Abstract

Salt intake as part of a western diet currently exceeds recommended limits, and the small amount found in the natural diet enjoyed by our Paleolithic ancestors. Excess salt is associated with the development of hypertension and cardiovascular disease, but other adverse effects of excess salt intake are beginning to be recognized, including the development of autoimmune and inflammatory disease. Over the last decade there has been an increasing body of evidence demonstrating that salt affects multiple components of both the innate and adaptive immune systems. In this review we outline the recent laboratory, animal and human data, highlighting the effect of salt on immunity, with a particular focus on the relevance to inflammatory kidney disease.

INTRODUCTION

Salt [sodium chloride (NaCl)] has been a prominent feature of the human diet for a relatively short time, at least in evolutionary terms. Our ancestors survived for millions of years on a diet that contained <1 g of salt per day. Salt is required for the maintenance of extracellular and therefore plasma volume, and given the limited salt composition of this natural diet, humans evolved under an intense evolutionary pressure for the selection of salt-conserving genes. Salt was discovered as a way of preserving food ∼5000 years ago and since then salt intake has increased rapidly to current estimates of 8 g/day in the UK, with even greater intake in other parts of the world [1]. Most of this salt is found in processed foods and restaurant meals; the remaining salt is added while cooking and eating. Current salt intake as part of a western diet is therefore far in excess of the amount we are physiologically programmed to handle.

Given our ability to retain salt, in steady state the minimum amount required to replace losses is <0.5 g/day. There is good evidence from both observational and interventional studies that excess salt intake is associated with the development of hypertension [2, 3], it is generally accepted that excess salt intake is linked to the development of cardiovascular disease [4], and there is some evidence for a role in the progression of chronic kidney disease (CKD) [5]. Based on this, the World Health Organization recommends limiting salt intake to <5 g/day, which it estimates could prevent 2.5 million deaths globally each year.

There are, however, perhaps less well-appreciated adverse effects of excess salt intake that are becoming apparent. These include the development of malignancy, in particular gastric cancer [6], obesity and metabolic syndrome [7]. Excess salt is also implicated in autoimmune and inflammatory disease. Observational studies have demonstrated that increased salt intake is associated with the development of rheumatoid arthritis [8] and correlates with disease severity in multiple sclerosis [9]. Furthermore, in renal transplantation, perioperative salt loading worsens short-term outcomes [10] and adherence to a low salt diet improves renal outcomes and survival in the longer term [11]. In this review we outline the recent data from laboratory, animal and human studies that demonstrate an effect of salt on both innate and adaptive immunity and postulate how these findings may be relevant in inflammatory kidney disease.

SALT AND INNATE IMMUNITY

Initial evidence of an effect of salt on macrophages

The mechanism by which excess salt causes hypertension is unclear and cannot be fully explained by an effect on extracellular volume [12]. Over the last decade, however, two concepts have been described that not only provide novel insights into the pathogenesis of hypertension, but also provide the initial evidence of salt impacting immune cells. First, it has been shown that salt loading results in sodium storage at interstitial sites within muscle and skin [13, 14]. This results in sodium concentrations in interstitial compartments (up to 250 mM) in excess of that found in plasma, where sodium is tightly regulated due to its effect on osmolality. Second, is the increasing awareness of a role for inflammation in the development of hypertension.

A link between both these concepts was demonstrated in a seminal paper in 2009 that demonstrated a high salt diet in rats results in elevated skin sodium concentrations, which in turn activates resident macrophages, which through production of vascular endothelial growth factor C, clear this stored sodium through the lymphatic system [13]. Macrophage depletion promoted skin sodium storage and the development of hypertension. Hence salt loading leads to storage at interstitial sites, with NaCl concentrations in excess of plasma, resulting in immune cell activation and salt clearance via the lymphatic system. Either excess salt or impaired immune-mediated salt clearance can lead to hypertension.

Further studies on the effect of salt on macrophage activation and its relevance in the clearance of cutaneous infection

Following this initial work, there have been several other studies investigating how salt affects macrophage activation. Like other cells of the immune system, macrophages display a spectrum of activation states with plasticity along this spectrum. At one end are pro-inflammatory M1 macrophages, which secrete inflammatory cytokines interleukin-1 (IL-1), IL-6, IL-12 and tumour necrosis factor-α (TNF-α) and activate effector T cells (Teffs), and at the other end are reparative M2 macrophages that secrete anti-inflammatory cytokines IL-10 and transforming growth factor-β (TGF-β), with reduction in Teffs and an increase in regulatory T cell (Treg) activity [15]. M2 macrophage activity is important in wound healing, and experimentally, they have been shown to reduce the development of autoimmune disease [16, 17].

Salt has been shown to have a pro-inflammatory effect on macrophages, increasing M1 activation and reducing M2 activation (Figure 1) [18, 19]. In cell culture, the addition of 40 mM NaCl increases M1 macrophage activation and this is dependent on a signalling pathway involving p38 mitogen-activated protein kinase (MAPK) and the transcription factor nuclear factor of activated T-cells 5 (NFAT5; also known as tonicity-responsive enhancer binding protein) [18]. Sodium accumulates at sites of skin infection in mice. In humans, adding NaCl to cell cultures in vitro led to increased M1 clearance of infection, and a high salt diet in animals resulted in enhanced clearance of cutaneous leishmaniasis. In contrast, M2 activation is blunted in vitro in high salt culture conditions, and a high salt diet is associated with reduced wound healing and reduced M2 activity in a mouse model of peritonitis [19]. Unlike the salt effect on M1 macrophages, and indeed that on T helper 17 (Th17) cells, which is dependent on NFAT5, the inhibitory salt effect on M2 macrophages is dependent on a reduction in signalling through a pathway involving protein kinase B (PKB/Akt) and the mammalian target of rapamycin.

The effect of salt on M1 and M2 macrophage polarization and the intracellular pathways involved.
FIGURE 1

The effect of salt on M1 and M2 macrophage polarization and the intracellular pathways involved.

Taken together, these data on macrophage activation states and high interstitial NaCl concentrations provide insight into the potential evolutionary reasons why the immune system responds to salt. Infection is associated with the creation of localized hypersalinity, which allows for local activation of pro-inflammatory cells while limiting such activation in the body more generally. A similar high salt environment is created at sites of body–environmental interface (e.g. skin, kidney, genitourinary tract), providing protection from pathogens at these locations.

The effect of salt on renal mononuclear phagocytes and the development of pyelonephritis

Further evidence for this role of hypersalinity in providing protection from infection was provided in a recent study that assessed the role of interstitial sodium concentrations on tissue resident mononuclear phagocytes (MNPs) in the kidney [20]. Tissue MNPs include distinct subsets of macrophages and dendritic cells. Berry et al. [20] demonstrated that CD14+ MNPs, a subset of MNPs with enhanced phagocytic capability, were enriched in the medulla compared with the cortex of human kidneys; this positioning was dependent on NFAT5-mediated production of the chemokines chemokine ligand 2 (CCL2) and chemokine ligand 1 (CX3CL1) by renal tubular epithelial cells in response to hypersalinity. Medullary CD14+ MNPs demonstrated increased phagocytosis of uropathogenic Escherichia coli and phagocytic activity increased with increasing extracellular sodium concentrations in an NFAT5-dependent manner. The induction of nephrogenic diabetes insipidus (NDI) resulted in increased bacteraemia and death following intravesical uropathogenic E. coli challenge in mice. This was interpreted as being due to disruption of the medullary renal sodium gradient. Moreover, NDI in patients, either in the setting of tolvaptan use or as a result of sickle cell disease, is associated with increased urinary tract infection risk [20]. Taken together, these data demonstrate that high interstitial salt concentrations in the kidney, primarily required for water reabsorption and the maintenance of extracellular volume, also promote medullary localization of specific subsets of MNPs, which in turn provide protection from infection. It is tempting to speculate that autoimmune kidney disease might arise in part due to the hypertonic environment found in the kidneys, created through evolution for these volume and infectious purposes.

SALT AND ADAPTIVE IMMUNITY

CD4+ T cell polarization and the imbalance of Th17 cells and Tregs in inflammatory kidney disease

Antigen is presented to naïve CD4+ T cells on major histocompatibility complex class II (MHC II). This is done by a select group of antigen-presenting cells, including dendritic cells, macrophages and B cells. After antigen presentation, naïve CD4+ T cells become activated and polarized towards one of the number of CD4+ T cell subsets, including Th1, Th2, Th17 and Tregs, each of which has different effects on the immune response, in part through the cell-specific cytokines produced.

Th17 cells are pro-inflammatory and provide protection against extracellular bacterial and fungal infections, particularly at epithelial cell surfaces [21]. Polarization requires stimulation by IL-1β, IL-6 and TGF-β, whereas full maturation requires IL-21 and IL-23. Th17 cells secrete various cytokines, including IL-17, which acts on stromal cells and is important for neutrophil recruitment, and others such as IL-26 and IL-22, which act on non-immune cells, enhancing inflammation or serving to attenuate damage, depending on the tissue and environment. Tregs have opposing effects to Th17 cells. Naturally occurring and peripherally induced Tregs are characterized by expression of the transcription factor Forkhead Box P3 (FOXP3) and provide peripheral tolerance through inhibiting the function of effector CD4+ and CD8+ T cells, B cells and cells of the innate immune system through cell–cell contact mechanisms and release of suppressive cytokines (e.g. TGF-β, IL-10) [22].

Th17 cells and Tregs both play a major role in autoimmune kidney disease and renal transplant rejection. Th17 cells are implicated in the development of both glomerular and tubulointerstitial disease in patients and animal models, including anti-neutrophil cytoplasmic antibody–associated glomerulonephritis [23, 24], lupus nephritis [25], immunoglobulin A nephropathy [26] and primary Sjögren’s syndrome–associated tubulointerstitial nephritis [27]. Conversely, Treg activity limits inflammatory glomerular disease and autoimmune disease relapses, both clinically and in experimental models [28]. Furthermore, it is becoming increasingly clear that excessive Th17 and reduced Treg activity are important components in the development of acute and chronic allograft rejection [29]. Hence excess Th17 activation and reduced Treg activity underlie a number of inflammatory kidney diseases in both the native and transplant setting. This augmented Th17 and attenuated Treg phenomenon is the same effect that salt has on these CD4+ T cell subtypes, as we describe below.

The effect of salt on Th17 cells and the development of multiple sclerosis

Two studies published in Nature in 2013 provided the initial evidence of an effect of salt on Th17 polarization [30, 31]. Kleinewietfeld et al. [30] isolated naïve CD4+ T cells from healthy volunteers and cultured these for 7 days in optimal Th17 polarizing conditions in standard media and in media supplemented with varying concentrations of NaCl (0–80 mM). There was a dose-dependent increase in Th17 polarization with salt up to 40 mM NaCl, which provided optimal polarization and cell survival. The authors concluded, on the basis of control experiments in which media was supplemented with sodium gluconate, magnesium chloride and mannitol, that sodium, as opposed to chloride or tonicity, was mediating this pro-inflammatory effect. Further in vitro work showed that salt stimulation affected naïve rather than memory CD4+ T cells, did not influence Th1 and Th2 polarization and not only polarized Th17 cells, but also promoted a pathogenic Th17 phenotype (increased expression of IL-2, TNF-α, IL-9 and colony-stimulating factor 2/granulocyte-macrophage colony-stimulating factor). It was subsequently shown that the intracellular pathway mediating this response to hypersalinity involved upregulation of phosphorylated p38 MAPK, NFAT5 and serum- and glucocorticoid-regulated kinase 1 (SGK1) (Figure 2). Knocking down any of these molecules resulted in a sharp reduction in salt-mediated Th17 polarization. The authors then investigated the salt effect on Th17 cells in vivo in an animal model of multiple sclerosis [experimental autoimmune encephalitis (EAE)], a Th17-mediated disease. A high salt diet exacerbated disease severity, and this was associated with increased Th17 cells in the central nervous system (CNS). The use of a p38 MAPK inhibitor abrogated both salt-induced Th17 CNS infiltration and clinical markers of disease severity.

Intracellular pathways involved in salt-induced Th17 polarization.
FIGURE 2

Intracellular pathways involved in salt-induced Th17 polarization.

Wu et al. [32] focused further on the role of SGK1 in salt-mediated Th17 polarization. They described the pathway downstream of SGK1, which involves phosphorylation of Forkhead box protein O1 (FOXO1) and upregulation of the main Th17 transcription factor, RAR-related orphan receptor gamma T (RORγT) (Figure 2). They also confirmed the polarizing effect of salt on Th17 cells in vitro and demonstrated that this Th17 polarization was prevented in SGK1 knockout cells. They showed that a high salt diet in mice is associated with increased gut Th17 cell frequency, confirmed that a high salt diet exacerbates EAE and showed that this was prevented in SGK1 knockout mice. Interestingly, since this initial work, the same group has demonstrated a pivotal role of SGK1 in mediating the balance between Th17 and Tregs in isotonic conditions [32]. Taken together, these initial studies demonstrate that Th17 cell polarization is enhanced by high salt conditions both in vitro and in vivo, that this is dependent on signalling through p38 MAPK, NFAT5 and SGK1 and that salt-mediated Th17 polarization exacerbates animal models of autoimmune disease.

The effect of salt on suppressor Treg function and transplant rejection

Given the interrelationship between Th17 cells and Tregs and the effect of salt on Th17 polarization, subsequent work investigated whether there was an effect of salt on Treg function, first in vitro and then in mouse models of graft-versus-host disease (GVHD), colitis and mismatched cardiac transplantation [33, 34]. Hernandez et al. [33] demonstrated that the addition of salt to co-culture of Tregs with effector T cells inhibited regulatory cell function in a dose-dependent manner over the range of 10–40 mM NaCl. Further analysis of purified Tregs cultured under high salt conditions demonstrated that the mechanism of this loss of suppressor function was a Treg shift to a Th1 phenotype with upregulation of pro-inflammatory molecules such as interferon γ (IFN-γ); knockdown of IFN-γ resulted in maintenance of the suppressor function. The authors demonstrated that the same pathway increased SGK1 activity and phosphorylation of FOXO1 and FOXO3, which is involved in Th17–Treg balance in isotonic conditions, and in the polarization of Th17 in hypertonic conditions, which also mediates the salt effect on Tregs. Inhibition or knockdown of SGK1 resulted in recovery of the suppressor function; high salt conditions increased phosphorylation of FOXO1 and FOXO3, which resulted in impaired stabilization of the FOXP3 locus. This inhibitory effect of salt on Tregs was then confirmed in vivo. A high salt diet was associated with increased IFN-γ-secreting Tregs in the spleen and mesenteric lymph nodes, and this was associated with increased SGK1 expression. Moreover, a high salt diet increased IFN-γ-secreting Tregs and worsened disease in a mouse model of GVHD, while transfer of Tregs cultured under high salt, as opposed to standard conditions, led to exacerbation of disease in an animal model of colitis [33].

Safa et al. [34] investigated the effect of high salt on the alloimmune response, using a mouse model of MHC II–mismatched cardiac transplantation, which is dependent on Treg suppressor function for graft survival. Despite having no effect on blood pressure, a high salt diet reduced allograft survival, and this was associated with a reduction in Tregs within the transplant and the spleen. This high salt effect on rejection was abolished in SGK1 knockout mice. Together, these initial Treg studies demonstrate an inhibitory effect of salt on Treg suppressor function, which is mediated through an SGK1–FOXO1/3 pathway. High salt exacerbates animal models of autoimmune disease and worsens murine transplant rejection.

The effect of salt on the development of autoimmune diseases

Subsequent studies have investigated further aspects of the effect of salt on immunity in models of multiple sclerosis (EAE), colitis and lupus nephritis and also in the setting of the progression of acute kidney injury (AKI) to CKD. The results of this work are summarized below.

Animal models of multiple sclerosis

Wilck et al. [35] proposed an alternative indirect in vivo mechanism by which high salt affects Th17 cells and the development of EAE. This was through alteration in the gut microbiome and provides a link to studies that demonstrate gut-derived Th17 cells mediate the development of extra-intestinal disease, including inflammatory kidney disease [36]. Salt loading inhibited the growth of Lactobacillus sp. in vitro in mice and in healthy human volunteers. In an EAE model, Lactobacillus administration ameliorated Th17 CNS infiltration and the development of disease. The mechanism of the effect on Th17 polarization was proposed to be due to reduced bacterial production of indole derivatives, which have previously been shown to associate with improvement in EAE. Hence multiple extracellular cues may affect Th17 polarization and several pathways may mediate the pro-inflammatory salt effect in vivo.

Jorg et al. [37] investigated whether alteration of dendritic cell function occurred in response to salt and mediated the in vitro effects of salt on Th17 and the development of EAE. They found no effect on dendritic cell function, suggesting the salt effect on Th17 is a direct effect on Th17 cells themselves. In contrast, Barbaro et al. [38] demonstrated that high salt conditions increased both macrophage and dendritic cell activation and that co-culture of salt activated dendritic cells with T cells from salt-sensitive mice led to increased pro-inflammatory cytokine production (IL-17 and IFN-γ) from CD4+ and CD8+ cells. The intracellular pathway mediating this effect involved sodium entry into dendritic cells through amiloride-sensitive sodium channels, formation of reactive oxygen species and the production of isolevuglandin protein adducts, which ultimately enhanced antigen presentation to T cells.

Animal models of inflammatory bowel disease

Several studies have investigated the role of salt on intestinal immunity and the development of colitis, perhaps unsurprising given the abundance of Th17 cells within the gut [33, 39, 40]. In these studies, salt loading of mice without disease results in increased gut Th17 cells and reduced Treg suppressor function. This is associated with increased SGK1 expression, increased gut permeability and histological features of inflammation. High salt has also been shown to worsen mouse models of inflammatory bowel disease, associated with increased gut IL-17. One study investigated the source of this IL-17 and demonstrated that salt not only affects CD4+ T cells, but also innate lymphoid cells (ILCs), in particular ILC3, the innate lymphoid equivalent of Th17 cells [40]. Indeed, recombination-activating gene knockout (i.e. lymphocyte depleted) mice still get IL-17-mediated gut inflammation in response to a high salt diet, highlighting that salt promotes IL-17 production from multiple immune cells.

Animal models of systemic lupus erythematosus

Elevated salt concentrations in vitro have been shown to increase follicular helper T cell polarization and a high salt diet worsened lupus features in an animal model [41]. A further study demonstrated that a high salt diet leads to exacerbated experimental lupus nephritis, with more significant disease on histology, increased proteinuria and worse survival [42]. This was associated with increased Th17 and Th1 cells in the spleen and fewer Tregs. The salt response of isolated CD4+ T cells from patients with systemic lupus erythematosus was then analysed. Th17 cell polarization increased with anti-CD3 and anti-CD28 stimulation in the presence of NaCl. This, as in healthy volunteers, was abrogated by the addition of an SGK1 inhibitor. Of note, high salt conditions do not promote increased polarization of naïve T cells to Th1 cells in vitro under Th1 polarizing conditions [43], but this and other studies have shown a high salt diet does promote a Th1 phenotype in vivo [42, 43]. This may reflect CD4+ T cell plasticity, with Th17 cells or Tregs developing a pro-inflammatory Th1 phenotype with expression of IFN-γ in response to high salt conditions.

Animal models of AKI

AKI, particularly if recurrent, can lead to CKD, and high salt has been shown to increase the progression of AKI to CKD in animal models [44]. The mechanism behind this phenomenon is unclear but has been proposed to be immune mediated, as immunosuppression with mycophenolate mofetil abrogates the salt effect. More recent work has demonstrated that Th17 cells mediate interstitial inflammation in AKI; salt loading worsened Th17-mediated inflammation and this resulted in more significant CKD [45]. This was prevented by angiotensin receptor blockade, which was shown in vitro to directly affect Th17 cells.

Human studies of dietary salt modification and their effect on Th17–Treg balance

Given the growing body of laboratory and animal work that have demonstrated a polarizing effect of salt on diverse immune cells (summarized in Table 1), using dietary modifications to limit salt intake in the management of patients with autoimmune disease provides an intriguing and potentially beneficial, low-cost therapeutic avenue. Initial interventional studies have investigated the effect of changing salt intake on the immune response in healthy volunteers and in patients with autoimmune disease. The results of these studies are outlined in Table 2. Together, these demonstrate dietary salt modification may be used to modulate the immune response to varying degrees, but whether this translates to changes in clinical outcomes is yet to be determined. Moreover, where salt exerts its pro-inflammatory effect on immune cells in vivo is unknown.

Table 1

Summary of the known effects of salt on immune cell function, the intracellular pathways involved and the in vivo clinical consequences of the high salt effect

Cell typeSalt effectIntracellular signalling molecules and pathways mediating the salt effectClinical consequences of high salt effect in vivo
Innate immune system
M1 macrophage [18]Activationp38 MAPK, NFAT5Enhanced clearance of cutaneous infection
M2 macrophage [19]InhibitionAkt, mammalian target of rapamycinReduced wound healing
Monocytes [46]Expansion of CD14++CD16+ (intermediate) monocytesFormation of reactive oxygen speciesRenal hypoxia and inflammation
Tissue resident MNPs [20]Increased phagocytic activity of CD14+ MNPsNFAT5Protection against pyelonephritis
Dendritic cell [38]ActivationNicotinamide adenine dinucleotide phosphate oxidase (reactive oxygen species formation), formation of isolevuglandin protein adductsHypertension
Neutrophils [40]Activation as a consequence of increased IL-23 production (direct effect unknown)UnknownExacerbated gut inflammation
ILCs [40]Activation of ILC3UnknownExacerbated gut inflammation
Adaptive immune system
Th17 [30, 31, 39, 40, 42, 45]Activationp38 MAPK, NFAT5, SGK1, FOXO1Exacerbated EAE, colitis and lupus nephritis. Increased progression AKI to CKD
Treg [33, 34]InhibitionSGK1, FOXO1/3Exacerbated transplant rejection, EAE
Th1 [42]Increased activity in vivo, possibly conversion from Th17 or Treg (no direct effect in vitro)N/AExacerbated lupus nephritis
Th2No effect [30]N/AN/A
Follicular helper T cell [41]ActivationTET2Exacerbated SLE
CD8+ T cell [38]Increased IFN-γ and IL-17 from CD8+ cells from salt-sensitive mice when cultured with salt primed dendritic cellsUnknownUnknown
B cellsUnknownUnknownUnknown
Cell typeSalt effectIntracellular signalling molecules and pathways mediating the salt effectClinical consequences of high salt effect in vivo
Innate immune system
M1 macrophage [18]Activationp38 MAPK, NFAT5Enhanced clearance of cutaneous infection
M2 macrophage [19]InhibitionAkt, mammalian target of rapamycinReduced wound healing
Monocytes [46]Expansion of CD14++CD16+ (intermediate) monocytesFormation of reactive oxygen speciesRenal hypoxia and inflammation
Tissue resident MNPs [20]Increased phagocytic activity of CD14+ MNPsNFAT5Protection against pyelonephritis
Dendritic cell [38]ActivationNicotinamide adenine dinucleotide phosphate oxidase (reactive oxygen species formation), formation of isolevuglandin protein adductsHypertension
Neutrophils [40]Activation as a consequence of increased IL-23 production (direct effect unknown)UnknownExacerbated gut inflammation
ILCs [40]Activation of ILC3UnknownExacerbated gut inflammation
Adaptive immune system
Th17 [30, 31, 39, 40, 42, 45]Activationp38 MAPK, NFAT5, SGK1, FOXO1Exacerbated EAE, colitis and lupus nephritis. Increased progression AKI to CKD
Treg [33, 34]InhibitionSGK1, FOXO1/3Exacerbated transplant rejection, EAE
Th1 [42]Increased activity in vivo, possibly conversion from Th17 or Treg (no direct effect in vitro)N/AExacerbated lupus nephritis
Th2No effect [30]N/AN/A
Follicular helper T cell [41]ActivationTET2Exacerbated SLE
CD8+ T cell [38]Increased IFN-γ and IL-17 from CD8+ cells from salt-sensitive mice when cultured with salt primed dendritic cellsUnknownUnknown
B cellsUnknownUnknownUnknown
Table 1

Summary of the known effects of salt on immune cell function, the intracellular pathways involved and the in vivo clinical consequences of the high salt effect

Cell typeSalt effectIntracellular signalling molecules and pathways mediating the salt effectClinical consequences of high salt effect in vivo
Innate immune system
M1 macrophage [18]Activationp38 MAPK, NFAT5Enhanced clearance of cutaneous infection
M2 macrophage [19]InhibitionAkt, mammalian target of rapamycinReduced wound healing
Monocytes [46]Expansion of CD14++CD16+ (intermediate) monocytesFormation of reactive oxygen speciesRenal hypoxia and inflammation
Tissue resident MNPs [20]Increased phagocytic activity of CD14+ MNPsNFAT5Protection against pyelonephritis
Dendritic cell [38]ActivationNicotinamide adenine dinucleotide phosphate oxidase (reactive oxygen species formation), formation of isolevuglandin protein adductsHypertension
Neutrophils [40]Activation as a consequence of increased IL-23 production (direct effect unknown)UnknownExacerbated gut inflammation
ILCs [40]Activation of ILC3UnknownExacerbated gut inflammation
Adaptive immune system
Th17 [30, 31, 39, 40, 42, 45]Activationp38 MAPK, NFAT5, SGK1, FOXO1Exacerbated EAE, colitis and lupus nephritis. Increased progression AKI to CKD
Treg [33, 34]InhibitionSGK1, FOXO1/3Exacerbated transplant rejection, EAE
Th1 [42]Increased activity in vivo, possibly conversion from Th17 or Treg (no direct effect in vitro)N/AExacerbated lupus nephritis
Th2No effect [30]N/AN/A
Follicular helper T cell [41]ActivationTET2Exacerbated SLE
CD8+ T cell [38]Increased IFN-γ and IL-17 from CD8+ cells from salt-sensitive mice when cultured with salt primed dendritic cellsUnknownUnknown
B cellsUnknownUnknownUnknown
Cell typeSalt effectIntracellular signalling molecules and pathways mediating the salt effectClinical consequences of high salt effect in vivo
Innate immune system
M1 macrophage [18]Activationp38 MAPK, NFAT5Enhanced clearance of cutaneous infection
M2 macrophage [19]InhibitionAkt, mammalian target of rapamycinReduced wound healing
Monocytes [46]Expansion of CD14++CD16+ (intermediate) monocytesFormation of reactive oxygen speciesRenal hypoxia and inflammation
Tissue resident MNPs [20]Increased phagocytic activity of CD14+ MNPsNFAT5Protection against pyelonephritis
Dendritic cell [38]ActivationNicotinamide adenine dinucleotide phosphate oxidase (reactive oxygen species formation), formation of isolevuglandin protein adductsHypertension
Neutrophils [40]Activation as a consequence of increased IL-23 production (direct effect unknown)UnknownExacerbated gut inflammation
ILCs [40]Activation of ILC3UnknownExacerbated gut inflammation
Adaptive immune system
Th17 [30, 31, 39, 40, 42, 45]Activationp38 MAPK, NFAT5, SGK1, FOXO1Exacerbated EAE, colitis and lupus nephritis. Increased progression AKI to CKD
Treg [33, 34]InhibitionSGK1, FOXO1/3Exacerbated transplant rejection, EAE
Th1 [42]Increased activity in vivo, possibly conversion from Th17 or Treg (no direct effect in vitro)N/AExacerbated lupus nephritis
Th2No effect [30]N/AN/A
Follicular helper T cell [41]ActivationTET2Exacerbated SLE
CD8+ T cell [38]Increased IFN-γ and IL-17 from CD8+ cells from salt-sensitive mice when cultured with salt primed dendritic cellsUnknownUnknown
B cellsUnknownUnknownUnknown
Table 2

Summary of studies investigating the immunological effects of dietary salt modification in healthy human volunteers and in patients with autoimmune disease

ReferencesSubjects includedDietary modificationMain results
Zhou et al. [46]Healthy volunteers (n = 20)3-day run-in on normal diet, then fixed daily salt intake of 15 g (high salt) for 7 days, then 5 g (low salt) for 7 days. Food provided to subjectsIncrease in CD14++CD16+ (intermediate) monocytes; decrease in CD14++CD16(classical) and CD14+CD16++ (non-classical) monocytes with high salt intake
Yi et al. [47] (part of Mars flight simulation study)Healthy volunteers (n = 6)Fixed daily salt intake of 12 g, 9 g, then 6 g for 1–2 months, then reverted back to 12 g for 1 month. Food provided to subjectsSerum cytokines analysed after each intervention period: increased pro-inflammatory cytokines (IL-6 and IL-23) and reduced anti-inflammatory cytokines (IL-10) with high salt intake
Luo et al. [48]Healthy volunteers (n = 15)3-day run-in on normal diet, then fixed daily salt intake of 15 g (high salt) for 7 days, then 5 g (low salt) for 7 days. Food provided to subjectsT cell subset analysis: increased Th17 and reduced Treg populations with high salt intake. Associated with increased expression of NFAT5 and SGK1
Wen et al. [49]Healthy volunteers (n = 49)Fixed daily salt intake of 3 g (low salt) for 7 days, then 7 days high salt (18 g), then 7 days high salt (18 g) plus potassium supplementation (4.5 g KCl; 60 mmol K)Serum IL-17 and PBMC IL-17 mRNA increased with high salt. Associated with increased SGK1. High salt effect abolished with potassium supplementation of diet
Scrivo et al. [50]Patients with RA (n = 15) and SLE (n = 15)Dietary counselling used to limit daily salt intake to 5 g for 3 weeks, then reverted back to standard diet for 2 weeks. 24-h urine Na measurement used to assess adherence to dietRA patients: no effect in Th17 or Treg subsets. SLE patients: increase in Treg after salt restriction; no effect on Th17 cells
ReferencesSubjects includedDietary modificationMain results
Zhou et al. [46]Healthy volunteers (n = 20)3-day run-in on normal diet, then fixed daily salt intake of 15 g (high salt) for 7 days, then 5 g (low salt) for 7 days. Food provided to subjectsIncrease in CD14++CD16+ (intermediate) monocytes; decrease in CD14++CD16(classical) and CD14+CD16++ (non-classical) monocytes with high salt intake
Yi et al. [47] (part of Mars flight simulation study)Healthy volunteers (n = 6)Fixed daily salt intake of 12 g, 9 g, then 6 g for 1–2 months, then reverted back to 12 g for 1 month. Food provided to subjectsSerum cytokines analysed after each intervention period: increased pro-inflammatory cytokines (IL-6 and IL-23) and reduced anti-inflammatory cytokines (IL-10) with high salt intake
Luo et al. [48]Healthy volunteers (n = 15)3-day run-in on normal diet, then fixed daily salt intake of 15 g (high salt) for 7 days, then 5 g (low salt) for 7 days. Food provided to subjectsT cell subset analysis: increased Th17 and reduced Treg populations with high salt intake. Associated with increased expression of NFAT5 and SGK1
Wen et al. [49]Healthy volunteers (n = 49)Fixed daily salt intake of 3 g (low salt) for 7 days, then 7 days high salt (18 g), then 7 days high salt (18 g) plus potassium supplementation (4.5 g KCl; 60 mmol K)Serum IL-17 and PBMC IL-17 mRNA increased with high salt. Associated with increased SGK1. High salt effect abolished with potassium supplementation of diet
Scrivo et al. [50]Patients with RA (n = 15) and SLE (n = 15)Dietary counselling used to limit daily salt intake to 5 g for 3 weeks, then reverted back to standard diet for 2 weeks. 24-h urine Na measurement used to assess adherence to dietRA patients: no effect in Th17 or Treg subsets. SLE patients: increase in Treg after salt restriction; no effect on Th17 cells

KCl, potassium chloride; PBMC, peripheral blood mononuclear cell; RA, rheumatoid arthritis; SLE, systemic lupus erythematosus.

Table 2

Summary of studies investigating the immunological effects of dietary salt modification in healthy human volunteers and in patients with autoimmune disease

ReferencesSubjects includedDietary modificationMain results
Zhou et al. [46]Healthy volunteers (n = 20)3-day run-in on normal diet, then fixed daily salt intake of 15 g (high salt) for 7 days, then 5 g (low salt) for 7 days. Food provided to subjectsIncrease in CD14++CD16+ (intermediate) monocytes; decrease in CD14++CD16(classical) and CD14+CD16++ (non-classical) monocytes with high salt intake
Yi et al. [47] (part of Mars flight simulation study)Healthy volunteers (n = 6)Fixed daily salt intake of 12 g, 9 g, then 6 g for 1–2 months, then reverted back to 12 g for 1 month. Food provided to subjectsSerum cytokines analysed after each intervention period: increased pro-inflammatory cytokines (IL-6 and IL-23) and reduced anti-inflammatory cytokines (IL-10) with high salt intake
Luo et al. [48]Healthy volunteers (n = 15)3-day run-in on normal diet, then fixed daily salt intake of 15 g (high salt) for 7 days, then 5 g (low salt) for 7 days. Food provided to subjectsT cell subset analysis: increased Th17 and reduced Treg populations with high salt intake. Associated with increased expression of NFAT5 and SGK1
Wen et al. [49]Healthy volunteers (n = 49)Fixed daily salt intake of 3 g (low salt) for 7 days, then 7 days high salt (18 g), then 7 days high salt (18 g) plus potassium supplementation (4.5 g KCl; 60 mmol K)Serum IL-17 and PBMC IL-17 mRNA increased with high salt. Associated with increased SGK1. High salt effect abolished with potassium supplementation of diet
Scrivo et al. [50]Patients with RA (n = 15) and SLE (n = 15)Dietary counselling used to limit daily salt intake to 5 g for 3 weeks, then reverted back to standard diet for 2 weeks. 24-h urine Na measurement used to assess adherence to dietRA patients: no effect in Th17 or Treg subsets. SLE patients: increase in Treg after salt restriction; no effect on Th17 cells
ReferencesSubjects includedDietary modificationMain results
Zhou et al. [46]Healthy volunteers (n = 20)3-day run-in on normal diet, then fixed daily salt intake of 15 g (high salt) for 7 days, then 5 g (low salt) for 7 days. Food provided to subjectsIncrease in CD14++CD16+ (intermediate) monocytes; decrease in CD14++CD16(classical) and CD14+CD16++ (non-classical) monocytes with high salt intake
Yi et al. [47] (part of Mars flight simulation study)Healthy volunteers (n = 6)Fixed daily salt intake of 12 g, 9 g, then 6 g for 1–2 months, then reverted back to 12 g for 1 month. Food provided to subjectsSerum cytokines analysed after each intervention period: increased pro-inflammatory cytokines (IL-6 and IL-23) and reduced anti-inflammatory cytokines (IL-10) with high salt intake
Luo et al. [48]Healthy volunteers (n = 15)3-day run-in on normal diet, then fixed daily salt intake of 15 g (high salt) for 7 days, then 5 g (low salt) for 7 days. Food provided to subjectsT cell subset analysis: increased Th17 and reduced Treg populations with high salt intake. Associated with increased expression of NFAT5 and SGK1
Wen et al. [49]Healthy volunteers (n = 49)Fixed daily salt intake of 3 g (low salt) for 7 days, then 7 days high salt (18 g), then 7 days high salt (18 g) plus potassium supplementation (4.5 g KCl; 60 mmol K)Serum IL-17 and PBMC IL-17 mRNA increased with high salt. Associated with increased SGK1. High salt effect abolished with potassium supplementation of diet
Scrivo et al. [50]Patients with RA (n = 15) and SLE (n = 15)Dietary counselling used to limit daily salt intake to 5 g for 3 weeks, then reverted back to standard diet for 2 weeks. 24-h urine Na measurement used to assess adherence to dietRA patients: no effect in Th17 or Treg subsets. SLE patients: increase in Treg after salt restriction; no effect on Th17 cells

KCl, potassium chloride; PBMC, peripheral blood mononuclear cell; RA, rheumatoid arthritis; SLE, systemic lupus erythematosus.

CONCLUSION

The immune system consists of opposing pro-inflammatory and anti-inflammatory cells. Salt affects the activity of multiple cells of both innate and adaptive immunity, promoting predominantly pro-inflammatory subtypes. In vitro, salt increases activation of M1 macrophages and Th17 cells, whereas it suppresses M2 macrophage and Treg function. Intracellular molecules that mediate this effect include NFAT5 and SGK1. The in vivo consequences of hypersalinity are increased clearance of infection, but also the development of autoimmune disease. Whether dietary salt manipulation can be used as an adjunctive therapy in inflammatory kidney diseases or transplantation remains to be investigated.

FUNDING

RE is a Kidney Research UK funded Clinical Training Fellow (Grant code: TF_007_20161125).

CONFLICT OF INTEREST STATEMENT

None declared.

REFERENCES

1

Powles
J
,
Fahimi
S
,
Micha
R
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
:
e003733

2

Intersalt Cooperative Research Group.

Intersalt: an international study of electrolyte excretion and blood pressure. Results for 24 hour urinary sodium and potassium excretion
.
BMJ
1988
;
297
:
319
328

3

Sacks
FM
,
Svetkey
LP
,
Vollmer
WM
et al.
Effects on blood pressure of reduced dietary sodium and the Dietary Approaches to Stop Hypertension (DASH) diet
.
N Engl J Med
2001
;
344
:
3
10

4

Whelton
PK
,
Appel
LJ
,
Sacco
RL
et al.
Sodium, blood pressure, and cardiovascular disease: further evidence supporting the American Heart Association sodium reduction recommendations
.
Circulation
2012
;
126
:
2880
2889

5

Smyth
A
,
O'Donnell
MJ
,
Yusuf
S
et al.
Sodium intake and renal outcomes: a systematic review
.
Am J Hypertens
2014
;
27
:
1277
1284

6

Joossens
JV
,
Hill
MJ
,
Elliott
P
et al.
Dietary salt, nitrate and stomach cancer mortality in 24 countries
.
Int J Epidemiol
1996
;
25
:
494
504

7

Lanaspa
MA
,
Kuwabara
M
,
Andres-Hernando
A
et al.
High salt intake causes leptin resistance and obesity in mice by stimulating endogenous fructose production and metabolism
.
Proc Natl Acad Sci USA
2018
;
115
:
3138
3143

8

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
493

9

Farez
MF
,
Fiol
MP
,
Gaitán
MI
et al.
Sodium intake is associated with increased disease activity in multiple sclerosis
.
J Neurol Neurosurg Psychiatry
2015
;
86
:
26
31

10

Adwaney
A
,
Randall
DW
,
Blunden
MJ
et al.
Perioperative Plasma-Lyte use reduces the incidence of renal replacement therapy and hyperkalaemia following renal transplantation when compared with 0.9% saline: a retrospective cohort study
.
Clin Kidney J
2017
;
10
:
838
844

11

Osté
MCJ
,
Gomes-Neto
AW
,
Corpeleijn
E
et al.
Dietary Approach to Stop Hypertension (DASH) diet and risk of renal function decline and all-cause mortality in renal transplant recipients
.
Am J Transplant
2018
;
18
:
2523
2533

12

Wenzel
UO
,
Bode
M
,
Kurts
C
et al.
Salt, inflammation, IL-17 and hypertension
.
Br J Pharmacol
2018
. doi:10.1111/bph.14359

13

Machnik
A
,
Neuhofer
W
,
Jantsch
J
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
552

14

Kopp
C
,
Linz
P
,
Dahlmann
A
et al.
23Na magnetic resonance imaging-determined tissue sodium in healthy subjects and hypertensive patients
.
Hypertens Dallas Tex 1979
2013
;
61
:
635
640

15

Sica
A
,
Mantovani
A.
Macrophage plasticity and polarization: in vivo veritas
.
J Clin Invest
2012
;
122
:
787
795

16

Jiang
H-R
,
Milovanović
M
,
Allan
D
et al.
IL-33 attenuates EAE by suppressing IL-17 and IFN-γ production and inducing alternatively activated macrophages
.
Eur J Immunol
2012
;
42
:
1804
1814

17

Parsa
R
,
Andresen
P
,
Gillett
A
et al.
Adoptive transfer of immunomodulatory M2 macrophages prevents type 1 diabetes in NOD mice
.
Diabetes
2012
;
61
:
2881
2892

18

Jantsch
J
,
Schatz
V
,
Friedrich
D
et al.
Cutaneous Na+ storage strengthens the antimicrobial barrier function of the skin and boosts macrophage-driven host defense
.
Cell Metab
2015
;
21
:
493
501

19

Binger
KJ
,
Gebhardt
M
,
Heinig
M
et al.
High salt reduces the activation of IL-4- and IL-13-stimulated macrophages
.
J Clin Invest
2015
;
125
:
4223
4238

20

Berry
MR
,
Mathews
RJ
,
Ferdinand
JR
et al.
Renal sodium gradient orchestrates a dynamic antibacterial defense zone
.
Cell
2017
;
170
:
860
874

21

Gaffen
SL
,
Jain
R
,
Garg
AV
et al.
The IL-23–IL-17 immune axis: from mechanisms to therapeutic testing
.
Nat Rev Immunol
2014
;
14
:
585
600

22

Kleinewietfeld
M
,
Hafler
DA.
The plasticity of human Treg and Th17 cells and its role in autoimmunity
.
Semin Immunol
2013
;
25
:
305
312

23

Gan
PY
,
Steinmetz
OM
,
Tan
DSY
et al.
Th17 cells promote autoimmune anti-myeloperoxidase glomerulonephritis
.
J Am Soc Nephrol
2010
;
21
:
925
931

24

Nogueira
E
,
Hamour
S
,
Sawant
D
et al.
Serum IL-17 and IL-23 levels and autoantigen-specific Th17 cells are elevated in patients with ANCA-associated vasculitis
.
Nephrol Dial Transplant
2010
;
25
:
2209
2217

25

Rother
N
,
van der Vlag
J.
Disturbed T cell signaling and altered Th17 and regulatory T cell subsets in the pathogenesis of systemic lupus erythematosus
.
Front Immunol
2015
;
6
:
610

26

Peng
Z
,
Tian
J
,
Cui
X
et al.
Increased number of Th22 cells and correlation with Th17 cells in peripheral blood of patients with IgA nephropathy
.
Hum Immunol
2013
;
74
:
1586
1591

27

Evans
R
,
Zdebik
A
,
Ciurtin
C
et al.
Renal involvement in primary Sjögren’s syndrome
.
Rheumatology (Oxford)
2015
;
54
:
1541
8
.

28

Paust
H-J
,
Riedel
J-H
,
Krebs
CF
et al.
CXCR3+ regulatory T cells control TH1 responses in crescentic GN
.
J Am Soc Nephrol
2016
;
27
:
1933
1942

29

Sullivan
JA
,
Adams
AB
,
Burlingham
WJ.
The emerging role of TH17 cells in organ transplantation
.
Transplantation
2014
;
97
:
483
489

30

Kleinewietfeld
M
,
Manzel
A
,
Titze
J
et al.
Sodium chloride drives autoimmune disease by the induction of pathogenic Th17 cells
.
Nature
2013
;
496
:
518
522

31

Wu
C
,
Yosef
N
,
Thalhamer
T
et al.
Induction of pathogenic TH17 cells by inducible salt-sensing kinase SGK1
.
Nature
2013
;
496
:
513
517

32

Wu
C
,
Chen
Z
,
Xiao
S
et al.
SGK1 governs the reciprocal development of Th17 and regulatory T cells
.
Cell Rep
2018
;
22
:
653
665

33

Hernandez
AL
,
Kitz
A
,
Wu
C
et al.
Sodium chloride inhibits the suppressive function of FOXP3+ regulatory T cells
.
J Clin Invest
2015
;
125
:
4212
4222

34

Safa
K
,
Ohori
S
,
Borges
TJ
et al.
Salt accelerates allograft rejection through serum- and glucocorticoid-regulated kinase-1–dependent inhibition of regulatory T cells
.
J Am Soc Nephrol
2015
;
26
:
2341
2347

35

Wilck
N
,
Matus
MG
,
Kearney
SM
et al.
Salt-responsive gut commensal modulates TH17 axis and disease
.
Nature
2017
;
551
:
585
589

36

Krebs
CF
,
Paust
H-J
,
Krohn
S
et al.
Autoimmune renal disease is exacerbated by S1P-receptor-1-dependent intestinal Th17 cell migration to the kidney
.
Immunity
2016
;
45
:
1078
1092

37

Jörg
S
,
Kissel
J
,
Manzel
A
et al.
High salt drives Th17 responses in experimental autoimmune encephalomyelitis without impacting myeloid dendritic cells
.
Exp Neurol
2016
;
279
:
212
222

38

Barbaro
NR
,
Foss
JD
,
Kryshtal
DO
et al.
Dendritic cell amiloride-sensitive channels mediate sodium-induced inflammation and hypertension
.
Cell Rep
2017
;
21
:
1009
1020

39

Wei
Y
,
Lu
C
,
Chen
J
et al.
High salt diet stimulates gut Th17 response and exacerbates TNBS-induced colitis in mice
.
Oncotarget
2017
;
8
:
70
82

40

Aguiar
SLF
,
Miranda
MCG
,
Guimarães
MAF
et al.
High-salt diet induces IL-17-dependent gut inflammation and exacerbates colitis in mice
.
Front Immunol
2017
;
8
:
1969

41

Wu
H
,
Huang
X
,
Qiu
H
et al.
High salt promotes autoimmunity by TET2-induced DNA demethylation and driving the differentiation of Tfh cells
.
Sci Rep
2016
;
6
:
28065

42

Yang
X
,
Yao
G
,
Chen
W
et al.
Exacerbation of lupus nephritis by high sodium chloride related to activation of SGK1 pathway
.
Int Immunopharmacol
2015
;
29
:
568
573

43

Hammer
A
,
Schliep
A
,
Jörg
S
et al.
Impact of combined sodium chloride and saturated long-chain fatty acid challenge on the differentiation of T helper cells in neuroinflammation
.
J Neuroinflammation
2017
;
14
:
184

44

Spurgeon-Pechman
KR
,
Donohoe
DL
,
Mattson
DL
et al.
Recovery from acute renal failure predisposes hypertension and secondary renal disease in response to elevated sodium
.
Am J Physiol Renal Physiol
2007
;
293
:
F269
F278

45

Mehrotra
P
,
Patel
JB
,
Ivancic
CM
et al.
Th-17 cell activation in response to high salt following acute kidney injury is associated with progressive fibrosis and attenuated by AT-1R antagonism
.
Kidney Int
2015
;
88
:
776
784

46

Zhou
X
,
Zhang
L
,
Ji
W-J
et al.
Variation in dietary salt intake induces coordinated dynamics of monocyte subsets and monocyte-platelet aggregates in humans: implications in end organ inflammation
.
PLoS One
2013
;
8
:
e60332

47

Yi
B
,
Titze
J
,
Rykova
M
et al.
Effects of dietary salt levels on monocytic cells and immune responses in healthy human subjects: a longitudinal study
.
Transl Res J Lab Clin Med
2015
;
166
:
103
110

48

Luo
T
,
Ji
W-J
,
Yuan
F
et al.
Th17/Treg imbalance induced by dietary salt variation indicates inflammation of target organs in humans
.
Sci Rep
2016
;
6
:
26767

49

Wen
W
,
Wan
Z
,
Ren
K
et al.
Potassium supplementation inhibits IL-17A production induced by salt loading in human T lymphocytes via p38/MAPK-SGK1 pathway
.
Exp Mol Pathol
2016
;
100
:
370
377

50

Scrivo
R
,
Massaro
L
,
Barbati
C
et al.
The role of dietary sodium intake on the modulation of T helper 17 cells and regulatory T cells in patients with rheumatoid arthritis and systemic lupus erythematosus
.
PLoS One
2017
;
12
:
e0184449

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://dbpia.nl.go.kr/journals/pages/open_access/funder_policies/chorus/standard_publication_model)

Comments

0 Comments
Submit a comment
You have entered an invalid code
Thank you for submitting a comment on this article. Your comment will be reviewed and published at the journal's discretion. Please check for further notifications by email.