scholarly journals Melatonin Prevents T Lymphocyte Infiltration to the Kidneys of Hypertensive Rats, Induced by a High-Salt Diet, by Preventing the Expression of CXCR3 Ligand Chemokines

Nutrients ◽  
2021 ◽  
Vol 13 (10) ◽  
pp. 3577
Author(s):  
Ariel Bier ◽  
Rawan Khashab ◽  
Yehonatan Sharabi ◽  
Ehud Grossman ◽  
Avshalom Leibowitz

In a previous study, we demonstrated that melatonin prevents kidney damage in a salt-induced hypertension model by decreasing oxidative stress. We hypothesized that this effect involves melatonin’s immunomodulatory properties. In vivo Study-Dahl salt-sensitive (DSS) rats were fed normal chow, a high-salt diet (HSD), or a HSD and melatonin (30 mg/kg/day) in their water for eight weeks. Kidneys were harvested for immediate lymphocyte isolation and characterization by Flow cytometry (CD3+CD4+ and CD3+CD8+) and for lymphocyte chemoattractant (mainly CXCL chemokines) gene expression studies. In vitro study-rat mesangial cells (RMC) were cultured in a high-salt medium without and with melatonin. A HSD was associated with significant renal infiltration of CD4+ and CD8+ T lymphocytes compared to control. Melatonin significantly reduced renal lymphocyte infiltration. A HSD significantly increased mRNA expression of CXCL chemokines. Adding melatonin to the HSD abolished this effect. Treating RMC cells with salt increased the expression of CXCL10 and CXCL11 but not CXCL9. Adding melatonin to the culture media prevented this increase. Treating HSD-fed rats with melatonin decreased renal lymphocyte chemoattractant mRNA expression and is associated with significantly reducing renal T lymphocyte infiltration. Salt may have a direct effect on chemokine-producing renal cells, which is blunted by melatonin treatment.

2013 ◽  
Vol 6 (273) ◽  
pp. ec97-ec97 ◽  
Author(s):  
Annalisa M. VanHook

In addition to contributing to the immune response against pathogens, helper T (TH ) cells that produce the cytokine interleukin-17 (IL-17) also contribute to autoimmune diseases. Maintenance of both normal and pathogenic TH17 cell activities depends on activation of the IL-23 receptor (IL-23R). By performing transcriptional profiling and network analysis of transcriptional changes in wild-type and Il23r–/– mouse T cells that were activated and induced to differentiate into TH17 cells, Wu et al. identified serum glucocorticoid kinase 1 (Sgk1) as a key node downstream of IL-23R. In vitro differentiation of naïve T cells from Sgk1–/– mice revealed that SGK1 was not required for primary TH17 cell differentiation but was required for maintenance of TH17 cells and continued signaling through IL-23R. Analysis of experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis, in Sgk1–/– animals showed that these mice had reduced incidence of disease, severity of symptoms, and production of IL-17 compared with EAE in wild-type animals. In vitro experiments were consistent with a model in which SGK1 phosphorylates the transcription factor Foxo1 to repress its ability to indirectly activate Il23r expression. SGK1 mediates sodium (Na+) homeostasis by modulating the activity of epithelial Na+ channels, so the authors tested the effect of Na+ on TH17 cell differentiation. Increasing the concentration of NaCl in the culture medium increased expression of Sgk1, Il23r, Il17, and other genes associated with TH17 differentiation in wild-type, but not Sgk1–/–, T cells that had been activated but not treated with factors to influence their development into a particular type of TH cell. Compared with a normal diet, a high-salt diet increased the number of TH17 cells in the guts of wild-type mice but induced a milder increase in the abundance of TH17 cells in Sgk1–/– mice. In the EAE model, mice on a high-salt diet showed increased severity of disease compared with those fed a normal diet. However, a high-salt diet had a much milder effect on disease symptoms in Sgk1–/– mice. In a related study, Kleinewietfeld etal. differentiated naïve human T cells in culture conditions that mimicked the interstitial fluid of animals fed a high-salt diet and found that the additional NaCl promoted differentiation of TH17 cells that expressed markers consistent with autoimmune activity. Further experiments indicated that this effect was mediated by the kinase p38, the transcription factor and p38 target NFAT5, and the NFAT5 target Sgk1. In vivo experiments performed in this study were consistent with those reported by Wu et al. These studies suggest that production of the pathogenic TH17 cells that contribute to autoimmunity may be exacerbated by dietary salt. Commentary by O’Shea and Jones considers the implications and limitations of these findings in the context of autoimmune disease.C. Wu, N. Yosef, T. Thalhamer, C. Zhu, S. Xiao, Y. Kishi, A. Regev, V. K. Kuchroo, Induction of pathogenic TH17 cells by inducible salt-sensing kinase SGK1. Nature496, 513–517 (2013). [PubMed]M. Kleinewietfeld, A. Manzel, J. Titze, H. Kvakan, N. Yosef, R. A. Linker, D. N. Muller, D. A. Hafler, Sodium chloride drives autoimmune disease by the induction of pathogenic TH17 cells. Nature496, 518–522 (2013). [PubMed]J. J. O’Shea, R. G. Jones, Rubbing salt in the wound. Nature496, 437–439 (2013). [PubMed]


2022 ◽  
Vol 8 (1) ◽  
Author(s):  
Juexiao Gong ◽  
Man Luo ◽  
Yonghong Yong ◽  
Shan Zhong ◽  
Peng Li

AbstractAlamandine (Ala) is a novel member of the renin–angiotensin-system (RAS) family. The present study aimed to explore the effects of Ala on hypertension and renal damage of Dahl salt-sensitive (SS) rats high-salt diet-induced, and the mechanisms of Ala on renal-damage alleviation. Dahl rats were fed with high-salt diets to induce hypertension and renal damage in vivo, and HK-2 cells were treated with sodium chloride (NaCl) to induce renal injury in vitro. Ala administration alleviated the high-salt diet-induced hypertension, renal dysfunction, and renal fibrosis and apoptosis in Dahl SS rats. The HK-2 cells’ damage, and the increases in the levels of cleaved (c)-caspase3, c-caspase8, and c-poly(ADP-ribose) polymerase (PARP) induced by NaCl were inhibited by Ala. Ala attenuated the NaCl-induced oxidative stress in the kidney and HK-2 cells. DETC, an inhibitor of SOD, reversed the inhibitory effect of Ala on the apoptosis of HK-2 cells induced by NaCl. The NaCl-induced increase in the PKC level was suppressed by Ala in HK-2 cells. Notably, PKC overexpression reversed the moderating effects of Ala on the NaCl-induced apoptosis of HK-2 cells. These results show that Ala alleviates high-salt diet-induced hypertension and renal dysfunction. Ala attenuates the renal damage via inhibiting the PKC/reactive oxygen species (ROS) signaling pathway, thereby suppressing the apoptosis in renal tubular cells.


2004 ◽  
Vol 287 (1) ◽  
pp. F95-F101 ◽  
Author(s):  
Frank Schweda ◽  
Martin Kammerl ◽  
Charlotte Wagner ◽  
Bernhard K. Krämer ◽  
Armin Kurtz

Although the regulation of cyclooxygenase-2 (COX-2) expression in the kidney cortex has been extensively characterized, the physiological control mechanisms of COX-2 expression at the level of the kidney and at the level of the tubular cells are not well understood. Based on the current hypothesis that tubular salt transport might be a crucial regulator of COX-2 expression, this study aimed to determine the impact of salt delivery to the tubules (glomerular filtration) for the regulation of COX-2 in the kidney cortex in vivo. To this end, glomerular filtration of the right kidney was abrogated by the ligation of the right ureter of male Sprague-Dawley rats. After 1 wk of ligation, the animals were treated with subcutaneous infusions of furosemide (12 mg·kg−1·day−1) or with a low-salt or a high-salt diet (0.02% wt/wt; 8% wt/wt), and COX-2 as well as renin mRNA expression were determined in the ligated and the nonligated contralateral kidney. During ureteral ligation, hydronephrosis developed with a reduction of medullary mass, while the cortex was preserved. Expressions of the Na-K-2Cl cotransporter isoforms A and B were both reduced in the hydronephrotic cortex to 70 and 35% of the corresponding contralateral intact kidney. Despite the abrogation of glomerular filtration, detected by inulin clearance measurements, renocortical COX-2 mRNA abundance was stimulated by furosemide treatment (3.2-fold) or low-salt diet (2.9-fold) to similar degrees compared with the intact contralateral kidney (2.7-fold for both treatments), whereas a high-salt diet did not significantly suppress COX-2 mRNA in the macula densa region of either kidney. Renin mRNA expression was regulated strictly in parallel in both kidneys, a low-salt diet or furosemide treatment stimulating and a high-salt diet suppressing it. We conclude from these findings that salt delivery to the tubules is not an essential requirement for the upregulation of COX-2 by salt deficiency or by loop diuretics in the rat kidney cortex nor is it for chronic stimulation of renin mRNA expression.


1981 ◽  
Vol 241 (5) ◽  
pp. F517-F524 ◽  
Author(s):  
P. Chaumet-Riffaud ◽  
J. P. Oudinet ◽  
J. Sraer ◽  
C. Lajotte ◽  
R. Ardaillou

Prostaglandin (PG) E2 and F2 alpha synthesis by isolated glomeruli and papillary homogenates prepared from control, salt-loaded, and salt-depleted rats was measured in vitro with and without added arachidonic acid using specific radioimmunoassays. Glomeruli from salt-depleted rats synthesized less PGE2 and more PGF2 alpha than glomeruli from control rats under both conditions. The effect of sodium restriction could be attributed to stimulation of glomerular 9-keto-PGE2 reductase activity unrelated to a change in the concentration of this enzyme. High salt diet had no effect on PG synthesis by glomeruli. Papillary homogenates prepared from salt-loaded rats synthesized more PGE2 than those from control rats both with and without added arachidonic acid. This finding suggests an effect of high salt diet at a stage further than phospholipid deacylation. Low salt diet had no effect on PG synthesis by papillary homogenates. The physiological control of PG synthesis in response to changes in the NaCl content of the diet is, therefore, different for the glomeruli and the papilla.


2020 ◽  
Vol 318 (4) ◽  
pp. F982-F993
Author(s):  
Ammar J. Alsheikh ◽  
John Henry Dasinger ◽  
Justine M. Abais-Battad ◽  
Daniel J. Fehrenbach ◽  
Chun Yang ◽  
...  

Studies examining mechanisms of Dahl salt-sensitive (SS) hypertension have implicated the infiltration of leukocytes in the kidneys, which contribute to renal disease and elevated blood pressure. However, the signaling pathways by which leukocytes traffic to the kidneys remain poorly understood. The present study nominated a signaling pathway by analyzing a kidney RNA sequencing data set from SS rats fed either a low-salt (0.4% NaCl) diet or a high-salt (4.0% NaCl) diet. From this analysis, chemokine (C-C motif) ligand 2 (CCL2) and chemokine (C-C motif) receptor 2 (CCR2) were nominated as a potential pathway modifying renal leukocyte infiltration and contributing to SS hypertension. The functional role of the CCL2/CCR2 pathway was tested by daily administration of CCR2 antagonist (RS-102895 at 5 mg·kg−1·day−1 in DMSO) or DMSO vehicle for 3 or 21 days by intraperitoneal injections during the high salt challenge. Blood pressure, renal leukocyte infiltration, and renal damage were evaluated. The results demonstrated that RS-102895 treatment ameliorated renal damage (urinary albumin excretion; 43.4 ± 5.1 vs. 114.7 ± 15.2 mg/day in vehicle, P < 0.001) and hypertension (144.3 ± 2.2 vs. 158.9 ± 4.8 mmHg in vehicle, P < 0.001) after 21 days of high-salt diet. It was determined that renal leukocyte infiltration was blunted by day 3 of the high-salt diet (1.4 ± 0.1 vs. 1.9 ± 0.2 in vehicle × 106 CD45+ cells/kidney, P = 0.034). An in vitro chemotaxis assay validated the effect of RS-102895 on leukocyte chemotaxis toward CCL2. The results suggest that increased CCL2 in SS kidneys is important in the early recruitment of leukocytes, and blockade of this recruitment by administering RS-102895 subsequently blunted the renal damage and hypertension.


2008 ◽  
Vol 294 (6) ◽  
pp. H2814-H2821 ◽  
Author(s):  
Micheline M. de Resende ◽  
Andrew S. Greene

We have previously shown that skeletal muscle angiogenesis induced by electrical stimulation is significantly attenuated when SS-13BN/Mcwi rats are fed a high-salt diet. This effect was associated with a large increase in endothelial cell (EC) apoptosis. We hypothesized that the low levels of ANG II during high-salt diet would increase EC apoptosis and consequently diminish the angiogenic response. To test this hypothesis, a series of in vitro and in vivo studies was performed. EC apoptosis and viability were evaluated after incubation with ANG II under serum-free conditions. After 24 h of incubation, ANG II increased EC viability and Bcl-2-to-Bax ratio along with a dose-dependent decrease in EC apoptosis. This effect was blocked by the ANG II type 1 receptor antagonist losartan. To confirm our in vitro results, ANG II (3 ng·kg−1·min−1) was chronically infused in rats fed a high-salt diet (4% NaCl). ANG II decreased EC apoptosis and produced a significant increase (40%) in skeletal muscle angiogenesis after electrical stimulation. These in vivo results were in agreement with our in vitro results and demonstrate that the attenuation of ANG II levels during a high-salt diet may induce EC apoptosis and consequently block the angiogenic response induced by electrical stimulation. Furthermore, under normal conditions, ANG II increases EC viability and protects EC from apoptosis possibly by inactivation of the mitochondrial apoptotic pathway.


2020 ◽  
Vol 22 (11) ◽  
pp. 2152-2155
Author(s):  
Eliane F. E. Wenstedt ◽  
Ester B. M. Remmerswaal ◽  
Nelly D. Bom‐Baylon ◽  
Esmee M. Schrooten ◽  
Frederike J. Bemelman ◽  
...  

2021 ◽  
Vol 18 (1) ◽  
Author(s):  
Mengyan Hu ◽  
Yinyao Lin ◽  
Xuejiao Men ◽  
Shisi Wang ◽  
Xiaobo Sun ◽  
...  

Abstract Background A high-salt diet (HSD) is one of the major risk factors for acute ischemic stroke (AIS). As a potential mechanism, surplus salt intake primes macrophages towards a proinflammatory phenotype. In this study, whether HSD could blunt the efferocytic capability of macrophages after ischemic stroke, thus exacerbating post-stroke neural inflammation, was investigated. Methods Wild-type male C57BL/6 mice were fed with fodder containing 8% sodium chloride for 4 weeks and subjected to transient middle cerebral occlusion (tMCAO). Disease severity, macrophage polarization as well as efferocytic capability were evaluated. Bone marrow-derived macrophages were cultured in vitro, and the impact of high salinity on their efferocytic activity, as well as their expression of phagocytic molecules, were analyzed. The relationships among sodium concentration, macrophage phenotype, and disease severity in AIS patients were explored. Results HSD-fed mice displayed increased infarct volume and aggravated neurological deficiency. Mice fed with HSD suffered exacerbated neural inflammation as shown by higher inflammatory mediator expression and immune cell infiltration levels. Infiltrated macrophages within stroke lesions in HSD-fed mice exhibited a shift towards proinflammatory phenotype and impaired efferocytic capability. As assessed with a PCR array, the expression of triggering receptor expressed on myeloid cells 2 (TREM2), a receptor relevant to phagocytosis, was downregulated in high-salt-treated bone marrow-derived macrophages. Enhancement of TREM2 signaling restored the efferocytic capacity and cellular inflammation resolution of macrophages in a high salinity environment in vitro and in vivo. A high concentration of urine sodium in AIS patients was found to be correlated with lower TREM2 expression and detrimental stroke outcomes. Conclusions HSD inhibited the efferocytic capacity of macrophages by downregulating TREM2 expression, thus impeding inflammation resolution after ischemic stroke. Enhancing TREM2 signaling in monocytes/macrophages could be a promising therapeutic strategy to enhance efferocytosis and promote post-stroke inflammation resolution.


Diabetes ◽  
2020 ◽  
Vol 69 (Supplement 1) ◽  
pp. 463-P
Author(s):  
TOMONORI KIMURA ◽  
YOSHITAKA HASHIMOTO ◽  
TAKAFUMI SENMARU ◽  
EMI USHIGOME ◽  
MASAHIDE HAMAGUCHI ◽  
...  
Keyword(s):  

Sign in / Sign up

Export Citation Format

Share Document