff G, Fanselow MS, Ennes HS, Palmer AA, et al. Behavioral differences among C57BL/6 substrains: implications for transgenic and knockout studies. J Neurogenet. 2008;22(4):315–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Simon MM, Greenaway S, White JK, Fuchs H, Gailus-Durner V, Wells S, et al. A comparative phenotypic and genomic analysis of C57BL/6J and C57BL/6N mouse strains. Genome Biol. 2013;14(7):R82.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  87. Huehnchen P, Boehmerle W, Endres M. High salt diet ameliorates functional, electrophysiological and histological characteristics of murine spontaneous autoimmune polyneuropathy. Neurobiol Dis. 2019;124:240–7.

    Article  CAS  PubMed  Google Scholar 

  88. Kolypetri P, Randell E, Van Vliet BN, Carayanniotis G. High salt intake does not exacerbate murine autoimmune thyroiditis. Clin Exp Immunol. 2014;176(3):336–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Powles J, Fahimi S, Micha R, Khatibzadeh S, Shi P, Ezzati M, et al. Global, regional and national sodium intakes in 1990 and 2010: a systematic analysis of 24 h urinary sodium excretion and dietary surveys worldwide. BMJ Open. 2013;3(12):e003733.

    Article  PubMed  PubMed Central  Google Scholar 

  90. Watanabe H, Saito Y, Terao S, Ando T, Kachi T, Mukai E, et al. Progression and prognosis in multiple system atrophy: an analysis of 230 Japanese patients. Brain. 2002;125(Pt 5):1070–83.

    Article  PubMed  Google Scholar 

  91. Wenning GK, Geser F, Krismer F, Seppi K, Duerr S, Boesch S, et al. The natural history of multiple system atrophy: a prospective European cohort study. Lancet Neurol. 2013;12(3):264–74.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Low PA, Reich SG, Jankovic J, Shults CW, Stern MB, Novak P, et al. Natural history of multiple system atrophy in the USA: a prospective cohort study. Lancet Neurol. 2015;14(7):710–9.

    Article  PubMed  PubMed Central  Google Scholar 

  93. Gilman S, Wenning GK, Low PA, Brooks DJ, Mathias CJ, Trojanowski JQ, et al. Second consensus statement on the diagnosis of multiple system atrophy. Neurology. 2008;71(9):670–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

The study was supported by grants of the Austrian Science Fund (FWF) F4414, W1206-08 and a grant of the European Community’s Seventh Framework Programme (FP7/2007–2013) under agreement n 602646 (Multisyn).

Author information

Authors and Affiliations

Authors

Contributions

AHG contributed to the acquisition, analysis, and interpretation of data, helped to draft and revise the manuscript, and gave final approval of the submission. VR contributed to the acquisition, analysis, and interpretation of data, revised the manuscript, and gave final approval of the submission. MR contributed to the interpretation of data, revised the manuscript, and gave final approval of the submission. GW and NS contributed to the conception and design, helped in the interpretation of data, revised the manuscript, and gave final approval of the submission. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Nadia Stefanova.

Ethics declarations

Ethics approval and consent to participate

All the experiments were performed according to the ethical guidelines of the EU (Directive 2010/63/EU for animal experiments) and the Austrian Federal Ministry of Science and Research (permission BMFWF-66.011/0018-WF/v/3b/2015).

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

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

Supplementary information

Additional file 1: Figure S1

. Animal body weight. Body weight of healthy control mice fed with normal diet (WT; white circles) or HSD (WT + HSD; gray circles) and PLP-hαSyn mice fed with normal diet (TG; black squares) or HSD (TG + HSD; red squares). Body weight was measured weekly throughout the treatment period.

Additional file 2: Figure S2

. High-salt diet does not affect astrogliosis in MSA mice. Gene expression (mRNA) levels in forebrain, midbrain, cerebellum and brainstem of the astrocyte marker Gfap. mRNA levels are expressed in fold change relative to WT mice fed with normal diet. White bars: WT mice; Gray bars: WT mice fed with HSD (TG + HSD); Black bars: TG mice; Red bars: TG mice fed with HSD (TG + HSD). Error bars indicate SEM. Two-way ANOVA: *** p < 0.001, **** p < 0.0001 (Bonferroni’s test).

Additional file 3: Figure S3

. Protein levels and gene expression (mRNA) levels in forebrain, cerebellum and brainstem of the main proteins and genes analyzed on the study. Protein levels are expressed in pg of protein of interest per mg of total proteins. mRNA levels are expressed in fold change relative to WT mice fed with normal diet. White bars: WT mice; Gray bars: WT mice fed with HSD (TG + HSD); Black bars: TG mice; Red bars: TG mice fed with HSD (TG + HSD). Error bars indicate SEM. Two-way ANOVA: * p < 0.05, ** p < 0.01, *** p < 0.001 (Bonferroni’s test).

Additional file 4: Tables S1

. Cytokine and chemokine protein levels in forebrain. The table represent the concentrations (pg/mg total protein) of the different cytokines and chemokines measured in forebrain of control and MSA mice fed with normal or high salt diet. Data are presented as mean ± SD. p-values where obtained by two-way ANOVA with Bonferroni’s post hoc test. Tables S2. Cytokine and chemokine protein levels in midbrain. The table represent the concentrations (pg/mg total protein) of the different cytokines and chemokines measured in forebrain of control and MSA mice fed with normal or high salt diet. Data are presented as mean ± SD. p-values where obtained by two-way ANOVA with Bonferroni’s post hoc test. Tables S3. Cytokine and chemokine protein levels in cerebellum. The table represent the concentrations (pg/mg total protein) of the different cytokines and chemokines measured in forebrain of control and MSA mice fed with normal or high salt diet. Data are presented as mean ± SD. p-values where obtained by two-way ANOVA with Bonferroni’s post hoc test. Tables S4. Cytokine and chemokine protein levels in brainstem. The table represent the concentrations (pg/mg total protein) of the different cytokines and chemokines measured in forebrain of control and MSA mice fed with normal or high salt diet. Data are presented as mean ± SD. p-values where obtained by two-way ANOVA with Bonferroni’s post hoc test.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. 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.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Heras-Garvin, A., Refolo, V., Reindl, M. et al. High-salt diet does not boost neuroinflammation and neurodegeneration in a model of α-synucleinopathy. J Neuroinflammation 17, 35 (2020). https://doi.org/10.1186/s12974-020-1714-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12974-020-1714-y

Keywords