Background/Aims: Vasopressin is a powerful stimulator of vascular calcification, augmenting osteogenic signaling in vascular smooth muscle cells (VSMCs) including upregulation of transcription factors such as core-binding factor α-1 (CBFA1), msh homeobox 2 (MSX2), and SRY-Box 9 (SOX9), as well as of tissue-nonspecific alkaline phosphatase (ALPL). Vasopressin-induced osteogenic signaling and calcification require the serum- and glucocorticoid-inducible kinase 1 (SGK1). Known effects of SGK1 include upregulation of Na+/H+ exchanger 1 (NHE1). NHE1 further participates in the regulation of reactive oxygen species (ROS). NHE1 has been shown to participate in the orchestration of bone mineralization. The present study, thus, explored whether vasopressin modifies NHE1 expression and ROS generation, as well as whether pharmacological inhibition of NHE1 disrupts vasopressin-induced osteogenic signaling and calcification in VSMCs. Methods: Human aortic smooth muscle cells (HAoSMCs) were treated with vasopressin in the absence or presence of SGK1 silencing, SGK1 inhibitor GSK-650394, and NHE1 blocker cariporide. Transcript levels were determined by using quantitative real-time polymerase chain reaction, protein abundance by Western blotting, ROS generation with 2′,7′-dichlorofluorescein diacetate fluorescence, and ALP activity and calcium content by using colorimetric assays. Results: Vasopressin significantly enhanced the NHE1 transcript and protein levels in HAoSMCs, effects significantly blunted by SGK1 inhibition with GSK-650394 or SGK1 silencing. Vasopressin increased ROS accumulation, an effect significantly blocked by the NHE1 inhibitor cariporide. Vasopressin further significantly increased osteogenic markers CBFA1, MSX2, SOX9, and ALPL transcript levels, as well as ALP activity and calcium content in HAoSMCs, all effects significantly blunted by SGK1 silencing or in the presence of GSK-650394 or cariporide. Conclusion: Vasopressin stimulates NHE1 expression and ROS generation, an effect dependent on SGK1 and required for vasopressin-induced stimulation of osteogenic signaling and calcification of VSMCs.

Medial vascular calcification is considered an important cause of cardiovascular diseases [1-7]. In chronic kidney disease, compromised renal phosphate excretion leads to hyperphosphatemia, which in turn stimulates vascular calcification at least partly by inducing osteo-/chondrogenic reprogramming of vascular smooth muscle cells (VSMCs) [1-3, 6]. Osteo-/chondrogenic reprogramming involves upregulation of the osteogenic and chondrogenic transcription factors such as core-binding factor α-1 (CBFA1), msh homeobox 2 (MSX2), and SRY-Box 9 (SOX9) with subsequent stimulation of tissue-nonspecific alkaline phosphatase (ALPL) expression and activity [8-11]. The enzyme contributes to calcium-phosphate precipitation and tissue calcification by degradation of the calcification inhibitor pyrophosphate [8-11].

VSMCs are a target for vasopressin [12]. Vasopressin is a powerful stimulator of vascular calcification promoting osteo-/chondrogenic reprogramming of VSMCs [13, 14], which is upregulated by insufficient hydration [15, 16]. Mechanisms involved in the procalcific effects of vasopressin include activation of the serum- and glucocorticoid-inducible kinase 1 (SGK1) [17] with subsequent upregulation of store-operated Ca2+ entry [14, 15, 18-20]. SGK1-induced downstream targets include the Na+/H+ exchanger NHE1 [21-28], which has been shown to participate in the regulation of calcification in osteoblasts [29, 30]. Aldosterone is an activator of SGK1 and upregulates Na+/H+ exchange in VSMCs [31]. In hypertensive rats, NHE1 is upregulated in VSMCs [32]. Mice with reduced NHE1 expression are protected from abdominal aortic aneurysm induced by angiotensin-II [33]. Furthermore, NHE1 may play a role in atherogenesis [34]. At least in endothelial cells, vasopressin stimulates Na+/H+ exchange activity [35]. NHE1-dependent cellular functions also include induction of oxidative stress [36, 37]. Thus, the present study explored whether in human aortic smooth muscle cells (HAoSMCs), NHE1 and ROS generation are upregulated by vasopressin and required for stimulation of osteogenic signaling.

Cell Culture

HAoSMCs (Thermo Fisher Scientific) were cultured in Medium 231 (Thermo Fisher Scientific), supplemented with 10% fetal bovine serum (Gibco, Grand Island, NE, USA) and 1% penicillin/streptomycin in a humidified atmosphere at 37°C and 5% CO2. HAoSMCs were grown to confluency and used in all experiments from passages 4 to 10. Where indicated, the cells were exposed to 100 nM vasopressin (Sigma, Steinheim, Germany) [13], to SGK1 inhibitor GSK-650394 (1 μM; Sigma) [38], and/or to NHE1 inhibitor cariporide (10 μM; Sigma-Aldrich, St. Louis, MO, USA) [24]. Vasopressin was dissolved in Medium 231, while GSK-650394 and cariporide were dissolved in DMSO. Medium pH was 7.289 ± 0.024 (control), 7.294 ± 0.022 (cariporide), 7.292 ± 0.019 (vasopressin), and 7.295 ± 0.018 (vasopressin and cariporide) after a 24-h incubation. Medium bicarbonate concentration was 18.84 ± 0.96 mM (control), 19.04 ± 0.89 mM (cariporide), 18.97 ± 0.78 mM (vasopressin) and 19.09 ± 0.74 mM (vasopressin and cariporide) after a 24-h incubation. The values were not significantly different between the absence or presence of vasopressin and/or cariporide.

Silencing of SGK1

HAoSMCs were transfected with 10 nM SGK1 siRNA (s740; Thermo Fisher Scientific, Karlsruhe, Germany) [17] or with 10 nM negative control siRNA (4390843; Thermo Fisher Scientific) using Lipofectamine 2000 transfection agent (Invitrogen, Thermo Fisher Scientific) according to the manufacturer’s instructions. HAoSMCs were collected after a 48-h transfection, and silencing efficiency was evaluated by quantitative real-time polymerase chain reaction (qRT-PCR).

Quantitative Real-Time Polymerase Chain Reaction

To determine transcript levels, total RNA was extracted by using TriFast reagent (Peqlab, Erlangen, Germany) according to the manufacturer’s protocol. After DNAse digestion, reverse transcription of total RNA was carried out using Oligo(dT)15 primer (Promega, Hilden, Germany) and GoScriptTM Reverse Transcriptase (Promega). RT-PCR amplification of the respective genes was set up in a total volume of 15 μL using 100 ng of cDNA, 500 nM forward and reverse primers, and 2× GoTaq® qPCR Master Mix (Promega) according to the manufacturer’s instructions. Cycling conditions were as follows: initial denaturation at 95°C for 3 min, followed by 40 cycles of 95°C for 15 s, 58°C for 30 s, and 72°C for 30 s. RT-PCR amplifications were performed on a CFX96 Real-Time System (Bio-Rad, Munich, Germany). The primers used in this study for amplification are listed in Table 1.

Table 1.

List of the primer sequences

List of the primer sequences
List of the primer sequences

Specificity of PCR products was confirmed by the analysis of a melting curve. All PCRs were performed in duplicate and relative mRNA expression, which was calculated by the 2−ΔΔCt method using GAPDH, β-actin, or B2M as an internal reference normalized to the control group.

Western Blotting

Protein abundance of NHE1, phosphorylated and thus activated SGK1, and total SGK1 was determined by Western blotting. The harvested cells were centrifuged for 5 min at 1,000 rpm and 4°C. The pellet was washed twice with ice-cold phosphate-buffered saline (PBS) and suspended in 40 μL ice-cold RIPA lysis buffer (Cell Signaling Technology, Danvers, MA, USA) containing Halt Protease and Halt Phosphatase Inhibitor Cocktail (Thermo Fisher Scientific, Danvers, MA, USA). Protein concentration was quantified using the Bradford assay (Bio-Rad Laboratories). Equal amounts of proteins were separated by 10% sodium dodecyl sulfate-polyacrylamide gel in Glycine-Tris buffer, electro-transferred onto polyvinylidene difluoride membranes and blocked with 5% nonfat milk in TBST at room temperature. The membranes were incubated with primary anti-SGK1 (phospho S422) antibody (1:1,000; Abcam, Cambridge, UK), anti-SGK1 antibody (1:1,000; Cell Signaling Technology), anti-NHE1 antibody (1:1,000; Sigma-Aldrich) [39], and anti-GAPDH antibody (1:1,000; Cell Signaling Technology) at 4°C overnight. After washing (TBST), the blots were incubated with secondary anti-rabbit antibody conjugated with horseradish peroxidase (1:2,000; Cell Signaling Technology) for 2 h at room temperature. Protein bands were detected after additional washes (TBST) with an ECL detection reagent (Thermo Fisher Scientific, Waltham, MA, USA). For densitometry image analysis, Western blots were scanned and analyzed by ImageJ software (NIH, Bethesda, MD, USA), and the results are shown as the ratio of total protein to GAPDH. To assign the right protein size, a protein marker (Thermo Fisher Scientific) was used.

Detection of ROS Generation

HAoSMCs were treated with 100 nM vasopressin in the absence and presence of NHE1 blocker cariporide. Harvested HAoSMCs were washed and incubated at a final concentration of 10 μM 2′,7′-dichlorofluorescein diacetate (Sigma-Aldrich) in the dark for 30 min at 37°C. Then, cells were washed three times with PBS and subsequently resuspended in 500 μL PBS. ROS production was immediately measured by flow cytometry (BD Biosciences, Heidelberg, Germany). Fluorescence intensity of 2′,7′-dichlorofluorescein diacetate was detected and measured at an excitation wavelength of 488 nm and an emission wavelength of 530 nm. The geometric mean was calculated using FlowJo_V10 software (Version V10; TreeStar, California, USA).

Alkaline Phosphatase Activity Assay

HAoSMCs were exposed to 100 nM vasopressin for 7 days, in the absence or presence of SGK1 silencing, SGK1 inhibitor GSK-650394, or NHE1 inhibitor cariporide at the concentrations indicated in the figure legends. Fresh media with agents were added every 2–3 days. Alkaline phosphatase (ALP) activity in VSMCs was determined using the ALP colorimetric assay kit (Abcam) according to the manufacturer’s protocol. The results are shown normalized to total protein concentration as assessed by the Bradford assay (Bio-Rad Laboratories).

Calcium Content Analysis

HAoSMCs were treated with 100 nM vasopressin for 14 days. Then, HAoSMCs were decalcified in 0.6 M HCl at 4°C for 24 h. Calcium content was evaluated colorimetrically by the QuantiChrom Calcium assay kit (BioAssay Systems, Hayward, CA, USA) according to the manufacturer’s protocol. HAoSMCs were lysed with 0.1 M NaOH/0.1% sodium dodecyl sulfate. Calcium content was normalized to total protein concentration as assessed by the Bradford assay (Bio-Rad Laboratories). To visualize calcification, HAoSMCs were exposed to 100 nM vasopressin and 1 mM CaCl2 (Sigma-Aldrich) for 14 days for alizarin red staining. Fresh media with agents were added every 2–3 days. VSMCs were fixed with 4% paraformaldehyde and stained with 1% Alizarin Red (pH 4.0). The calcified areas are shown as red staining.

Statistical Analysis

Data are provided as means ± SD, and n represents the number of experiments. Statistical analysis was performed using statistic package for social science (SPSS; Version 22.0) and GraphPad Prism (Version 8.0.2). All data were tested for significance using unpaired t test (Student’s t test) or two-way ANOVA followed by post hoc Tukey’s test for multiple comparisons. A p < 0.05 was considered statistically significant.

To investigate the effect of vasopressin on the expression of SGK1, HAoSMCs were exposed to vasopressin (100 nM). DMSO was used as the vehicle. A 24-h incubation of HAoSMCs with vasopressin elevated the transcript levels of SGK1 (Fig. 1a). Moreover, the protein expression of phospho-SGK1 (S422) (Fig. 1b, c; online suppl. Fig. 1; for all online suppl. material, see www.karger.com/doi/10.1159/000524050) normalized to GAPDH was significantly upregulated. However, the protein expression of phospho-SGK1 (S422) (Fig. 1b, d; online suppl. Fig. 1) normalized to total-SGK1 was unchanged.

Fig. 1.

Effects of vasopressin on SGK1 expression in HAoSMCs. a Scatter dot plots with arithmetic means ± SD (n= 6) of SGK1 transcript levels in HAoSMCs following a 24-h culture without (Ctrl) and with (AVP) 100 nM vasopressin. b Representative original blots showing the protein levels of phospho-SGK1 (S422) and total-SGK1 following a 24-h treatment without (Ctrl) and with (AVP) 100 nM vasopressin. c Quantification showing the protein levels of phospho-SGK1 (S422) normalized to GAPDH following a 24-h treatment without (Ctrl) and with (AVP) 100 nM vasopressin. d Quantification showing the protein levels of phospho-SGK1 (S422) normalized to total-SGK1 following a 24-h treatment without (Ctrl) and with (AVP) 100 nM vasopressin. *p< 0.05 indicates a statistically significant difference to the absence of vasopressin (unpaired t test).

Fig. 1.

Effects of vasopressin on SGK1 expression in HAoSMCs. a Scatter dot plots with arithmetic means ± SD (n= 6) of SGK1 transcript levels in HAoSMCs following a 24-h culture without (Ctrl) and with (AVP) 100 nM vasopressin. b Representative original blots showing the protein levels of phospho-SGK1 (S422) and total-SGK1 following a 24-h treatment without (Ctrl) and with (AVP) 100 nM vasopressin. c Quantification showing the protein levels of phospho-SGK1 (S422) normalized to GAPDH following a 24-h treatment without (Ctrl) and with (AVP) 100 nM vasopressin. d Quantification showing the protein levels of phospho-SGK1 (S422) normalized to total-SGK1 following a 24-h treatment without (Ctrl) and with (AVP) 100 nM vasopressin. *p< 0.05 indicates a statistically significant difference to the absence of vasopressin (unpaired t test).

Close modal

In order to determine whether vasopressin modifies the expression of Na+/H+ exchanger 1 (NHE1) in VSMCs, HAoSMCs were incubated without or with vasopressin (100 nM), and NHE1 transcript levels and NHE1 protein abundance were subsequently determined by using qRT-PCR and Western blotting. As shown in Figure 2a and b and online supplementary Figure 2, vasopressin treatment significantly increased the NHE1 transcript levels and NHE1 protein abundance in HAoSMCs. In order to explore whether the effect of vasopressin on NHE1 expression involved SGK1, experiments were performed in the absence or presence of the SGK1 inhibitor GSK-650394 (1 μM). As illustrated in Figure 2c and d and online supplementary Figure 3, GSK-650394 significantly blunted the effect of vasopressin treatment on NHE1 transcript levels and NHE1 protein abundance.

Fig. 2.

Effect of vasopressin on NHE1 expression in the absence or presence of SGK1 inhibitor GSK-650394. a Scatter dot plots with arithmetic means ± SD (n= 6) of NHE1 transcript levels following a 24-h culture without (Ctrl) and with (AVP) 100 nM vasopressin. b Original Western blots and quantification showing NHE1 protein abundance following a 24-h culture without (Ctrl) and with (AVP) 100 nM vasopressin. c Scatter dot plots with arithmetic means ± SD (n= 6) of NHE1 transcript levels in HAoSMCs following a 24-h culture without (Ctrl) and with (AVP) 100 nM vasopressin in the absence (Veh) or presence of 1 μM GSK-650394 (GSK). d Original Western blots and quantification showing NHE1 protein abundance following a 24-h culture without (Ctrl) and with (AVP) 100 nM vasopressin in the absence (Veh) or presence of 1 μM GSK-650394 (GSK). *p< 0.05 and **p< 0.01 indicate statistically significant differences to the absence of vasopressin (unpaired t test or ANOVA), and #p< 0.05 indicates a statistically significant difference to vasopressin treatment alone (ANOVA).

Fig. 2.

Effect of vasopressin on NHE1 expression in the absence or presence of SGK1 inhibitor GSK-650394. a Scatter dot plots with arithmetic means ± SD (n= 6) of NHE1 transcript levels following a 24-h culture without (Ctrl) and with (AVP) 100 nM vasopressin. b Original Western blots and quantification showing NHE1 protein abundance following a 24-h culture without (Ctrl) and with (AVP) 100 nM vasopressin. c Scatter dot plots with arithmetic means ± SD (n= 6) of NHE1 transcript levels in HAoSMCs following a 24-h culture without (Ctrl) and with (AVP) 100 nM vasopressin in the absence (Veh) or presence of 1 μM GSK-650394 (GSK). d Original Western blots and quantification showing NHE1 protein abundance following a 24-h culture without (Ctrl) and with (AVP) 100 nM vasopressin in the absence (Veh) or presence of 1 μM GSK-650394 (GSK). *p< 0.05 and **p< 0.01 indicate statistically significant differences to the absence of vasopressin (unpaired t test or ANOVA), and #p< 0.05 indicates a statistically significant difference to vasopressin treatment alone (ANOVA).

Close modal

To explore a role of NHE1 in oxidative stress, HAoSMCs were treated without or with vasopressin (100 nM) in the absence and presence of the NHE1 inhibitor cariporide (10 μM). As shown in Figure 3, vasopressin stimulated ROS accumulation in HAoSMCs, an effect significantly blunted by inhibition of NHE1 with cariporide.

Fig. 3.

Sensitivity of vasopressin-induced ROS generation of HAoSMCs in response to NHE1 inhibitor cariporide. a Representative overlay histograms from the flow cytometry analysis using 2′,7′-dichlorofluorescein diacetate (DCFDA) illustrating ROS generation following a 24-h stimulation without (Ctrl) and with (AVP) 100 nM vasopressin in the absence (Veh) or presence of 10 μM NHE1 inhibitor cariporide (Cari). b Scatter dot plots with arithmetic means ± SD (n= 6) showing the quantification of ROS production. ***p< 0.001 indicates a statistically significant difference to the absence of vasopressin, and ##p< 0.01 indicates a statistically significant difference to vasopressin treatment alone (ANOVA).

Fig. 3.

Sensitivity of vasopressin-induced ROS generation of HAoSMCs in response to NHE1 inhibitor cariporide. a Representative overlay histograms from the flow cytometry analysis using 2′,7′-dichlorofluorescein diacetate (DCFDA) illustrating ROS generation following a 24-h stimulation without (Ctrl) and with (AVP) 100 nM vasopressin in the absence (Veh) or presence of 10 μM NHE1 inhibitor cariporide (Cari). b Scatter dot plots with arithmetic means ± SD (n= 6) showing the quantification of ROS production. ***p< 0.001 indicates a statistically significant difference to the absence of vasopressin, and ##p< 0.01 indicates a statistically significant difference to vasopressin treatment alone (ANOVA).

Close modal

In order to explore whether the SGK1-sensitive NHE1 regulation by vasopressin is relevant for the effects of vasopressin on osteogenic signaling, the effects of vasopressin (100 nM) on the transcript levels of osteogenic transcription factors CBFA1, MSX2, and SOX9 as well as the osteogenic enzyme ALPL were quantified after treatment of HAoSMCs for 24 h with vasopressin in the absence or presence of the SGK1 inhibitor GSK-650394 (1 μM) or NHE1 inhibitor cariporide (10 μM). As illustrated in Figure 4, each GSK-650394 and cariporide significantly blunted the effects of vasopressin treatment on CBFA1, MSX2, SOX9, and ALPL transcript levels.

Fig. 4.

Effect of vasopressin on CBFA1, MSX2, SOX9, and ALPL transcription in the absence or presence of SGK1 inhibitor GSK-650394 or NHE1 inhibitor cariporide. Scatter dot plots with arithmetic means ± SD (n= 6) of CBFA1(a), MSX2(b), SOX9(c), and ALPL(d) transcript levels in HAoSMCs following a 24-h culture without (Ctrl) and with (AVP) 100 nM vasopressin in the absence (Veh) or presence of 1 μM GSK-650394 (GSK) or 10 μM cariporide (Cari). **p< 0.01 and ***p< 0.001 indicate statistically significant differences to the absence of vasopressin, and #p< 0.05, ##p< 0.01, and ###p< 0.001 indicates statistically significant differences to vasopressin treatment alone (ANOVA).

Fig. 4.

Effect of vasopressin on CBFA1, MSX2, SOX9, and ALPL transcription in the absence or presence of SGK1 inhibitor GSK-650394 or NHE1 inhibitor cariporide. Scatter dot plots with arithmetic means ± SD (n= 6) of CBFA1(a), MSX2(b), SOX9(c), and ALPL(d) transcript levels in HAoSMCs following a 24-h culture without (Ctrl) and with (AVP) 100 nM vasopressin in the absence (Veh) or presence of 1 μM GSK-650394 (GSK) or 10 μM cariporide (Cari). **p< 0.01 and ***p< 0.001 indicate statistically significant differences to the absence of vasopressin, and #p< 0.05, ##p< 0.01, and ###p< 0.001 indicates statistically significant differences to vasopressin treatment alone (ANOVA).

Close modal

As illustrated in Figure 5, vasopressin further significantly increased ALP activity in HAoSMCs, an effect significantly blunted in the presence of GSK-650394 (1 μM) or cariporide (10 μM). As shown in Figure 6, vasopressin significantly enhanced the formation of calcium deposits, an effect again significantly blunted in the presence of GSK-650394 (1 μM) or cariporide (10 μM).

Fig. 5.

Effect of vasopressin on ALP activity in the absence or presence of SGK1 inhibitor GSK-650394 or NHE1 inhibitor cariporide. Scatter dot plots with arithmetic means ± SD (n= 6) of ALP activity in HAoSMCs after a 7-day culture without (Ctrl) and with (AVP) 100 nM vasopressin in the absence (Veh) or presence of 1 μM GSK-650394 (GSK) (a) or 10 μM cariporide (Cari) (b). **p< 0.01 and ***p< 0.001 indicate statistically significant differences to the absence of vasopressin, and ##p< 0.01 and ###p< 0.001 indicate statistically significant differences to vasopressin treatment alone (ANOVA).

Fig. 5.

Effect of vasopressin on ALP activity in the absence or presence of SGK1 inhibitor GSK-650394 or NHE1 inhibitor cariporide. Scatter dot plots with arithmetic means ± SD (n= 6) of ALP activity in HAoSMCs after a 7-day culture without (Ctrl) and with (AVP) 100 nM vasopressin in the absence (Veh) or presence of 1 μM GSK-650394 (GSK) (a) or 10 μM cariporide (Cari) (b). **p< 0.01 and ***p< 0.001 indicate statistically significant differences to the absence of vasopressin, and ##p< 0.01 and ###p< 0.001 indicate statistically significant differences to vasopressin treatment alone (ANOVA).

Close modal
Fig. 6.

Effect of vasopressin on calcium deposits in the absence or presence of SGK1 inhibitor GSK-650394 or NHE1 inhibitor cariporide. Original photographs showing calcium deposits following a 14-day culture of HAoSMCs without (Ctrl) and with (AVP) 100 nM vasopressin and 1 mM CaCl2 in the absence (Veh) or presence of 1 μM GSK-650394 (GSK) (a) or 10 μM cariporide (Cari) (b). Scatter dot plots with arithmetic means ± SD (n= 6) of calcium content in HAoSMCs after a 14-day culture without (Ctrl) and with (AVP) 100 nM vasopressin in the absence (Veh) or presence of 1 μM GSK-650394 (GSK) (c) or 10 μM cariporide (Cari) (d). **p< 0.01 indicates a statistically significant difference to the absence of vasopressin, and #p< 0.05 indicates a statistically significant difference to vasopressin treatment alone (ANOVA).

Fig. 6.

Effect of vasopressin on calcium deposits in the absence or presence of SGK1 inhibitor GSK-650394 or NHE1 inhibitor cariporide. Original photographs showing calcium deposits following a 14-day culture of HAoSMCs without (Ctrl) and with (AVP) 100 nM vasopressin and 1 mM CaCl2 in the absence (Veh) or presence of 1 μM GSK-650394 (GSK) (a) or 10 μM cariporide (Cari) (b). Scatter dot plots with arithmetic means ± SD (n= 6) of calcium content in HAoSMCs after a 14-day culture without (Ctrl) and with (AVP) 100 nM vasopressin in the absence (Veh) or presence of 1 μM GSK-650394 (GSK) (c) or 10 μM cariporide (Cari) (d). **p< 0.01 indicates a statistically significant difference to the absence of vasopressin, and #p< 0.05 indicates a statistically significant difference to vasopressin treatment alone (ANOVA).

Close modal

Similar to pharmacological inhibition, silencing of the SGK1 gene in HAoSMCs disrupted vasopressin-induced NHE1 expression and osteo-/chondrogenic signaling. SGK1 mRNA level was significantly decreased after transfection with SGK1 siRNA (10 nM), as compared to negative control siRNA-transfected HAoSMCs (Fig. 7a). Vasopressin (100 nM) treatment upregulated SGK1 transcription in negative control siRNA-transfected HAoSMCs (Fig. 7a). The vasopressin-induced NHE1 transcript levels (Fig. 7b) and NHE1 protein abundance (Fig. 7c, d; online suppl. Fig. 4) were significantly blunted in SGK1 siRNA-transfected HAoSMCs. Furthermore, the vasopressin-induced transcript levels of CBFA1, MSX2, SOX9, and ALPL were significantly blunted in SGK1 siRNA-transfected HAoSMCs (Fig. 8a–d). Also, the increased ALP activity and the enhanced calcium deposition by vasopressin were reversed by SGK1 knockdown (Fig. 8e, f).

Fig. 7.

Silencing of SGK1 blunted vasopressin-induced NHE1 expression in HAoSMCs. Scatter dot plots with arithmetic means ± SD (n= 6) of SGK1(a) and NHE1(b) transcript levels in HAoSMCs following silencing for 48 h with 10 nM negative control siRNA (Neg.si) or 10 nM SGK1 siRNA (SGK1si) and without (Ctrl) or with (AVP) additional 100 nM vasopressin treatment for 24 h. Original Western blots (c) and quantification (d) showing NHE1 protein abundance in HAoSMCs following silencing for 48 h with 10 nM negative control siRNA (Neg.si) or 10 nM SGK1 siRNA (SGK1si) and without (Ctrl) or with (AVP) additional 100 nM vasopressin treatment for 24 h. *p< 0.05, **p< 0.01, and ***p< 0.001 indicate statistically significant differences to Neg.si-silenced HAoSMCs, and #p< 0.05 indicates a statistically significant difference to Neg.si-silenced and vasopressin-treated HAoSMCs (ANOVA).

Fig. 7.

Silencing of SGK1 blunted vasopressin-induced NHE1 expression in HAoSMCs. Scatter dot plots with arithmetic means ± SD (n= 6) of SGK1(a) and NHE1(b) transcript levels in HAoSMCs following silencing for 48 h with 10 nM negative control siRNA (Neg.si) or 10 nM SGK1 siRNA (SGK1si) and without (Ctrl) or with (AVP) additional 100 nM vasopressin treatment for 24 h. Original Western blots (c) and quantification (d) showing NHE1 protein abundance in HAoSMCs following silencing for 48 h with 10 nM negative control siRNA (Neg.si) or 10 nM SGK1 siRNA (SGK1si) and without (Ctrl) or with (AVP) additional 100 nM vasopressin treatment for 24 h. *p< 0.05, **p< 0.01, and ***p< 0.001 indicate statistically significant differences to Neg.si-silenced HAoSMCs, and #p< 0.05 indicates a statistically significant difference to Neg.si-silenced and vasopressin-treated HAoSMCs (ANOVA).

Close modal
Fig. 8.

Silencing of SGK1 blunted vasopressin-induced osteogenic signaling and calcification of HAoSMCs. Scatter dot plots with arithmetic means ± SD (n= 6) of CBFA1(a), MSX2(b), SOX9(c), and ALPL(d) transcript levels in HAoSMCs following silencing for 48 h with 10 nM negative control siRNA (Neg.si) or 10 nM SGK1 siRNA (SGK1si) and without (Ctrl) or with (AVP) additional 100 nM vasopressin treatment for 24 h. e Scatter dot plots with arithmetic means ± SD (n= 6) of ALP activity in HAoSMCs after 7 days of silencing with 10 nM negative control siRNA (Neg.si) or 10 nM SGK1 siRNA (SGK1si) and without (Ctrl) or with (AVP) additional 100 nM vasopressin treatment. f Scatter dot plots with arithmetic means ± SD (n= 6) of calcium content in HAoSMCs following 14 days of silencing with 10 nM negative control siRNA (Neg.si) or 10 nM SGK1 siRNA (SGK1si) and without (Ctrl) or with (AVP) additional 100 nM vasopressin treatment. **p< 0.01 and ***p< 0.001 indicate statistically significant differences to Neg.si-silenced HAoSMCs, and #p< 0.05 and ##p< 0.01 indicate statistically significant differences to Neg.si-silenced and vasopressin-treated HAoSMCs (ANOVA).

Fig. 8.

Silencing of SGK1 blunted vasopressin-induced osteogenic signaling and calcification of HAoSMCs. Scatter dot plots with arithmetic means ± SD (n= 6) of CBFA1(a), MSX2(b), SOX9(c), and ALPL(d) transcript levels in HAoSMCs following silencing for 48 h with 10 nM negative control siRNA (Neg.si) or 10 nM SGK1 siRNA (SGK1si) and without (Ctrl) or with (AVP) additional 100 nM vasopressin treatment for 24 h. e Scatter dot plots with arithmetic means ± SD (n= 6) of ALP activity in HAoSMCs after 7 days of silencing with 10 nM negative control siRNA (Neg.si) or 10 nM SGK1 siRNA (SGK1si) and without (Ctrl) or with (AVP) additional 100 nM vasopressin treatment. f Scatter dot plots with arithmetic means ± SD (n= 6) of calcium content in HAoSMCs following 14 days of silencing with 10 nM negative control siRNA (Neg.si) or 10 nM SGK1 siRNA (SGK1si) and without (Ctrl) or with (AVP) additional 100 nM vasopressin treatment. **p< 0.01 and ***p< 0.001 indicate statistically significant differences to Neg.si-silenced HAoSMCs, and #p< 0.05 and ##p< 0.01 indicate statistically significant differences to Neg.si-silenced and vasopressin-treated HAoSMCs (ANOVA).

Close modal

The transcript levels of SGK1 and NHE1 as well as CBFA1, MSX2, SOX9, and ALPL were compared not only to GAPDH but also to β-actin or B2M as an internal reference. Importantly, no pseudogenes were found in B2M [40]. As shown in online supplementary Figure 5, vasopressin significantly upregulated the transcript levels of SGK1 and NHE1 as well as CBFA1, MSX2, SOX9, and ALPL normalized to β-actin or B2M.

Similar to what has been shown in earlier studies [13, 14], the present observations demonstrate the ability of vasopressin to upregulate the expression of osteogenic transcription factors CBFA1, MSX2, and SOX9 as well as the osteogenic enzyme ALPL in VSMCs. As a result, vasopressin promotes calcium deposition in VSMCs [13, 14]. During the formation of vascular calcification, many cells, including VSMCs, transform into cells with some osteoblast-like or chondrocytes-like properties [41]. This transformation contributes to an active calcification process [14, 20]. ALP activity is an important marker for early osteoblast differentiation, which cleaves the calcification inhibitor pyrophosphate and subsequently alters the local environment to allow for calcification [41, 42]. Thus, upregulation of ALP is considered to precede and allow for vascular calcification [43, 44].

The present study further reveals the involvement of the Na+/H+ exchanger NHE1 in the upregulation of osteogenic signaling and calcium deposition in VSMCs by vasopressin. Vasopressin-induced osteogenic signaling and calcium deposition are virtually abolished in the presence of the NHE1 inhibitor cariporide. Mechanisms linking NHE1 activity to osteogenic signaling may include luminal pH in intracellular vesicles. NHE1 is inserted into phagosomal membranes and may contribute to luminal acidification of intracellular vesicles [45]. In VSMCs, luminal acidification may be involved in phosphate-induced calcification [46] and calcium-phosphate-induced cell death [47]. NHE1 may further be required for the generation of oxidative stress [48-52], which has in turn been shown to participate in the orchestration of osteogenic signaling [53-57]. In addition, NHE1 activity may be linked to production of inflammatory cytokines [58], which may contribute to osteogenic mechanisms in VSMCs [59]. However, the exact mechanisms mediating the procalcific effects of NHE1 in VSMCs require further study.

As NHE1 is tightly regulated by cytosolic pH and turned off by alkaline cytosolic pH [60], enhanced NHE1 expression counteracts cytosolic acidification but does not affect alkaline cytosolic pH. In the absence of NHE1, H+ generation or entry leads to cytosolic acidification. The effects of vasopressin on NHE1 expression were significantly blunted in the presence of SGK1 inhibitor GSK-650394 or SGK1 silencing. According to the present observations, vasopressin upregulates SGK1 transcription and phospho-SGK1 (S422) protein abundance in HAoSMCs. Furthermore, pharmacological inhibition or genetic silencing of SGK1 suppressed vasopressin-induced osteogenic signaling and calcium deposition.

The present paper did not elucidate the signaling mediating the upregulation of SGK1 by vasopressin. As vasopressin stimulates Ca2+ entry [14, 61], which in turn upregulates SGK1 [62-64], the signaling could involve increase of cytosolic Ca2+. SGK1 upregulates the store-operated Ca2+ channel Orai1 [18]; at least in theory, Ca2+ entry through Orai1 could further enhance cytosolic Ca2+, thus augmenting the effect of vasopressin on SGK1 and NHE1.

Ample experimental evidence points to a critical role of both, vasopressin and SGK1, in the pathophysiology of dehydration [15]. According to earlier studies [17], SGK1 is decisive for stimulation of osteogenic signaling by phosphate. SGK1 has further been shown to participate in the effects of vasopressin on VSMCs [14]. SGK1 has been shown to mediate upregulation of NHE1 expression [22-26, 28]. Thus, SGK1-dependent upregulation of NHE1 may contribute to calcification of VSMCs. SGK1-dependent upregulation of NHE1 may further contribute to the regulation of diverse functions in several cell types, such as aldosterone-induced renal salt retention, hypertension and cardiac remodeling [22, 23, 27, 65, 66], cell proliferation and apoptosis [24, 67], activation of dendritic cells with bacterial lipopolysaccharides or dexamethasone [21, 68], endothelial cell swelling and brain edema [28], as well as endothelial leukocyte recruitment [26]. SGK1-sensitive upregulation of NHE1 with subsequent cell swelling and cytosolic alkalinization may further contribute to stimulation of cell proliferation and counteraction of suicidal cell death [24, 67, 69-71].

The present observations may be important for vascular calcification in chronic kidney disease. A role for vasopressin in vascular calcification has already been suggested in patients with renal insufficiency. The concentrations of copeptin, considered a marker for vasopressin release [72], are related to coronary artery calcification score in patients with chronic kidney disease [73]. In type 1 diabetes, serum copeptin concentrations are associated with coronary atherosclerosis and diabetic kidney disease [74]. Furthermore, in the general population, higher copeptin levels are linked to vascular stiffness [75]. Copeptin also has been connected to mortality in elderly people [76]. Interfering with the vasopressin-calcification pathway may ameliorate vascular calcification and reduce cardiovascular risk.

Vasopressin treatment increases NHE1 expression and ROS generation, an effect requiring SGK1 and participating in the upregulation of osteogenic signaling and calcification of VSMCs.

The authors gratefully acknowledge the meticulous preparation of the manuscript by Lejla Subasic.

An ethics statement was not required for this study type, and no human or animal subjects or materials were used.

The authors state that they have no conflicts of interest.

Xuexue Zhu, Kuo Zhou, and Jibin Liu are supported by the Chinese Scholarship Council. The sponsor(s) had no role in study design, the collection, analysis, and interpretation of data; in the writing of the report; and in the decision to submit the article for publication.

Xuexue Zhu, Ke Ma, Kuo Zhou, Xia Pan, and Jibin Liu performed experiments and analyzed data; Florian Lang, Jakob Voelkl, Ioana Alesutan, and Bernd Nürnberg designed research; and Florian Lang drafted and wrote the manuscript.

All data generated or analyzed during this study are included in this published article and its online supplementary material. Further enquiries can be directed to the corresponding author.

1.
Foley
RN
,
Parfrey
PS
,
Sarnak
MJ
.
Clinical epidemiology of cardiovascular disease in chronic renal disease
.
Am J Kidney Dis
.
1998 Nov
;
32
(
5 Suppl 3
):
S112
9
. .
2.
Blacher
J
,
Guerin
AP
,
Pannier
B
,
Marchais
SJ
,
London
GM
.
Arterial calcifications, arterial stiffness, and cardiovascular risk in end-stage renal disease
.
Hypertension
.
2001 Oct
;
38
(
4
):
938
42
. .
3.
London
GM
,
Guérin
AP
,
Marchais
SJ
,
Métivier
F
,
Pannier
B
,
Adda
H
.
Arterial media calcification in end-stage renal disease: impact on all-cause and cardiovascular mortality
.
Nephrol Dial Transplant
.
2003 Sep
;
18
(
9
):
1731
40
. .
4.
Towler
DA
,
Shao
JS
,
Cheng
SL
,
Pingsterhaus
JM
,
Loewy
AP
.
Osteogenic regulation of vascular calcification
.
Ann N Y Acad Sci
.
2006 Apr
;
1068
:
327
33
. .
5.
Fadini
GP
,
Pauletto
P
,
Avogaro
A
,
Rattazzi
M
.
The good and the bad in the link between insulin resistance and vascular calcification
.
Atherosclerosis
.
2007 Aug
;
193
(
2
):
241
4
. .
6.
Mizobuchi
M
,
Towler
D
,
Slatopolsky
E
.
Vascular calcification: the killer of patients with chronic kidney disease
.
J Am Soc Nephrol
.
2009 Jul
;
20
(
7
):
1453
64
. .
7.
Haarhaus
M
,
Brandenburg
V
,
Kalantar-Zadeh
K
,
Stenvinkel
P
,
Magnusson
P
.
Alkaline phosphatase: a novel treatment target for cardiovascular disease in CKD
.
Nat Rev Nephrol
.
2017 Jul
;
13
(
7
):
429
42
. .
8.
Steitz
SA
,
Speer
MY
,
Curinga
G
,
Yang
HY
,
Haynes
P
,
Aebersold
R
,
Smooth muscle cell phenotypic transition associated with calcification: upregulation of Cbfa1 and downregulation of smooth muscle lineage markers
.
Circ Res
.
2001 Dec 7
;
89
(
12
):
1147
54
.
9.
Lang
F
,
Ritz
E
,
Voelkl
J
,
Alesutan
I
.
Vascular calcification: is aldosterone a culprit?
Nephrol Dial Transplant
.
2013 May
;
28
(
5
):
1080
4
. .
10.
Lang
F
,
Ritz
E
,
Alesutan
I
,
Voelkl
J
.
Impact of aldosterone on osteoinductive signaling and vascular calcification
.
Nephron Physiol
.
2014
;
128
(
1–2
):
40
5
. .
11.
Kapustin
AN
,
Chatrou
ML
,
Drozdov
I
,
Zheng
Y
,
Davidson
SM
,
Soong
D
,
Vascular smooth muscle cell calcification is mediated by regulated exosome secretion
.
Circ Res
.
2015 Apr 10
;
116
(
8
):
1312
23
.
12.
Nemenoff
RA
.
Vasopressin signaling pathways in vascular smooth muscle
.
Front Biosci
.
1998 Feb 15
;
3
:
d194
207
. .
13.
Nishiwaki-Yasuda
K
,
Suzuki
A
,
Kakita
A
,
Sekiguchi
S
,
Asano
S
,
Nishii
K
,
Vasopressin stimulates Na-dependent phosphate transport and calcification in rat aortic smooth muscle cells
.
Endocr J
.
2007 Feb
;
54
(
1
):
103
12
.
14.
Zhu
X
,
Ma
K
,
Zhou
K
,
Liu
J
,
Nürnberg
B
,
Lang
F
.
Vasopressin-stimulated ORAI1 expression and store-operated Ca(2+) entry in aortic smooth muscle cells
.
J Mol Med
.
2021 Mar
;
99
(
3
):
373
82
.
15.
Lang
F
,
Guelinckx
I
,
Lemetais
G
,
Melander
O
.
Two liters a day keep the doctor away? Considerations on the pathophysiology of suboptimal fluid intake in the common population
.
Kidney Blood Press Res
.
2017
;
42
(
3
):
483
94
. .
16.
Perrier
ET
,
Armstrong
LE
,
Bottin
JH
,
Clark
WF
,
Dolci
A
,
Guelinckx
I
,
Hydration for health hypothesis: a narrative review of supporting evidence
.
Eur J Nutr
.
2021 Apr
;
60
(
3
):
1167
80
.
17.
Voelkl
J
,
Luong
TT
,
Tuffaha
R
,
Musculus
K
,
Auer
T
,
Lian
X
,
SGK1 induces vascular smooth muscle cell calcification through NF-kappaB signaling
.
J Clin Invest
.
2018 Jul 2
;
128
(
7
):
3024
40
.
18.
Lang
F
,
Shumilina
E
.
Regulation of ion channels by the serum- and glucocorticoid-inducible kinase SGK1
.
FASEB J
.
2013 Jan
;
27
(
1
):
3
12
. .
19.
Sahu
I
,
Pelzl
L
,
Sukkar
B
,
Fakhri
H
,
Al-Maghout
T
,
Cao
H
,
NFAT5-sensitive Orai1 expression and store-operated Ca(2+) entry in megakaryocytes
.
FASEB J
.
2017 Aug
;
31
(
8
):
3439
48
.
20.
Ma
K
,
Liu
P
,
Al-Maghout
T
,
Sukkar
B
,
Cao
H
,
Voelkl
J
,
Phosphate-induced ORAI1 expression and store-operated Ca(2+) entry in aortic smooth muscle cells
.
J Mol Med
.
2019 Oct
;
97
(
10
):
1465
75
.
21.
Rotte
A
,
Pasham
V
,
Eichenmüller
M
,
Yang
W
,
Bhandaru
M
,
Lang
F
.
Influence of dexamethasone on Na+/H+ exchanger activity in dendritic cells
.
Cell Physiol Biochem
.
2011
;
28
(
2
):
305
14
. .
22.
Voelkl
J
,
Lin
Y
,
Alesutan
I
,
Ahmed
MS
,
Pasham
V
,
Mia
S
,
SGK1 sensitivity of Na(+)/H(+) exchanger activity and cardiac remodeling following pressure overload
.
Basic Res Cardiol
.
2012 Mar
;
107
(
2
):
236
.
23.
Voelkl
J
,
Pasham
V
,
Ahmed
MS
,
Walker
B
,
Szteyn
K
,
Kuhl
D
,
Sgk1-dependent stimulation of cardiac Na+/H+ exchanger Nhe1 by dexamethasone
.
Cell Physiol Biochem
.
2013
;
32
(
1
):
25
38
.
24.
Chatterjee
S
,
Schmidt
S
,
Pouli
S
,
Honisch
S
,
Alkahtani
S
,
Stournaras
C
,
Membrane androgen receptor sensitive Na+/H+ exchanger activity in prostate cancer cells
.
FEBS Lett
.
2014 May 2
;
588
(
9
):
1571
9
.
25.
Lang
F
,
Stournaras
C
,
Alesutan
I
.
Regulation of transport across cell membranes by the serum- and glucocorticoid-inducible kinase SGK1
.
Mol Membr Biol
.
2014 Feb
;
31
(
1
):
29
36
. .
26.
Qadri
SM
,
Su
Y
,
Cayabyab
FS
,
Liu
L
.
Endothelial Na+/H+ exchanger NHE1 participates in redox-sensitive leukocyte recruitment triggered by methylglyoxal
.
Cardiovasc Diabetol
.
2014 Sep 30
;
13
:
134
. .
27.
Lou
Y
,
Zhang
F
,
Luo
Y
,
Wang
L
,
Huang
S
,
Jin
F
.
Serum and glucocorticoid regulated kinase 1 in sodium homeostasis
.
Int J Mol Sci
.
2016 Aug 10
;
17
(
8
):
1307
. .
28.
Klug
NR
,
Chechneva
OV
,
Hung
BY
,
O’Donnell
ME
.
High glucose-induced effects on Na(+)-K(+)-2Cl(-) cotransport and Na(+)/H(+) exchange of blood-brain barrier endothelial cells: involvement of SGK1, PKCbetaII, and SPAK/OSR1
.
Am J Physiol Cell Physiol
.
2021 Apr 1
;
320
(
4
):
C619
34
.
29.
Liu
L
,
Schlesinger
PH
,
Slack
NM
,
Friedman
PA
,
Blair
HC
.
High capacity Na+/H+ exchange activity in mineralizing osteoblasts
.
J Cell Physiol
.
2011 Jun
;
226
(
6
):
1702
12
. .
30.
Blair
HC
,
Larrouture
QC
,
Tourkova
IL
,
Liu
L
,
Bian
JH
,
Stolz
DB
,
Support of bone mineral deposition by regulation of pH
.
Am J Physiol Cell Physiol
.
2018 Oct 1
;
315
(
4
):
C587
97
.
31.
Ebata
S
,
Muto
S
,
Okada
K
,
Nemoto
J
,
Amemiya
M
,
Saito
T
,
Aldosterone activates Na+/H+ exchange in vascular smooth muscle cells by nongenomic and genomic mechanisms
.
Kidney Int
.
1999 Oct
;
56
(
4
):
1400
12
.
32.
LaPointe
MS
,
Ye
M
,
Bacallao
R
,
Batlle
D
.
NHE-1 protein in vascular smooth muscle and lymphocytes from the spontaneously hypertensive rat
.
Hypertension
.
1997 Oct
;
30
(
4
):
880
5
. .
33.
Liu
CL
,
Liu
X
,
Wang
Y
,
Deng
Z
,
Liu
T
,
Sukhova
GK
,
Reduced Nhe1 (Na(+)-H(+) exchanger-1) function protects ApoE-deficient mice from Ang II (angiotensin II)-induced abdominal aortic aneurysms
.
Hypertension
.
2020 Jul
;
76
(
1
):
87
100
.
34.
Sarigianni
M
,
Tsapas
A
,
Mikhailidis
DP
,
Kaloyianni
M
,
Koliakos
G
,
Fliegel
L
,
Na+ H+ exchanger-1: a link with atherogenesis?
Expert Opin Investig Drugs
.
2010 Dec
;
19
(
12
):
1545
56
.
35.
Lam
TI
,
Wise
PM
,
O’Donnell
ME
.
Cerebral microvascular endothelial cell Na/H exchange: evidence for the presence of NHE1 and NHE2 isoforms and regulation by arginine vasopressin
.
Am J Physiol Cell Physiol
.
2009 Aug
;
297
(
2
):
C278
89
. .
36.
Rothstein
EC
,
Byron
KL
,
Reed
RE
,
Fliegel
L
,
Lucchesi
PA
.
H(2)O(2)-induced Ca(2+) overload in NRVM involves ERK1/2 MAP kinases: role for an NHE-1-dependent pathway
.
Am J Physiol Heart Circ Physiol
.
2002 Aug
;
283
(
2
):
H598
605
. .
37.
Garciarena
CD
,
Caldiz
CI
,
Correa
MV
,
Schinella
GR
,
Mosca
SM
,
Chiappe de Cingolani
GE
,
Na+/H+ exchanger-1 inhibitors decrease myocardial superoxide production via direct mitochondrial action
.
J Appl Physiol
.
2008 Dec
;
105
(
6
):
1706
13
.
38.
Borst
O
,
Schmidt
EM
,
Münzer
P
,
Schönberger
T
,
Towhid
ST
,
Elvers
M
,
The serum- and glucocorticoid-inducible kinase 1 (SGK1) influences platelet calcium signaling and function by regulation of Orai1 expression in megakaryocytes
.
Blood
.
2012 Jan 5
;
119
(
1
):
251
61
.
39.
Zeng
N
,
Zhou
Y
,
Zhang
S
,
Singh
Y
,
Shi
B
,
Salker
MS
,
1α,25(OH) 2D3 sensitive cytosolic pH regulation and glycolytic flux in human endometrial ishikawa cells
.
Cell Physiol Biochem
.
2017
;
41
(
2
):
678
88
.
40.
Balaji
S
,
Vanniarajan
A
.
Implication of pseudo reference genes in normalization of data from reverse transcription-quantitative PCR
.
Gene
.
2020 Oct 5
;
757
:
144948
. .
41.
Jiang
W
,
Zhang
Z
,
Li
Y
,
Chen
C
,
Yang
H
,
Lin
Q
,
The cell origin and role of osteoclastogenesis and osteoblastogenesis in vascular calcification
.
Front Cardiovasc Med
.
2021
;
8
:
639740
.
42.
Stamnitz
S
,
Klimczak
A
.
Mesenchymal stem cells, bioactive factors, and scaffolds in bone repair: from research perspectives to clinical practice
.
Cells
.
2021 Jul 29
;
10
(
8
):
1925
. .
43.
Speer
MY
,
Yang
HY
,
Brabb
T
,
Leaf
E
,
Look
A
,
Lin
WL
,
Smooth muscle cells give rise to osteochondrogenic precursors and chondrocytes in calcifying arteries
.
Circ Res
.
2009 Mar 27
;
104
(
6
):
733
41
.
44.
Hortells
L
,
Sosa
C
,
Guillén
N
,
Lucea
S
,
Millán
Á
,
Sorribas
V
.
Identifying early pathogenic events during vascular calcification in uremic rats
.
Kidney Int
.
2017 Dec
;
92
(
6
):
1384
94
. .
45.
Hackam
DJ
,
Rotstein
OD
,
Zhang
WJ
,
Demaurex
N
,
Woodside
M
,
Tsai
O
,
Regulation of phagosomal acidification. Differential targeting of Na+/H+ exchangers, Na+/K+-ATPases, and vacuolar-type H+-atpases
.
J Biol Chem
.
1997 Nov 21
;
272
(
47
):
29810
20
.
46.
Alesutan
I
,
Musculus
K
,
Castor
T
,
Alzoubi
K
,
Voelkl
J
,
Lang
F
.
Inhibition of phosphate-induced vascular smooth muscle cell osteo-/chondrogenic signaling and calcification by bafilomycin A1 and methylamine
.
Kidney Blood Press Res
.
2015
;
40
(
5
):
490
9
. .
47.
Ewence
AE
,
Bootman
M
,
Roderick
HL
,
Skepper
JN
,
McCarthy
G
,
Epple
M
,
Calcium phosphate crystals induce cell death in human vascular smooth muscle cells: a potential mechanism in atherosclerotic plaque destabilization
.
Circ Res
.
2008 Aug 29
;
103
(
5
):
e28
34
.
48.
Singh
Y
,
Zhou
Y
,
Zhang
S
,
Abdelazeem
KNM
,
Elvira
B
,
Salker
MS
,
Enhanced reactive oxygen species production, acidic cytosolic pH and upregulated Na+/H+ exchanger (NHE) in dicer deficient CD4+ T cells
.
Cell Physiol Biochem
.
2017
;
42
(
4
):
1377
89
.
49.
Zhou
Y
,
Shi
X
,
Chen
H
,
Zhang
S
,
Salker
MS
,
Mack
AF
,
DJ-1/Park7 sensitive Na(+)/H(+) exchanger 1 (NHE1) in CD4(+) T cells
.
J Cell Physiol
.
2017 Nov
;
232
(
11
):
3050
9
.
50.
Yeves
AM
,
Burgos
JI
,
Medina
AJ
,
Villa-Abrille
MC
,
Ennis
IL
.
Cardioprotective role of IGF-1 in the hypertrophied myocardium of the spontaneously hypertensive rats: a key effect on NHE-1 activity
.
Acta Physiol
.
2018 Oct
;
224
(
2
):
e13092
. .
51.
Medina
AJ
,
Pinilla
OA
,
Portiansky
EL
,
Caldiz
CI
,
Ennis
IL
.
Silencing of the Na(+)/H(+) exchanger 1(NHE-1) prevents cardiac structural and functional remodeling induced by angiotensin II
.
Exp Mol Pathol
.
2019 Apr
;
107
:
1
9
.
52.
Escudero
DS
,
Brea
MS
,
Caldiz
CI
,
Amarillo
ME
,
Aranda
JO
,
Portiansky
EL
,
PDE5 inhibition improves cardiac morphology and function in SHR by reducing NHE1 activity: repurposing Sildenafil for the treatment of hypertensive cardiac hypertrophy
.
Eur J Pharmacol
.
2021 Jan 15
;
891
:
173724
.
53.
Duer
M
,
Cobb
AM
,
Shanahan
CM
.
DNA damage response: a molecular lynchpin in the pathobiology of arteriosclerotic calcification
.
Arterioscler Thromb Vasc Biol
.
2020 Jul
;
40
(
7
):
e193
202
. .
54.
Saito
Y
,
Nakamura
K
,
Ito
H
.
Effects of eicosapentaenoic acid on arterial calcification
.
Int J Mol Sci
.
2020 Jul 30
;
21
(
15
):
21
. .
55.
Wang
P
,
Zhang
N
,
Wu
B
,
Wu
S
,
Zhang
Y
,
Sun
Y
.
The role of mitochondria in vascular calcification
.
J Transl Int Med
.
2020 Jun
;
8
(
2
):
80
90
. .
56.
Boraldi
F
,
Lofaro
FD
,
Quaglino
D
.
Apoptosis in the extraosseous calcification process
.
Cells
.
2021 Jan 12
;
10
(
1
):
131
. .
57.
Phadwal
K
,
Vrahnas
C
,
Ganley
IG
,
MacRae
VE
.
Mitochondrial dysfunction: cause or consequence of vascular calcification?
Front Cell Dev Biol
.
2021
;
9
:
611922
. .
58.
Wang
J
,
Cheng
X
,
Xiang
MX
,
Alanne-Kinnunen
M
,
Wang
JA
,
Chen
H
,
IgE stimulates human and mouse arterial cell apoptosis and cytokine expression and promotes atherogenesis in Apoe-/- mice
.
J Clin Investig
.
2011 Sep
;
121
(
9
):
3564
77
.
59.
Voelkl
J
,
Egli-Spichtig
D
,
Alesutan
I
,
Wagner
CA
.
Inflammation: a putative link between phosphate metabolism and cardiovascular disease
.
Clin Sci
.
2021 Jan 15
;
135
(
1
):
201
27
.
60.
Koliakos
G
,
Paletas
K
,
Kaloyianni
M
.
NHE-1: a molecular target for signalling and cell matrix interactions
.
Connect Tissue Res
.
2008
;
49
(
3
):
157
61
. .
61.
Martinsen
A
,
Baeyens
N
,
Yerna
X
,
Morel
N
.
Rho kinase regulation of vasopressin-induced calcium entry in vascular smooth muscle cell: comparison between rat isolated aorta and cultured aortic cells
.
Cell Calcium
.
2012 Dec
;
52
(
6
):
413
21
. .
62.
Imai
S
,
Okayama
N
,
Shimizu
M
,
Itoh
M
.
Increased intracellular calcium activates serum and glucocorticoid-inducible kinase 1 (SGK1) through a calmodulin-calcium calmodulin dependent kinase kinase pathway in Chinese hamster ovary cells
.
Life Sci
.
2003 Apr 4
;
72
(
20
):
2199
209
. .
63.
Hills
CE
,
Bland
R
,
Bennett
J
,
Ronco
PM
,
Squires
PE
.
High glucose up-regulates ENaC and SGK1 expression in HCD-cells
.
Cell Physiol Biochem
.
2006
;
18
(
6
):
337
46
. .
64.
Taruno
A
,
Niisato
N
,
Marunaka
Y
.
Intracellular calcium plays a role as the second messenger of hypotonic stress in gene regulation of SGK1 and ENaC in renal epithelial A6 cells
.
Am J Physiol Renal Physiol
.
2008 Jan
;
294
(
1
):
F177
86
. .
65.
Sakurada
M
,
Yoshimoto
T
,
Sekizawa
N
,
Hirono
Y
,
Suzuki
N
,
Hirata
Y
.
Vasculoprotective effect of cilostazol in aldosterone-induced hypertensive rats
.
Hypertens Res
.
2010 Mar
;
33
(
3
):
229
35
. .
66.
de Las Heras
N
,
Galiana
A
,
Ballesteros
S
,
Olivares-Álvaro
E
,
Fuller
PJ
,
Lahera
V
,
Proanthocyanidins maintain cardiac ionic homeostasis in aldosterone-induced hypertension and heart failure
.
Int J Mol Sci
.
2021 Sep 4
;
22
(
17
):
9602
.
67.
Lang
F
,
Perrotti
N
,
Stournaras
C
.
Colorectal carcinoma cells: regulation of survival and growth by SGK1
.
Int J Biochem Cell Biol
.
2010 Oct
;
42
(
10
):
1571
5
. .
68.
Rotte
A
,
Pasham
V
,
Eichenmüller
M
,
Mahmud
H
,
Xuan
NT
,
Shumilina
E
,
Effect of bacterial lipopolysaccharide on Na(+)/H(+) exchanger activity in dendritic cells
.
Cell Physiol Biochem
.
2010
;
26
(
4–5
):
553
62
.
69.
Schneider
D
,
Gerhardt
E
,
Bock
J
,
Müller
MM
,
Wolburg
H
,
Lang
F
,
Intracellular acidification by inhibition of the Na+/H+-exchanger leads to caspase-independent death of cerebellar granule neurons resembling paraptosis
.
Cell Death Differ
.
2004 Jul
;
11
(
7
):
760
70
.
70.
Lang
F
,
Föller
M
,
Lang
K
,
Lang
P
,
Ritter
M
,
Vereninov
A
,
Cell volume regulatory ion channels in cell proliferation and cell death
.
Methods Enzymol
.
2007
;
428
:
209
25
.
71.
Lang
F
,
Hoffmann
EK
.
CrossTalk proposal: cell volume changes are an essential step in the cell death machinery
.
J Physiol
.
2013 Dec 15
;
591
(
24
):
6119
21
. .
72.
Golembiewska
E
,
Machowska
A
,
Stenvinkel
P
,
Lindholm
B
.
Prognostic value of copeptin in chronic kidney disease: from general population to end-stage renal disease
.
Curr Protein Pept Sci
.
2017
;
18
(
12
):
1232
43
. .
73.
Golembiewska
E
,
Qureshi
AR
,
Dai
L
,
Lindholm
B
,
Heimbürger
O
,
Söderberg
M
,
Copeptin is independently associated with vascular calcification in chronic kidney disease stage 5
.
BMC Nephrol
.
2020 Feb 7
;
21
(
1
):
43
.
74.
Bjornstad
P
,
Maahs
DM
,
Jensen
T
,
Lanaspa
MA
,
Johnson
RJ
,
Rewers
M
,
Elevated copeptin is associated with atherosclerosis and diabetic kidney disease in adults with type 1 diabetes
.
J Diabetes Complications
.
2016 Aug
;
30
(
6
):
1093
6
.
75.
Schill
F
,
Persson
M
,
Engström
G
,
Melander
O
,
Enhörning
S
.
Copeptin as a marker of atherosclerosis and arteriosclerosis
.
Atherosclerosis
.
2021 Nov 1
;
338
:
64
8
.
76.
Smaradottir
MI
,
Andersen
K
,
Gudnason
V
,
Näsman
P
,
Rydén
L
,
Mellbin
LG
.
Copeptin is associated with mortality in elderly people
.
Eur J Clin Invest
.
2021 Jul
;
51
(
7
):
e13516
. .
Open Access License / Drug Dosage / Disclaimer
This article is licensed under the Creative Commons Attribution-NonCommercial 4.0 International License (CC BY-NC). Usage and distribution for commercial purposes requires written permission. Drug Dosage: The authors and the publisher have exerted every effort to ensure that drug selection and dosage set forth in this text are in accord with current recommendations and practice at the time of publication. However, in view of ongoing research, changes in government regulations, and the constant flow of information relating to drug therapy and drug reactions, the reader is urged to check the package insert for each drug for any changes in indications and dosage and for added warnings and precautions. This is particularly important when the recommended agent is a new and/or infrequently employed drug. Disclaimer: The statements, opinions and data contained in this publication are solely those of the individual authors and contributors and not of the publishers and the editor(s). The appearance of advertisements or/and product references in the publication is not a warranty, endorsement, or approval of the products or services advertised or of their effectiveness, quality or safety. The publisher and the editor(s) disclaim responsibility for any injury to persons or property resulting from any ideas, methods, instructions or products referred to in the content or advertisements.