Skip to main content
Erschienen in: Journal of Neuroinflammation 1/2017

Open Access 01.12.2017 | Short Report

Impact of combined sodium chloride and saturated long-chain fatty acid challenge on the differentiation of T helper cells in neuroinflammation

verfasst von: Anna Hammer, Anne Schliep, Stefanie Jörg, Aiden Haghikia, Ralf Gold, Markus Kleinewietfeld, Dominik N. Müller, Ralf A. Linker

Erschienen in: Journal of Neuroinflammation | Ausgabe 1/2017

Abstract

Background

There has been a marked increase in the incidence of autoimmune diseases like multiple sclerosis (MS) in the last decades which is most likely driven by a change in environmental factors. Here, growing evidence suggests that ingredients of a Western diet like high intake of sodium chloride (NaCl) or saturated fatty acids may impact systemic immune responses, thus increasing disease susceptibility. Recently, we have shown that high dietary salt or long-chain fatty acid (LCFA) intake indeed aggravates T helper (Th) cell responses and neuroinflammation.

Methods

Naïve CD4+ T cells were treated with an excess of 40 mM NaCl and/or 250 μM lauric acid (LA) in vitro to analyze effects on Th cell differentiation, cytokine secretion, and gene expression. We employed ex vivo analyses of the model disease murine experimental autoimmune encephalomyelitis (EAE) to investigate whether salt and LCFA may affect disease severity and T cell activation in vivo.

Results

LCFA, like LA, together with NaCl enhance the differentiation of Th1 and Th17 cells as well as pro-inflammatory cytokine and gene expression in vitro. In cell culture, we observed an additive effect of LA and hypertonic extracellular NaCl (NaCl + LA) in Th17 differentiation assays as well as on IL-17, GM-CSF, and IL-2 gene expression. In contrast, NaCl + LA reduced Th2 frequencies. We employed EAE as a model of Th1/Th17 cell-mediated autoimmunity and show that the combination of a NaCl- and LA-rich diet aggravated the disease course and increased T cell infiltration into the central nervous system (CNS) to the same extent as dietary NaCl.

Conclusions

Our findings demonstrate a partially additive effect of NaCl and LA on Th cell polarization in vitro and on Th cell responses in autoimmune neuroinflammation. These data may help to better understand the pathophysiology of autoimmune diseases such as MS.
Abkürzungen
ahr
Aryl hydrocarbon receptor
BBB
Blood-brain barrier
CFA
Complete Freund’s Adjuvant
CNS
Central nervous system
EAE
Experimental autoimmune encephalomyelitis
GM-CSF
Granulocyte macrophage colony-stimulating factor
IFNγ
Interferon gamma
IL
Interleukin
LA
Lauric acid
LCFA
Long-chain fatty acids
MOG
Myelin oligodendrocyte glycoprotein
MRI
Magnetic resonance imaging
MS
Multiple sclerosis
NaCl
Sodium chloride
RORγ
RAR-related orphan receptor gamma
SGK1
Serum- and glucocorticoid-regulated kinase-1
T-bet
T-box-containing protein expressed in T cells
Th
T helper
Tnf
Tumor necrosis factor

Background

The increasing incidence of autoimmune diseases with a high prevalence in Western countries [1] and the rapid evolution of multiple sclerosis (MS) in former low prevalence countries like Japan [2] indicate a role of environmental factors that do play a role beyond predisposing genetic risk factors. Here, most impressively, dietary habits like the so-called Western diet, high in both fat and salt, may play a role (as reviewed in [3, 4]).
High dietary salt (sodium chloride (NaCl)) intake is a major culprit in cardiovascular disease [5], cancer [6], chronic inflammation [7], and also autoimmune diseases [8, 9]. Mimicking in vivo situations in the tissue after a high-salt diet by raising sodium concentrations in vitro has been shown to affect adaptive immune cells and promote the differentiation of murine and human T helper (Th)17 cells with a pathogenic phenotype [10, 11]. Here, the serum- and glucocorticoid-regulated kinase-1 (SGK1) is one of the key mediators of salt-augmented differentiation of Th17 cells. Further studies on the effects of NaCl-induced hypertonicity on the activation and function of myeloid dendritic cells (DC) provide evidence that, in autoimmune neuroinflammation, high-salt effects on T cells are rather directly exerted than primarily mediated via DC [12]. In experimental autoimmune encephalomyelitis (EAE), a Th1/Th17 model disease mimicking many features of MS [13], a high-salt diet augmented disease onset and severity and correlated with enhanced breakdown of the blood-brain barrier (BBB) as well as more severe brain pathology [10, 11]. Exacerbated disease was accompanied by increased induction of Th17 cells and elevated numbers of central nervous system (CNS)-infiltrating pathogenic Th17 cells. In MS patients, a recently published study demonstrated an enhanced disease activity, relapse risk, and increased magnetic resonance imaging (MRI) activity in subjects with an increased dietary sodium intake [9].
Similar to a high-salt diet, an increased intake of long-chain fatty acids (LCFA), like lauric acid (LA), exacerbated autoimmunity in the CNS [14]. This was due to an increased infiltration of Th1 and Th17 cells in the spinal cord. In vitro, the differentiation of murine and human CD4+ T cells into Th1 and Th17 cells was significantly increased by the addition of LCFA. In murine EAE, diets rich in LCFA were shown to modulate the microbiome, exposing CD4+ T cells to increased LCFA concentrations in the small intestine, thus inducing pro-inflammatory T cell responses [14].
Of note, the consumption of “Westernized food,” including high-salt, high-fat, high-protein, and high-sugar intake, has already been associated with an increased prevalence of various diseases [15, 16]. However, there is still little understanding on the mechanisms linking environmental factors to disease mechanisms, genetic predisposition, and the immune system. Here, we investigated whether a combination of increased NaCl and LA may have additional effects on CD4+ T cell populations in vitro and in vivo, thus further underlining the relevance of a “Western diet” as a risk factor for autoimmune diseases.

Methods

Isolation of naïve T cells

Splenic T cells were isolated by magnetic-activated cell sorting using the “pan T cell isolation kit II” according to the manufacturer’s instructions (Miltenyi Biotech, Bergisch Gladbach, Germany). Cells were fluorescently stained with an antibody cocktail containing αCD4-FITC (RM4-5, eBioscience, San Diego, CA), αCD44-PE (IM7, BioLegend, San Diego, CA), αCD62L-APC (MEL-14, eBioscience), and αCD25-PECy5 (PC61.5, eBioscience) and were subsequently isolated by fluorescence-activated cell sorting on MoFlo (Beckman-Coulter, Brea, CA) in the FACS core unit in Erlangen, Germany.

T cell culture and differentiation

Sorted naïve T cells (CD4+CD62L+CD44lowCD25) were stimulated by plate-bound anti-CD3 (2 μg/ml, 145-2C11, BD Pharmingen, San Diego, CA) and soluble anti-CD28 (2 μg/ml, 37.51, BD Pharmingen) and cultured for 4 days in the presence of interleukin (IL)-6 (40 ng/ml, R&D Systems, Minneapolis, MN) and IL-23 (20 ng/ml, BioLegend) for Th17, IL-12p70 (20 ng/ml, R&D Systems) and anti-IL-4 (10 μg/ml, BioLegend) for Th1, or IL-4 (50 ng/ml, Miltenyi Biotech) and anti-interferon gamma (IFNγ; 10 μg/ml, BioLegend) for Th2 differentiation. To determine the influence of LA and NaCl on T cell differentiation, cells were cultured with and without 250 μM LA and/or an excess of 40 mM NaCl. T cell frequencies were analyzed by flow cytometry (FACSCantoII, BD Biosiences, San Jose, CA).

Animal experiments

Mice strains were backcrossed on C57BL/6J background for at least 10 generations. All mice were housed at the Präklinisches Experimentelles Tierzentrum (PETZ), the animal care facility of the University Erlangen-Nürnberg, under a 12-h day-night cycle and standardized environmental conditions receiving normal chow (SNIFF E15431-34EF R/M; 0.4% NaCl, 4.2% crude fat) and tap water ad libitum. All experiments were in accordance with the German laws for animal protection and were approved by local ethic committees (Erlangen AZ 54-2532.1-56/12 and 55.2 DMS 2532-2-27).
For experiments under the high-salt diet, mice received chow containing 4% NaCl (based on SNIFF E15431-34EF) and tap water containing 1% NaCl. Mice were adapted to high-salt chow for 4 weeks prior to induction of active myelin oligodendrocyte glycoprotein (MOG)35–55 EAE. For experiments under high-fat diet, mice received a chow containing 30.9% crude fat, rich in LA (13.47%; based on SNIFF E15431-34EF). Mice were adapted to high-fat chow 4 weeks before EAE induction. For experiments under high-salt and high-fat diet, mice received a chow containing 30% crude fat and 4% NaCl (based on SNIFF E15431-34EF) and tap water containing 1% NaCl. Mice were adapted to high-salt + high-fat chow 4 weeks before EAE induction.
For EAE induction, 10–12-week-old mice were anesthetized and subcutaneously injected with 200 μg MOG35–55 and 200 μg Complete Freund’s Adjuvant (CFA), containing 4 mg/ml Mycobacterium tuberculosis (H37RA, BD Biosciences). Pertussis toxin (200 ng/mouse, List Biological Laboratories, Campbell, CA) was applied intraperitoneally on days 0 and 2 post immunization (p.i.). Daily clinical evaluation was performed via a 5-point scale.

Isolation of splenic cells

Spleens were removed on day 10 p.i. and disrupted with a 5-ml glass homogenizer. The tissue was then filtered through a 100 μm cell strainer followed by an erythrocyte lysis. After washing with cold PBS, cells were processed by intracellular cytokine staining and ex vivo flow cytometry analysis.

Isolation of CNS-infiltrating cells

The spinal cord was removed on day 21 p.i. and disrupted with a 5-ml glass homogenizer. Tissue was strained through a 100 μm cell strainer followed by a three-step density gradient (30, 45, and 70% isotonic Percoll™, GE Healthcare, Uppsala, Sweden). After centrifugation (20 min at 800g without brake), cells were harvested from the interphases, washed with cold PBS, and re-suspended in medium for intracellular cytokine staining and ex vivo flow cytometry analysis.

In vitro MOG restimulation assay

Splenocytes from EAE mice were obtained on day 10 p.i. and seeded at a density of 6 × 106 cells/ml. MOG35–55 (20 μg/ml) and concanavalin A (1.25 μg/ml) were added for stimulation, and supernatants were harvested 48 h later and analyzed for cytokine production.

Flow cytometry

Ex vivo-obtained splenic cells, CNS-infiltrating cells, and in vitro-differentiated T cells were analyzed by staining for extra- and intracellular markers. Dead cells were excluded by a fixable viability dye eFluor®780 (0.2 μl/test, eBioscience). Nonspecific Fc-mediated interactions were blocked by the addition of 0.5 μl αCD16/32 (93, eBioscience). For surface staining, the cells were treated with the respective fluorochrome-conjugated antibodies: αCD4-FITC (RM4-5, eBioscience) and αCD25-APC (PC61.5, eBioscience). For intracellular cytokine staining, cells were stimulated for 4 h with ionomycin (1 μM, Sigma-Aldrich, St. Louis, MO) and PMA (50 ng/ml, Sigma-Aldrich) in the presence of monensin (2 μM, eBioscience), fixed with 1% paraformaldehyde and made permeable by saponin or Fix/Perm buffer (eBioscience) treatment. Intracellular cytokines were stained with the respective fluorochrome-conjugated antibodies: αFoxP3-PE (FJK-16s, eBioscience), αIFNγ-APC (XMG1.2, eBioscience), αIL-17A-PE (eBio17B7, eBioscience), IL-4-PE (11B11, BioLegend), and GATA3-PerCP/eFluor710 (TWAJ, eBioscience). Probes were measured with a flow cytometer (FACSCantoII, BD Biosiences).

Cytokine detection

Cytokine concentrations in cell culture supernatants were analyzed by enzyme-linked immunosorbent assays for the secretion of IL-17A and IFNγ (DuoSet ELISA Kits, R&D Systems, Minneapolis, MN) according to the manufacturer’s instructions.

Real-time PCR

Total RNA was isolated using the RNeasy kit (Qiagen, Venlo, Netherlands) and reversely transcribed into cDNA using the QuantiTect® Reverse Transcription Kit (Qiagen). PCR reactions were performed with a qTower real-time PCR System (Analytik Jena, Jena, Germany). Relative quantification was performed by the ΔΔCT method, normalizing the target gene expression on actb/β-Actin as a housekeeping gene. The following TaqMan® real-time PCR assays from Thermo Fisher Scientific were used: actb (β-Actin) Mm00607939_s1, ahr Mm00478932_m1, csf2 Mm01290062_m1, il2 Mm00434256_m1, il6 Mm99999064_m1, il17 Mm00439618_m1, il23a Mm01160011_g1, rorc Mm01261019_g1, sgk1 Mm00441380_m1, tbx21 Mm00450960_m1, and tnf Mm00443258_m1.

Statistical analysis

Statistical analysis was performed using GraphPad Prism (GraphPad Software Inc., La Jolla, CA). All in vitro and ex vivo data were analyzed by one-way ANOVA followed by Tukey’s post-test, unpaired t test, or Wilcoxon rank sum test after checking for normal distribution (unless indicated otherwise in the legends). EAE data were analyzed by Kruskal-Wallis test. Data are presented as mean ± SEM; *p < 0.05, **p < 0.01, or ***p < 0.001 were considered to be statistically significant.

Results

NaCl and LA display additive effects on the differentiation of Th17 cells

To investigate a potential additive effect of a combination of NaCl and LA on the differentiation of naïve T cells under Th1, Th17, or Th2 cell-polarizing conditions in vitro, we added either 40 mM NaCl, 250 μM LA, or both to murine CD4+ T cell cultures (Fig. 1ae). In accordance with published data, both NaCl and LA fostered the differentiation of Th17 cells (Fig. 1c) by around 50% as compared to control conditions whereas only LA increased the frequency of Th1 cells (Fig. 1a). Moreover, NaCl and LA diminished Th2 differentiation significantly (Fig. 1e). NaCl together with LA (NaCl + LA) further enhanced the differentiation of Th17 cells but reduced Th2 frequencies by about one third versus NaCl or LA alone (Fig. 1c, e). No additional effect of NaCl + LA was observed for Th1 cell differentiation compared to LA treatment alone (Fig. 1a). NaCl and LA also increased the production of the signature cytokines IFNγ (Fig. 1b) and IL-17 (Fig. 1d) in the supernatants of Th1 or Th17 differentiating cultures, respectively. In accordance with Th17 cell frequencies, an additional effect of NaCl + LA was observed for the production of IL-17 in culture supernatants (Fig. 1d).

NaCl and LA challenge alters the expression profile of Th1 and Th17 cells

Expression analyses of Th1 or Th17 cell gene signatures revealed increased mRNA expression of tbx21 (T-bet) and csf2 (granulocyte macrophage colony-stimulating factor (GM-CSF)) in NaCl + LA-treated Th1 cells compared to controls, NaCl, or LA treatment (Fig. 2a, b). In contrast, the il6 expression was most significantly enhanced by LA alone (Fig. 2c).
In differentiated Th17 cells, NaCl as well as LA and NaCl + LA treatment increased the expression of a number of pro-inflammatory genes (Fig. 2dl). An additional effect of NaCl + LA versus NaCl or LA alone was observed for il17 (Fig. 2d), csf2 (Fig. 2f), and il2 (Fig. 2j) gene expression supporting the increased generation of Th17 cells under high NaCl + LA conditions in vitro. Aryl hydrocarbon receptor (ahr) expression in Th17 cells was similarly increased by NaCl, LA, and NaCl + LA (Fig. 2k), whereas expression of sgk1, as a known key mediator of salt-augmented activation of Th17 cells, was mainly induced by NaCl or NaCl + LA treatment (Fig. 2l).

NaCl + LA-rich diet impacts on Th1 and Th17 cell-mediated CNS autoimmunity in vivo

The distinct in vitro effects of NaCl + LA on naïve CD4+ T cells prompted us to examine their effects in vivo using murine MOG35–55 EAE as a model of Th1/Th17 cell-mediated autoimmunity. C57BL/6J mice were fed standardized and otherwise completely matched diets rich in either NaCl or NaCl + LA and were compared to mice on a control diet after EAE induction. Mice on the NaCl + LA-rich diet did not display different body weights compared to those on the NaCl-rich or control diet (mean ± SEM on day 21 of EAE, 22.2 ± 0.65 g NaCl + LA diet versus 20.5 ± 0.85 g NaCl diet versus 22.0 ± 0.74 g control diet). Mice fed the NaCl- or NaCl + LA-rich diet displayed a more severe disease course compared to controls (Fig. 3a), although disease incidence and mortality were unaffected. Upon ex vivo phenotyping of spinal cord infiltrates by flow cytometry, the NaCl as well as NaCl + LA diet increased Th1 (Fig. 3b) and Th17 (Fig. 3c) cell frequencies in the CNS on day 21 p.i.. However, in contrast to our in vitro experiments, we observed no additional effect of a NaCl- and LA-rich diet on disease severity or CNS cell infiltration compared to a NaCl-rich diet (Fig. 3ac). Analysis of the spleen showed enhanced Th1 (Fig. 3d) and Th17 (Fig. 3e) but decreased Treg (Fig. 3f) cell counts in mice fed a NaCl- or NaCl + LA-rich diet compared to the control group. Well in line with our previous data, a NaCl- and LA-rich diet had no additive effect on T cell counts (Fig. 3df). Ex vivo recall assays revealed an increase of the pro-inflammatory cytokines IFNγ and IL-17A in splenocytes derived from EAE mice on a NaCl-rich or NaCl + LA-rich diet compared to mice fed a control diet (Fig. 3g, h).

Discussion

Recent epidemiological studies reveal that lifestyle factors such as smoking, obesity, and salt intake may constitute risk factors for MS [17, 18]. LCFA, like LA, or NaCl alone are already established environmental triggers of T cell differentiation that may act as risk factors for Th1 and/or Th17 cell-mediated autoimmune diseases like MS [11, 12, 14]. Yet, a so-called Western diet is typically rich in both, NaCl and LCFA. Thus, our present data offer an important extension to the already existing immunological and functional groundwork and epidemiological observations in MS patients and identify a combined challenge of saturated LCFA and NaCl together as additional dietary, non-infectious triggers involved in T cell differentiation.
Here, we show that NaCl + LA treatment exerts direct effects on Th1, Th2, and Th17 cells under polarizing conditions in vitro and, most importantly, enhances Th17 cell differentiation to a much greater extent than LA or NaCl alone. The concentration of sodium in the plasma is approximately 140 mM, similar to standard cell culture media. However, in the interstitium and lymphoid tissue, considerably higher sodium concentrations may be encountered. This setting is mimicked by adding an excess of 40 mM NaCl to cell culture media which is able to induce pathogenic Th17 cells [11, 19]. Thus, this dietary challenge may be a mechanism for decreasing immune activation in the blood while favoring a pro-inflammatory response in lymphoid tissues. With a regulatory role of Th2 responses in autoimmune neuroinflammation [20], a shift from Th2 towards Th1 cell polarization may add to the detrimental effects of a high-salt and/or high-fat diet alongside with opposing effects on Th17 and Treg cells. The lack of an additional effect of a combined LA and NaCl challenge on Th1 cells may be explained by several factors. All NaCl-induced effects on immune cells so far have been ascribed to Th17 cells [11], Treg cells [21], and macrophages including M2 polarization [22]. No comparable outcomes on the differentiation of Th1 cells were observed [11]. These studies support the concept that, in contrast to fatty acids, distinct immune cell subtypes respond differently to NaCl and that the salt-induced generation of a pro-inflammatory environment involves specific effects on immune cells rather than unspecific activation of all lymphocytes and antigen-presenting cells [12]. Therefore, in contrast to Th17 cell differentiation, NaCl- and LA-induced signaling cascades are not cooperative to further induce Th1 polarization.
Furthermore, the responsible receptors mediating effects of NaCl or LA remain to be identified. Given the plasticity of Th cells and their complex, divergent intercellular signaling pathways during differentiation, it is likely that several mechanisms act in concert. In addition to mitogen-activated protein (MAP) kinase family members, SGK1, which is critically involved in Th17 differentiation [10], was differentially expressed in LA- as well as NaCl-challenged T cells under Th17 cell-polarizing conditions.
In the setting of autoimmunity, a NaCl- and LA-rich diet contributed to a more severe course of EAE and enhanced Th1 and Th17 infiltration into the CNS to the same extent as a NaCl-rich diet alone. It is well conceivable that the pro-inflammatory microenvironment generated by dietary NaCl in immune compartments of the body cannot be further boosted by the addition of LCFA leading to similar disease severity in the animal model.
Although no long-term clinical trials currently exist, our data suggest that the cellular and molecular immunological basis underlying the impact of dietary components like saturated fats and sodium on inflammatory processes and autoimmunity deserves further investigation. While no definite associations between dietary restrictions and the modulation of autoimmune diseases have been firmly established yet, a large proportion of patients already consider special diets or dietary supplements as alternative therapeutic measures. In the first place, the efficacy of dietary interventions in autoimmune diseases may crucially depend on how well the disease pathology is controlled by established immunomodulatory and anti-inflammatory treatment options. Thus, further studies combining standard immunotherapy with dietary approaches in patients are highly warranted.

Conclusions

LCFA, like LA, together with excessive NaCl enhance the differentiation of Th1 and Th17 cells but reduce Th2 frequencies. For Th2 and Th17 cells, an additive effect of LA and NaCl was observed in differentiation assays and on IL-17, GM-CSF, and IL-2 gene expression. In the EAE model, a combination of a NaCl- and LA-rich diet aggravated the disease course and increased T cell infiltration into the CNS to the same extent as dietary NaCl. In summary, different nutritional components may exert independent effects on distinct cells of the immune system but may also influence the functioning of these cells via cooperative mechanisms. These data further underline the relevance of a “Western diet” as a risk factor for MS. Our findings may help to better understand the pathophysiology of MS, thus ultimately enabling protocols for dietary intervention studies in neuroinflammation.

Acknowledgements

We wish to thank S. Seubert for expert technical assistance. Cell sorting was supported by the FACS Core Unit, Nikolaus-Fiebiger-Center, Friedrich-Alexander-University Erlangen-Nürnberg, Germany.

Funding

MKL was supported by the European Research Council (ERC) under the European Union’s Horizon 2020 research and innovation program (640116), by a SALK grant from the government of Flanders, Belgium, and by an Odysseus grant of the Research Foundation Flanders (FWO), Belgium.

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Disclosures

RAL received compensation for activities with or research support from Allmirall, Bayer Health Care, Biogen, Fresenius, Genzyme, Merck, Novartis Pharma, Roche, and TEVA. MKL received compensation for activities with TEVA. RG received compensation for activities with or research support from Bayer Health Care, Biogen, Genzyme, Merck, Novartis Pharma, and TEVA. AH received compensation for activities with or research support from Bayer Health Care, Biogen, Fresenius, Novartis Pharma, TEVA.

Ethics approval

All experiments were in accordance with the German laws for animal protection and were approved by local ethic committees (Erlangen AZ 54-2532.1-56/12 and 55.2 DMS 2532-2-27).
Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis 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.
Literatur
2.
Zurück zum Zitat Houzen H, Niino M, Hata D, Nakano F, Kikuchi S, Fukazawa T, et al. Increasing prevalence and incidence of multiple sclerosis in northern Japan. Mult Scler J. 2008;14:887–92.CrossRef Houzen H, Niino M, Hata D, Nakano F, Kikuchi S, Fukazawa T, et al. Increasing prevalence and incidence of multiple sclerosis in northern Japan. Mult Scler J. 2008;14:887–92.CrossRef
3.
Zurück zum Zitat Jörg S, Grohme DA, Erzler M, Binsfeld M, Haghikia A, Müller DN, et al. Environmental factors in autoimmune diseases and their role in multiple sclerosis. Cell Mol Life Sci. 2016;73:4611–22.CrossRefPubMedPubMedCentral Jörg S, Grohme DA, Erzler M, Binsfeld M, Haghikia A, Müller DN, et al. Environmental factors in autoimmune diseases and their role in multiple sclerosis. Cell Mol Life Sci. 2016;73:4611–22.CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Manzel A, Muller DN, Hafler DA, Erdman SE, Linker RA, Kleinewietfeld M. Role of “western diet” in inflammatory autoimmune diseases. Curr Allergy Asthma Rep. 2014;14:404.CrossRefPubMedPubMedCentral Manzel A, Muller DN, Hafler DA, Erdman SE, Linker RA, Kleinewietfeld M. Role of “western diet” in inflammatory autoimmune diseases. Curr Allergy Asthma Rep. 2014;14:404.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Mozaffarian D, Fahimi S, Singh GM, Micha R, Khatibzadeh S, Engell RE, et al. Global sodium consumption and death from cardiovascular causes. N Engl J Med. 2014;371:624–34.CrossRefPubMed Mozaffarian D, Fahimi S, Singh GM, Micha R, Khatibzadeh S, Engell RE, et al. Global sodium consumption and death from cardiovascular causes. N Engl J Med. 2014;371:624–34.CrossRefPubMed
6.
Zurück zum Zitat D’Elia L, Galletti F, Strazzullo P. Dietary salt intake and risk of gastric cancer. Cancer Treat Res. 2014;159:83–95.CrossRefPubMed D’Elia L, Galletti F, Strazzullo P. Dietary salt intake and risk of gastric cancer. Cancer Treat Res. 2014;159:83–95.CrossRefPubMed
7.
Zurück zum Zitat Sundström B, Johansson I, Rantapää-Dahlqvist S. Interaction between dietary sodium and smoking increases the risk for rheumatoid arthritis: results from a nested case-control study. Rheumatology (Oxford). 2015;54:487–93.CrossRef Sundström B, Johansson I, Rantapää-Dahlqvist S. Interaction between dietary sodium and smoking increases the risk for rheumatoid arthritis: results from a nested case-control study. Rheumatology (Oxford). 2015;54:487–93.CrossRef
8.
Zurück zum Zitat Hucke S, Wiendl H, Klotz L. Implications of dietary salt intake for multiple sclerosis pathogenesis. Mult Scler. 2016;22:133–9.CrossRefPubMed Hucke S, Wiendl H, Klotz L. Implications of dietary salt intake for multiple sclerosis pathogenesis. Mult Scler. 2016;22:133–9.CrossRefPubMed
9.
Zurück zum Zitat Farez MF, Fiol MP, Gaitán MI, Quintana FJ, Correale J. Sodium intake is associated with increased disease activity in multiple sclerosis. J Neurol Neurosurg Psychiatry. 2015;86:26–31.CrossRefPubMed Farez MF, Fiol MP, Gaitán MI, Quintana FJ, Correale J. Sodium intake is associated with increased disease activity in multiple sclerosis. J Neurol Neurosurg Psychiatry. 2015;86:26–31.CrossRefPubMed
10.
Zurück zum Zitat 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;496:513–7.CrossRefPubMedPubMedCentral 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;496:513–7.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat 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;496:518–22.CrossRefPubMedPubMedCentral 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;496:518–22.CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Jörg S, Kissel J, Manzel A, Kleinewietfeld M, Haghikia A, Gold R, et al. High salt drives Th17 responses in experimental autoimmune encephalomyelitis without impacting myeloid dendritic cells. Exp Neurol. 2016;279:212–22.CrossRefPubMed Jörg S, Kissel J, Manzel A, Kleinewietfeld M, Haghikia A, Gold R, et al. High salt drives Th17 responses in experimental autoimmune encephalomyelitis without impacting myeloid dendritic cells. Exp Neurol. 2016;279:212–22.CrossRefPubMed
13.
Zurück zum Zitat 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;129:1953–71.CrossRef 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;129:1953–71.CrossRef
14.
Zurück zum Zitat Haghikia A, Jörg S, Duscha A, Berg J, Manzel A, Waschbisch A, et al. Dietary fatty acids directly impact central nervous system autoimmunity via the small intestine. Immunity. 2015;43:817–29.CrossRefPubMed Haghikia A, Jörg S, Duscha A, Berg J, Manzel A, Waschbisch A, et al. Dietary fatty acids directly impact central nervous system autoimmunity via the small intestine. Immunity. 2015;43:817–29.CrossRefPubMed
15.
Zurück zum Zitat Odegaard AO, Koh WP, Yuan J-M, Gross MD, Pereira MA. Western-style fast food intake and cardiometabolic risk in an eastern country. Circulation. 2012;126:182–8.CrossRefPubMedPubMedCentral Odegaard AO, Koh WP, Yuan J-M, Gross MD, Pereira MA. Western-style fast food intake and cardiometabolic risk in an eastern country. Circulation. 2012;126:182–8.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Thorburn AN, Macia L, Mackay CR. Diet, metabolites, and “western-lifestyle” inflammatory diseases. Immunity. 2014;40:833–42.CrossRefPubMed Thorburn AN, Macia L, Mackay CR. Diet, metabolites, and “western-lifestyle” inflammatory diseases. Immunity. 2014;40:833–42.CrossRefPubMed
17.
Zurück zum Zitat Hedström AK, Sundqvist E, Bäärnhielm M, Nordin N, Hillert J, Kockum I, et al. Smoking and two human leukocyte antigen genes interact to increase the risk for multiple sclerosis. Brain J Neurol. 2011;134:653–64.CrossRef Hedström AK, Sundqvist E, Bäärnhielm M, Nordin N, Hillert J, Kockum I, et al. Smoking and two human leukocyte antigen genes interact to increase the risk for multiple sclerosis. Brain J Neurol. 2011;134:653–64.CrossRef
18.
Zurück zum Zitat Hedström AK, Lima Bomfim I, Barcellos L, Gianfrancesco M, Schaefer C, Kockum I, et al. Interaction between adolescent obesity and HLA risk genes in the etiology of multiple sclerosis. Neurology. 2014;82:865–72.CrossRefPubMedPubMedCentral Hedström AK, Lima Bomfim I, Barcellos L, Gianfrancesco M, Schaefer C, Kockum I, et al. Interaction between adolescent obesity and HLA risk genes in the etiology of multiple sclerosis. Neurology. 2014;82:865–72.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Machnik A, Neuhofer W, Jantsch J, Dahlmann A, Tammela T, Machura K, et al. Macrophages regulate salt-dependent volume and blood pressure by a vascular endothelial growth factor-C-dependent buffering mechanism. Nat Med. 2009;15:545–52.CrossRefPubMed Machnik A, Neuhofer W, Jantsch J, Dahlmann A, Tammela T, Machura K, et al. Macrophages regulate salt-dependent volume and blood pressure by a vascular endothelial growth factor-C-dependent buffering mechanism. Nat Med. 2009;15:545–52.CrossRefPubMed
20.
Zurück zum Zitat Kuchroo VK, Prabhu Das M, Brown JA, Ranger AM, Zamvil SS, Sobel RA, et al. B7-1 and B7-2 costimulatory molecules activate differentially the Th1/Th2 developmental pathways: application to autoimmune disease therapy. Cell. 1995;80:707–18.CrossRefPubMed Kuchroo VK, Prabhu Das M, Brown JA, Ranger AM, Zamvil SS, Sobel RA, et al. B7-1 and B7-2 costimulatory molecules activate differentially the Th1/Th2 developmental pathways: application to autoimmune disease therapy. Cell. 1995;80:707–18.CrossRefPubMed
21.
Zurück zum Zitat Hernandez AL, Kitz A, Wu C, Lowther DE, Rodriguez DM, Vudattu N, et al. Sodium chloride inhibits the suppressive function of FOXP3+ regulatory T cells. J Clin Invest. 2015;125:4212–22.CrossRefPubMedPubMedCentral Hernandez AL, Kitz A, Wu C, Lowther DE, Rodriguez DM, Vudattu N, et al. Sodium chloride inhibits the suppressive function of FOXP3+ regulatory T cells. J Clin Invest. 2015;125:4212–22.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat 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;125:4223–38.CrossRefPubMedPubMedCentral 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;125:4223–38.CrossRefPubMedPubMedCentral
Metadaten
Titel
Impact of combined sodium chloride and saturated long-chain fatty acid challenge on the differentiation of T helper cells in neuroinflammation
verfasst von
Anna Hammer
Anne Schliep
Stefanie Jörg
Aiden Haghikia
Ralf Gold
Markus Kleinewietfeld
Dominik N. Müller
Ralf A. Linker
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
Journal of Neuroinflammation / Ausgabe 1/2017
Elektronische ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-017-0954-y

Weitere Artikel der Ausgabe 1/2017

Journal of Neuroinflammation 1/2017 Zur Ausgabe