Next Article in Journal
A Novel Murine Multi-Hit Model of Perinatal Acute Diffuse White Matter Injury Recapitulates Major Features of Human Disease
Next Article in Special Issue
Intrarenal Dopaminergic System Is Dysregulated in SS-Resp18mutant Rats
Previous Article in Journal
Immune Checkpoint Molecules and Glucose Metabolism in HIV-Induced T Cell Exhaustion
Previous Article in Special Issue
Epithelial Sodium Channel Alpha Subunit (αENaC) Is Associated with Inverse Salt Sensitivity of Blood Pressure
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Inverse Salt Sensitivity of Blood Pressure Is Associated with an Increased Renin-Angiotensin System Activity

1
Department of Pathology, The University of Virginia, Charlottesville, VA 22903, USA
2
Division of Endocrinology and Metabolism, Department of Medicine, The University of Virginia, Charlottesville, VA 22903, USA
3
Division of Renal Diseases & Hypertension, Department of Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
4
Department of Physiology/Pharmacology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
*
Author to whom correspondence should be addressed.
Biomedicines 2022, 10(11), 2811; https://doi.org/10.3390/biomedicines10112811
Submission received: 14 October 2022 / Revised: 30 October 2022 / Accepted: 31 October 2022 / Published: 4 November 2022
(This article belongs to the Special Issue State-of-the-Art Hypertension and Biomedicine in the USA)

Abstract

:
High and low sodium diets are associated with increased blood pressure and cardiovascular morbidity and mortality. The paradoxical response of elevated BP in low salt diets, aka inverse salt sensitivity (ISS), is an understudied vulnerable 11% of the adult population with yet undiscovered etiology. A linear relationship between the number of single nucleotide polymorphisms (SNPs) in the dopamine D2 receptor (DRD2, rs6276 and 6277), and the sodium myo-inositol cotransporter 2 (SLC5A11, rs11074656), as well as decreased expression of these two genes in urine-derived renal proximal tubule cells (uRPTCs) isolated from clinical study participants suggest involvement of these cells in ISS. Insight into this newly discovered paradoxical response to sodium is found by incubating cells in low sodium (LS) conditions that unveil cell physiologic differences that are then reversed by mir-485-5p miRNA blocker transfection and bypassing the genetic defect by DRD2 re-expression. The renin-angiotensin system (RAS) is an important counter-regulatory mechanism to prevent hyponatremia under LS conditions. Oversensitive RAS under LS conditions could partially explain the increased mortality in ISS. Angiotensin-II (AngII, 10 nmol/L) increased sodium transport in uRPTCs to a greater extent in individuals with ISS than SR. Downstream signaling of AngII is verified by identifying lowered expression of nuclear factor erythroid 2-related factor 2 (NRF2), CCCTC-binding factor (CTCF), and manganese-dependent mitochondrial superoxide dismutase (SOD2) only in ISS-derived uRPTCs and not SR-derived uRPTCs when incubated in LS conditions. We conclude that DRD2 and SLC5A11 variants in ISS may cause an increased low sodium sensitivity to AngII and renal sodium reabsorption which can contribute to inverse salt-sensitive hypertension.

1. Introduction

Reducing dietary sodium (as NaCl) intake is one of the best proved non-pharmacological interventions for the prevention and treatment of hypertension [1,2,3]. Much is known about the molecular mechanisms involved in salt sensitivity (SS) since many studies have demonstrated a sustained increase in blood pressure (BP) with an increase in salt consumption [1,2,3,4,5,6,7,8,9,10,11,12,13,14,15]. Cardiovascular mortality is higher in individuals consuming high salt diet in salt-sensitive hypertensive and salt-sensitive normotensive than salt-resistant individuals [13,14,15,16,17,18,19]. However, little is known about individuals at the opposite end of the SS spectrum, i.e., inverse salt sensitivity (ISS) [20]. What is known is that death from cardiovascular diseases such as stroke, myocardial infarction, and congestive heart failure are also increased in some individuals who consume low amounts of sodium (<1.2–2.5 g/day) [14,17,18,21,22,23,24,25]. A previously unanswered question is why should low salt intake lead to an increase in cardiovascular mortality? [14,15,16,17,18,21,22,23,24,25]. Others have measured the consequences (e.g., cardiovascular disease and death) in individuals with ISS. Long-term clinical studies (median 16 years, IQR 12–17 years [26,27]) showed that individuals with the ISS phenotype had a 15-fold greater risk of target organ damage than individuals with the salt resistance (SR) phenotype, as determined by clinical evaluation of the heart, kidney, vasculature, and brain, as well as a history of cardiovascular events similar to those seen in salt-sensitive individuals [26]. Our studies are the first to examine the biological, genetic, and epigenetic mechanisms associated with the paradoxical increase in BP in adults consuming a low sodium diet [4]. When approached from a mechanistic viewpoint, future studies will be better equipped to stratify cohorts with ISS appropriately determine if clinical outcomes can be related to known mechanisms of BP regulation in these individuals.
The dietary sodium recommendation by the Institute of Medicine/American College of Cardiology (ACC)/American Heart Association (AHA) [1,19] is intended as a “one size fits all” recommendation. However, it is becoming clear that each individual is genetically programmed with a “personal index of salt sensitivity” [28,29]. Currently, the guidelines set the upper limits but not the lower limits of NaCl intake to prevent and treat hypertension. As aforementioned, cardiovascular risk is increased above and below the intake of 2.5 to 6.0 g sodium/day [5,14,17,18,30,31,32]. Thus, more research is needed to understand the J-curve effect of salt intake on cardiovascular disease and death, especially since both ISS and SS constitute 11% and 18% of the general population, respectively, depending on cohort stratification [22,23,24,30].
In a study of 465 Pennsylvania Amish by Montasser et al. [23], a high sodium (Na) diet (280 mmol/d = 6440 mg/d) for 6 days, and then after a 6- to 14-day washout period, a standardized low-Na diet (40 mmol/d = 920 mg/d) was consumed for 6 days. The potassium (K) in the diet was held constant at 140 mmol/d in both diets. SS was determined by an increase in ambulatory BP [≥5 mm Hg mean arterial pressure (MAP) or systolic BP ≥ 4 mm Hg] on high Na diet. In the studies of Overlack et al., the effects of 1 week of low-salt (20 mmol sodium/d) and 1 week of high-salt (300 mmol sodium/d) diet were investigated in a single-blind randomized study in 163 white non-obese normotensive subjects. Delta BP ≥ 4 mm Hg was use as a cutoff between SR and SS [24,25]. Castiglioni used a low sodium (30 mmol NaCl per day) and a high-sodium (200 mmol NaCl per day) diet, each for 5 days, in random order in 71 Italian participants [32]. The authors of all four studies observed a population of study participants that experienced a change in BP that was opposite to that of individuals with SS [23,24,25,32], that others have shown to be associated with increased mortality and morbidity [17,26].
Urinary surrogate markers, including renal proximal tubule cells (RPTCs), exosomes, and miRNA, hold promise for cost-effective methods to screen for SS and ISS [5,20,28,33,34,35]. Some genetic determinants of SS have been reported, as well [4,28,36,37,38]. Variants of the G protein-coupled receptor kinase type 4 (GRK4) have been shown to be associated with SR and SS in humans [4,37,39,40,41], and cause SR [42] or SS [43] when these human genes are expressed in mice. The genetic background of the mice and the variant expressed determine the resulting BP [44,45]. Variants in the human SLC4A5 gene that encodes the sodium bicarbonate electrogenic cotransporter type 2 (NBCe2) are strongly associated with SS [4]. Angiotensin (Ang) type 1 receptor (AT1R) is an antinatriuretic G protein-coupled receptor previously shown to increase calcium signaling via Ang II in ISS [20]. Other components of the renin-angiotensin system (RAS) may also play a role in the pathogenesis of ISS, such as aminopeptidase N (APN) which converts Ang III into angiotensin IV (Ang IV), reducing the levels of Ang III, the preferred endogenous agonist for the natriuretic Ang II type 2 receptor (AT2R), resulting in sodium retention and elevated BP [46,47]. APN has variable effects on BP [48,49,50]. Decreased cerebral APN activity increases Ang III levels and BP; the cerebral and paraventricular nucleus infusion of APN decreases BP [49]. However, in the kidney, a defect in Ang III signaling through the AT2R occurs in both prehypertensive and hypertensive spontaneously hypertensive rats (SHR) [51]; increased production of Ang III and decreased renal APN activity rescues this defect and inhibits renal sodium transport and decreases BP in SHR [50]. By contrast, nanomolar concentrations of Ang IV have been reported to increase BP in rats by interacting with the AT1R [48]. Some effects of Ang IV in the kidney may be through the binding and inhibition of insulin-regulated aminopeptidase (IRAP) activity [52]; global IRAP knockout mice have increased vasopressin sensitivity, yet no change in blood pressure [53].
The dopamine type 2 receptor (D2R) negatively interacts with the AT1R in many tissues, including the kidney [54,55] DRD2 rs6276 (1347G>A) at 3′UTR (MAF 0.484) has decreased D2R function and DRD2 rs6277 (957C>T exon7 synonymous mutation, MAF = 0.273) has decreased DRD2 mRNA stability [56]. D2R has a protective role against inflammation and these DRD2 variants increase inflammation and fibrosis in human and mouse RPTCs [57,58,59,60]. The present studies tested the hypothesis that DRD2 variant-mediated dysregulation of the RAS contributes to the pathogenesis of ISS. Other genes that are expressed in the kidney, such as SMIT1 (SLC5A3) [61] and SMIT2 (SLC5A11, SGLT6) (Na+/myoinositol cotransporters), may be related to ISS. SMIT2 is responsible for the apical sodium/myoinositol transport in the RPT [62].
Although the harmful effects of lowering sodium consumption is controversial for the general population that is not hypertensive or salt-sensitive, the simple observation that there are individuals in every SS study that have a paradoxical blood pressure response to lowering sodium consumption is not. Our group has previously demonstrated direct ex vivo cell physiologic differences in urine-derived renal proximal tubule cells (uRPTCs) in ISS individuals. We extend these studies by culturing these uRPTCs to show that they have enduring sodium-induced phenotypic changes that are amenable to genetic and epigenetic phenotypic reversal centered on D2R expression and Ang II sensitivity.

2. Materials and Methods

2.1. Living Renal Proximal Tubules Obtained from Spot Urines

The human tissues and urine specimens used in our studies were obtained in accordance with a University of Virginia Institutional Review Board-approved protocol that adheres to the Declaration of Helsinki and the most recent version of the USA Code of Federal Regulations Title 45, Part 46 [33]. We obtained spot urine collections from salt study participants described in a previous publication [4]. Briefly, we used the statistical tests to classify participants who successfully completed our salt diets (based on 24-h urine salt balance evaluation) into ISS, SR, and SS. Statistical justification for the cutoff points between the ISS, SR, and SS groups was performed, as previously published [47].

2.2. uRPTC Culture

Spot, mid-stream and clean-catch, urine (>200 mL) was centrifuged within 2 h after collection in 50 mL tubes at 1000 relative centrifugal force (RCF) for 3 min. The supernatant was removed, and the pellets were combined into one 50 mL tube, filled with PBS (Dulbecco’s PBS + CaCl2 + MgCl2, D8662, Sigma-Aldrich, St. Louis, MO, USA), centrifuged again, and the supernatant removed. Two ml of culture medium (REGM, cc-3190, Walkersville, MD, USA) were added to the pellet and plated in a 96-well tissue culture plate at 200 µL per well. Fifty µL of spent media were exchanged with fresh media every other day. The cell colonies appeared in 7–14 days. The cells were immortalized with lentiviral hTERT, as previously described [63].

2.3. Plasma Membrane D1R and AT2R Expression

uRPTCs were directly isolated and imaged for D1R [20], and for AT1R [64], as previously described, with the exception that a monoclonal antibody was generated related with the prospects of developing diagnostic clinical assays. Briefly, in TERT-immortalized uRPTCs, intracellular sodium was increased by the ionophore monensin [65] (MON, 10 μmol/L, 1 h), and D1R and AT2R recruitments were measured directly, using fluorescently labeled antibodies in collagen-coated glass-bottomed 96-well plates. The effect of the D1-like receptor agonist fenoldopam (FEN, 1 μmol/L, 30 min)-induced plasma membrane AT2R recruitment was performed identically, as in the D1R assay [20], except for the additional use of AT2R monoclonal antibodies.

2.4. Plasma Membrane D2R and APN Expression

Fixed, non-permeabilized in-cell western blotting, using directly labeled primary antibody against a synthetic (PLKEAARRC) peptide, corresponding to amino acids 239–246 of the third extracellular loop of the human D2R (324393, EMD Millipore, Burlington, MA, USA) was performed, as previously described [58,59,60,66]. Briefly, uRPTCs, plated in collagen-coated 96-well glass bottom microplates, were fixed with 4% paraformaldehyde without detergent in phosphate-buffered saline (PBS), blocked with Odyssey blocking reagent (927–40003, Licor, Lincoln, NE, USA) in PBS, and incubated with an extracellular epitope-specific rabbit anti-D2R antibody (EMD Millipore), directly conjugated with Alexa-647 (ab269823, Abcam, Cambridge, MA, USA), or purified monoclonal antibody against APN, also known as CD13, clone 452 (provided by Dr. Meenhard Herlyn, The Wistar Institute of Anatomy and Biology, Philadelphia, PA, USA), directly conjugated with Alexa-647, and read in a fluorescent microplate reader in well-mode (PHERAstar FS, BMG, Raleigh, NC, USA). Plating efficiency was normalized by taking the in-cell D2R fluorescence values, divided by simultaneous measurement of Hoechst 33,342 (735969, 1:2000, Thermo Fisher, Waltham, MA, USA) staining.

2.5. Total Cellular CTCF, NRF2, and SOD2 Expression

Fixed-permeabilized in-cell western blotting, using directly labeled primary antibody against CCCTC-binding factor (CTCF, sc-271474, Santa Cruz Biotechnology, Dallas, TX, USA), nuclear factor erythroid 2-related factor 2 (NRF2, sc-365949, Santa Cruz Biotechnology), and manganese-dependent mitochondrial superoxide dismutase (SOD2, sc-137254, Santa Cruz Biotechnology) was performed as above, with the exception that the cells were permeabilized with 0.2% Triton-X100 for 5 min, following fixation.

2.6. D2R Binding Assay

D2R fluorescence binding was measured by treating live cells with a fluorescently labeled D2R antagonist, spiperone (sc-206629, Santa Cruz Biotechnology) [67,68,69], or bodipy-propanoic acid N-phenethylspiperone amide (aka Spiperone-FL, 100 nmol/L, 2 h). The cells were washed, and the fluorescence was read in a fluorescent plate reader (ex 488 em 510). Spiperone has similarly high affinities for two members of the dopamine D2-like receptor family, D2R and D4R, and 10-fold lower affinity for the D3R and many-fold lower affinity for members of the dopamine D1-like receptor family, D1R, and D5R [67,68]. However, addition of a D4R selective antagonist (L-741742, 200 nmol/L), or a D3R antagonist (U99194A, 1 µmol/L) [69,70,71] did not significantly decrease spiperone-FL binding (data not shown).

2.7. Dopamine D2R siRNA

uRPTCs were transfected overnight with 50 nmol/L DRD2 siRNA or control non-targeting siRNA (D-001210-02-50, Dharmacon, Cambridge, UK), using Lipofectamine RNAiMAX (13778075, Thermo Fisher). D2R binding was measured 48 h later.

2.8. D2R Overexpression

We purchased a lentiviral construct (DRD2 long) from GeneCopoeia. The plasmid was packaged into virus with compatible packaging plasmids, using HEK-293T cells (Clontech Laboratories, Mountain View, CA, USA). The lentivirus was added to uRPTCs at 30–40% confluence for 18–20 h, then removed and replaced with regular growth medium. After 48 h, the medium was changed to selection medium, containing puromycin (Sigma, 5 μg/mL). The DRD2 long was used, instead of DRD2 short, because the former is the isoform expressed in the kidney [71].

2.9. Sodium Influx Assay

Details and specificity of this assay, in which ouabain was used to inhibit Na+-K+/ATPase activity, permitting the quantification of sodium accumulation in RPTCs, were previously published [72]. Briefly, the uRPTCs were cultured in 96-well glass bottom collagen-coated Matrical plates (Dot Scientific, Burton, MI, USA, MGB096-1-2LGL) at 37 °C until they reached 80% confluence. The cells were serum-starved overnight prior to loading with the sodium ion indicator, sodium-binding benzofuran isophthalate acetoxymethyl ester (SBFI, 5 μmol/L; ThermoFisher), with 0.04% Pluronic F-127 for 2 h in PBS with calcium (0.9 mmol/L) and magnesium (0.49 mmol/L). The cells were washed twice and allowed to recover at 37 °C in serum-free media for 30 min. They were washed two more times with PBS and incubated at room temperature. The cells were then placed in a fluorescence plate reader (Pherastar FS, BMG) and ratiometric readings (340 nm excitation and 510 nm emission/380 nm excitation and 510 nm emission) were recorded every 30 s for 30 min. A 5-min baseline reading was acquired prior to automatic injection of ouabain (100 μmol/L), to inhibit basolateral sodium transport. The change in sodium accumulation was measured as the change in 340/380 ratio at the 30-min time point minus the 5-min average reading.

2.10. Seminaphtharhodafluor (pH-Sensitive Dye, SNARF), Sodium-Hydrogen Exchanger Type 3 (NHE3), and Sodium-Dependent pH Recovery Assay in Low Sodium Concentrations

uRPTCs, plated in thin bottom plastic tissue culture-treated 96 well plates (uClear, Greiner Bio-One, Stonehouse, UK) were incubated in low sodium concentration (90 mmol/L sodium) for 2 h, using serum-free DMEM/F12 media diluted with water and mannitol to make the solution iso-osmolar (290 mOsm/L). In order to control for other factors in the physiologic serum-free media, normal serum-free media in DMEM/F12 was also diluted with the same amount of water as low sodium media but adjusted to 140 mmol/L sodium as sodium chloride. uRPTCs were loaded with carboxy SNARF-1 AM acetoxymethyl ester acetate in low-sodium Hank’s Balanced Salt Solution (HBSS, 90 mmol/L sodium, osmolality adjusted to 290 mOsm/L with mannitol) for 1 h, washed and incubated with 20 mmol/L ammonium chloride for 10 min, and changed to serum-free HEPES-buffered saline (HBS, 5 mmol/L HEPES, 140 mmol/L choline chloride, 1 mmol/L calcium chloride, 1 mmol/L magnesium chloride, 4 mmol/L potassium chloride), to stabilize the extracellular pH in the absence of CO2, but without bicarbonate which would lower the intracellular pH, which is necessary in order to measure sodium-dependent pH recovery, without interference with NBCe2 activity [70]. The microplate was transferred to a fluorescence microplate reader (PHERAstar FS, BMG), equipped with a single excitation (520 nm) and dual emission (640 nm/580 nm) custom filter set for SNARF-1 and baseline reading taken for 5 min before switching to low sodium containing HBS (5 mmol/L HEPES, 90 mmol/L sodium chloride, 50 mmol/L choline chloride, 1 mmol/L calcium chloride, 1 mmol/L magnesium chloride, 4 mmol/L potassium chloride). Full plate measurements were taken every 10 s and the slope of the first 2 min was used to record pH recovery. The pH was internally calibrated using the nigericin high potassium calibration method [73].

2.11. Micro Ribonucleic Acid 485-5p (miR-485-5p) Mimic and Blocker Transfection

miR-485-5p mimic (50 nmol/L), miR-485-5p blocker [74,75,76], or control miRNA (Mission, Sigma-Aldrich) was transfected overnight into the uRPTCs, using Lipofectamine RNAiMAX™ and D2R binding measured after 48 h.

2.12. SLC5A11 SNP and qRT-PCR

SLC5A11 SNP rs11074656 (C/T), DRD2 SNPs rs6276 and rs6277 were identified using Taqman PCR probes (Thermo Fisher), a two-step fast cycling real-time PCR machine (BioRad CFX96 Connect, Hercules, CA, USA), and SNP determination by CFX Maestro 2. SLC5A11 mRNA was quantified using real-time quantitative reverse transcriptase PCR (qRT-PCR), as previous described [77]. Primers: SLC5A11 sense 5′-GCCTCCACAGTTAGATCCCC-3′; SLC5A11 anti-sense 5′-CAGAACTAGCACCGCGATG-3′; actin sense 5′-AGAAAATCTGGCACCACACC-3′; Actin anti-sense 5′-CTCCTTAATGTCACGCACGA-3′.

2.13. Statistical Analysis

Statistical analysis was performed using GraphPad Prism 9 software (GraphPad, San Diego, CA, USA) as well as engaging the University of Virginia’s Medical School’s Department of Biomedical Statistics. Response differences were evaluated using t-test and analysis of variance with post hoc Bonferroni and Tukey tests for repeated measures. For most studies, we used power calculations based on the endpoints of major interest: sodium transport, expression, and activity of the members of the RAS, D2R expression and activity, miRNA expression, and downstream messengers. We justified selecting a Delta MAP of ≥+7 mm Hg for Defining SS and ≤−7 mm Hg for ISS (With the assistance of our collaborating biostatistician our statistical method that assures us that the cutoffs for classifying ISS and SR are no longer arbitrary [7].

3. Results

3.1. D1R and AT2R Plasma Membrane Recruitment

With 1-h 10 µM monensin (MON) treatment, more D1Rs (Figure 1A) and AT2Rs (Figure 1B) are recruited to the plasma membrane in cells from individuals with ISS, but not in cells from individuals with SR (D1R: MON/VEH: SR, 1.022 ± 0.038, n = 6; ISS, 1.635 ± 0.178, n = 6; t-test, p < 0.01; AT2R: MON/VEH: SR, 0.965 ± 0.04, n = 6; ISS, 1.434 ± 0.107, n = 6; t-test, p < 0.01).

3.2. Direct AT2R Plasma Membrane Recruitment in uRPTC

We have previously shown that the sodium-induced recruitment of the D1R to human RPTC plasma membrane correlates with the degree of SS in a clinical study [20]. Here, we extend those studies by showing the relationship between D1-like receptor agonist FEN-induced AT2R plasma membrane recruitment and the degree of SS. Unlike the linear relationship previously found for D1R and SS [20], AT2R plasma membrane recruitment was only increased in subjects with ISS (AT2R: FEN/VEH: ISS, 1.511 ± 0.13, n = 4; SR, 0.955 ± 0.04, n = 4; SS, 1.148 ± 0.082, n = 3; one-way ANOVA, Holm-Sidak post hoc test ** p < 0.01) (Figure 1C).

3.3. D2R Binding

Immunostaining showed that D2R was expressed in the human RPT (Figure 2A). To determine if D2R participates in the pathogenesis of ISS, ten uRPTCs, 5 isolated from clinical study participants with ISS and 5 isolated from clinical participants with SR, were grown, isolated, immortalized with Tert, and plated as 8 technical replicates in 96-well glass-bottom collagen-coated Matrical plates and grown to 80% confluence. Plasma membrane D2R expression was measured using an extracellular epitope-specific rabbit anti-D2R antibody and found to be reduced by 25.3 ± 0.5% in uRPTCs from clinical study participants with ISS (p < 0.01, n = 54/group), relative to SR cells (Figure 2B,C).

3.4. Total Cellular CTCF, NRF2, and SOD2 Expression

Since DRD2 contains five topologically associated domains containing CCCTC-binding factor (CTCF) studied by Chipseq [78], we measured CTCF expression in uRPTCs. CTCF was reduced by 16.75 ± 1.22% in ISS uRPTCs incubated in low salt concentration (90 mmol/L sodium) (p < 0.01 ISS 90 mmol/L sodium vs. ISS 140 mmol/L (normal) or 190 mmol/L sodium as NaCl (high) salt concentration, n = 9) (Figure 3A). Because D2Rs and dopamine are implicated in redox signaling in the RPT, we measured the expression of the protein responsible for controlling the majority of antioxidant response elements [79,80], i.e., basic leucine zipper (bZIP) protein (NRF2, Nuclear Factor Erythroid 2 Like 2, aka NFE2L2). NRF2 expression in uRPTCs was reduced by 12.73 ± 0.49% in ISS only under low salt (90 mmol/L) concentration (p = 0.0032 ISS low salt vs. ISS 140 or 190 mmol/L salt, as NaCl, n = 18) (Figure 3B). To validate the functionality of the reduced expression of NRF2, superoxide dismutase 2 (SOD2), a downstream antioxidant gene regulated by NRF2 [81], was studied. SOD2 expression in uRPTCs was reduced by 34.23 ± 6.52% in ISS only under low NaCl concentration (p < 0.01 ISS low salt vs. ISS 140 or 190 mmol/L NaCl, n = 18) (Figure 3C).

3.5. Ang II-Stimulated Sodium Influx in ISS and SR

Our previous study showed that uRPTCs from clinical study participants with ISS fed a low salt diet (10 mEq/day) for one week that had an increase in BP, displayed a heightened Ang II-mediated increase in intracellular calcium, relative to individuals with SR or SS [20]. Therefore, Ang II sensitivity was evaluated in these uRPTCs. We measured the effect of Ang II (10 nmol/L) on sodium influx with the sodium-sensitive ion indicator, SBFI. The uRPTCs were incubated with vehicle or Ang II (10 nmol/L, 10 min), in which basolateral sodium outflux was inhibited by incubating cells in media with ouabain (100 µmol/L). Sodium influx was similar in vehicle-treated uRPTCs from individuals with SR and ISS. uRPTCs showed a robust increase in sodium influx when stimulated with Ang II in SR (15,426 ± 900 RFU SR vehicle (VEH) vs. 20,357 ± 667 RFU SR Ang II, n = 15, * p < 0.01) and ISS (15,571 ± 904 RFU ISS VEH vs. 26,942 ± 815 RFU ISS Ang II, n = 15, ‡ p < 0.001) individuals (Figure 4). However, the sodium influx in response to Ang II was greater in uRPTCs from individuals with ISS than individuals with SR (ISS 26942 ± 815 RFU vs. SR 20357 ± 667 RFU, n = 15, † p < 0.001) (Figure 4).

3.6. D2R Extracellular Epitope Binding and D2R Overexpression in Living uRPTCs

To determine the role of D2R in the differential effect of Ang II in the sodium influx in uRPTCs from individuals with SR or ISS, we decreased D2R expression with D2R siRNA or overexpressed D2R and measured D2-like receptor binding by live-96-well fluorometry, using fluorescent spiperone, a D2-like receptor antagonist with high affinities to the members of the D2-like receptor family, D2R = D4R = D3R, [67,68]. In uRPTCs treated with control siRNA, spiperone binding was lesser in individuals with ISS than those with SR (36.9% ± 2.6% reduction in ISS control siRNA vs. SR control siRNA, n = 5, † p < 0.01) (Figure 5A), confirming the results using the extracellular epitope-specific D2R antibody (Figure 2). Spiperone binding in SR uRPTCs was reduced by DRD2 siRNA (37.1% ± 2.0% reduction in SR DRD2 siRNA vs. SR control, n = 5, * p < 0.01). Spiperone binding in ISS uRPTCs was reduced by DRD2 siRNA (42.7% ± 6.0% reduction in ISS DRD2 siRNA vs. ISS control, n = 5, * p < 0.01). The percent reduction in spiperone binding was not different between the SR and ISS uRPTCs. D2R overexpression increased spiperone binding in SR RPTCs (36.7% ± 7.9% increase over SR control siRNA (n = 5, ‡ p < 0.01). D2R overexpression also increased spiperone binding in ISS uRPTCs (22.5% ± 4.8% increase over SR control siRNA (n = 5, ‡ p < 0.01). The percent increase in spiperone binding in D2R overexpression in uRPTCs was not different between individuals with SR or ISS (Figure 5A). These results suggest that the spiperone binding assay is specific and sensitive enough to measure endogenous and exogenous D2R expression in uRPTCs, and not affected by SS, ISS or otherwise.

3.7. Epigenetic Regulation of the D2R by Mir-485-5p

We studied the effect of miR-485-5p mimic and blocker on spiperone (D2-like receptor antagonist) binding to test the hypothesis that endogenous miRNA may regulate D2R expression in uRPTCs [67,68]. The DRD2 SNP rs6726, located in the DRD2 3′UTR (untranslated region), results in a sequence that could cause an epigenetic downregulation of the synthesis of the D2R [72]. miR-9-5p, miR-485-5p, and miR-137 regulate schizophrenia risk genes and miR-9-5p targets the DRD2, a drug-target in schizophrenia [74,75,76]. An miR-485-5p mimic and blocker were synthesized and transfected into uRPTCs to increase or decrease miR-485-5p binding to target genes, respectively. Spiperone binding was lesser in control miRNA-treated uRPTCs from individuals with ISS than those with SR (Figure 5B), similar to that found in untreated uRPTCs (Figure 5A). miR-485-5p blocker increased while miR-485-5p mimic decreased the expression of the D2R only in uRPTCs from individuals with ISS and DRD2 SNPs rs6276/rs6277 (52,619 ± 2001 RFU ISS control miRNA vs. 69,496 ± 2108 RFU ISS miRNA blocker or vs. 30,434 ± 1824 RFU ISS miRNA mimic, n = 9, †, ‡ p < 0.01) (Figure 5B). Thus, miR-485-5p only binds to the DRD2 SNPs, leading to the reduction in D2R expression. The miRNA mimic behaves just like the endogenous miR-485-5p miRNA and reduced the already decreased expression of D2R in uRPTCs from individuals with ISS. The spiperone binding returned to normal when the cells were transfected with a miR-485-5p miRNA blocker which inhibited the endogenous miRNA. Because miR-485-5p has the highest energy binding level to DRD2 rs6276 [74], the above result suggests that rs6276 creates a neomorphic genetic alteration since the miR-485-5p mimic and blocker have no significant effect on D2R wild-type. The lower D2R expression in ISS uRPTCs due to miR-485-5p binding at the rs6276 SNP site can be bypassed by stable overexpression (Figure 5A), since the D2R lentiviral construct has a different 3′UTR (sv40 3′UTR and poly A sequence), relative to the endogenous DRD2 gene.

3.8. The Effect of D2R Overexpression on Sodium Transport in uRPTCs

Stable DRD2 long (D2L)-overexpressing uRPTCs were then evaluated to determine if the Ang II-stimulated sodium transport observed in uRPTCs from individuals with ISS when incubated in media with a low salt (NaCl) concentration, could be reverted to cells that respond to Ang II in a normal manner, similar to uRPTCs from individuals with SR. SNARF1 microplate fluorometry was utilized to measure NHE3-dependent pH recovery. Ang II increased pH recovery in low salt (90 mM) concentration only in ISS uRPTCs without DRD2-long (wo) overexpression (ISS Ang II vs. ISS VEH, n = 5 per group, * p < 0.05), an effect that was completely blocked by the AT1R antagonist losartan (LOS) (ISS Ang II vs. ISS Ang II LOS, n = 5/group, ‡ p < 0.05) (Figure 6A), which by itself had no effect. Stable overexpression of DRD2 long (w D2L) completely blocked the low salt Ang II-dependent increase in pH in ISS uRPTCs (ISS Ang II vs. ISS Ang II w D2L, n = 5/group, † p < 0.05) (Figure 6A).
The Ang II precursor peptide, Ang I, increased pH recovery in low salt concentration only in ISS uRPTCs without DRD2long (wo) overexpression (ISS Ang I vs. ISS VEH, n = 5 per group, * p < 0.05), an effect that was completely blocked by the AT1R antagonist LOS (ISS Ang I vs. ISS Ang I LOS, n = 5 per group, ‡ p < 0.05), which by itself had no effect (Figure 6B). Stable overexpression of DRD2 long (w D2L) completely blocked the low salt Ang I-dependent increase in pH in ISS uRPTCs (ISS Ang I vs. ISS Ang I w D2L, n = 5 per group, † p < 0.05) (Figure 6B).

3.9. RPTC Plasma Membrane Amino Peptidase N Expression

Amino peptidases A (APA) and N (APN) convert Ang II to Ang III and Ang III to Ang IV, respectively, with Ang III abrogating the effects of Ang II [46,50,82]. Plasma membrane APN expression was measured in uRPTCs from individuals with ISS or those with SR in either normal salt (140 mM sodium as NaCl) (NS) containing media or iso-osmotic media (290 mOsm/L) containing 90 mM sodium as NaCl (LS), as above. In-cell immunofluorescence staining in 96 well plates using an extracellular epitope-specific antibody to APN showed that plasma membrane APN expression was greater in ISS than SR uRPTCs. APN expression (60.20 ± 6.95%) was higher in ISS than SR uRPTCs incubated in 140 mmol/L sodium as NaCl (p < 0.01, ISS vs. SR, n = 18/group) and increased further to 72.71 ± 9.83% with incubation in 90 mmol/L sodium (p < 0.05, ISS in 90 mmol/L sodium as NaCl vs. ISS 140 mmol/L sodium as NaCl, n = 18/group) (Figure 7).

3.10. D2 SNPs and SLC5A11 SNP

The number of SNPs in DRD2 (rs6276, rs6277) correlates with the increase in MAP [57]. We also found that a SLC5A11 SNP, rs11074656, is associated with ISS (Table 1). The odds ratio is 2.29, indicating that the odds of expressing ISS are 2.29 times higher in individuals who have rs11074656 compared with individuals who express the normal homozygous major variant. In subjects carrying both SLC5A11 and DRD2 SNPs, those expressing more total SNPs had higher increase in MAP on low salt diet, relative to high salt diet, i.e., ∆MAP (Low Salt MAP minus High Salt MAP) (Figure 8A). The ∆MAP was significantly higher in those with 5–6 SNPs than those without or with 1 SNP (0–1: −2.12 ± 0.81, n = 58; 2–4: −1.14 ± 0.52, n = 190; 5–6: 0.98 ± 0.88, n = 54 p < 0.05, one-way ANOVA, Dunnett’s test) (Figure 8B). Moreover, there was significantly lower SLC5A11 expression in those with homozygous SNPs than those with wild-type SLC5A11 (Figure 8C), possibly contributing to their elevated BP on low sodium diet, as a result of additional increase in renal sodium reabsorption via the sodium/myo-inositol cotransporter 2 (SLC5A11).

4. Discussion

Understanding the causes and pathogeneses of hypertension are important because it is one of the most important risk factors for CVD [1,2,3,5,6,7,8,11,12,13,14,15,17,18,19,83]. Hypertension can be caused or exacerbated by an increase in the intake of salt, in individuals with SS [1,2,3,4,5,6,7,8,9,10,11,12,13,14,15,17,18,19,29,36,37,38,39,40]. However, about 11% of individuals have increased BP when the intake of salt is very low (<1.2–3 g/day) [23,24,25,47,84,85]. In one recent long-term study (median 16 years, IQR 12–17 years [26]) individuals with ISS had a 15-fold increase of end-organ damage and mortality compared with individuals with SR. Whereas several genes have been proposed to cause SS [4,36,37,38,39,40,41,43,45], little is known about genes that contribute to ISS. We hypothesize that there may be many genetic contributors to chronic diseases such as ISS since no single gene has been shown to be solely responsible for SS of BP [36].
We now report that the D2R may contribute to the pathogenesis of ISS since the number of variants in the DRD2 UTR, i.e., rs6276/rs6277 is associated with an increase in BP in individuals fed a low sodium diet. The proposed model of ISS in RPTCs under low sodium condition is depicted in cartoon form (Figure 9). In the left panel is the RPTC of individual with SR, in low sodium conditions. The angiotensin peptides are converted by enzymes found in the apical brush boarder and bind and stimulate the corresponding preferred receptors and lead to the proper regulation of sodium hydrogen exchanger type 3, NHE3 and sodium pump, Na+K+/ATPase to maintain normal sodium reabsorption, maintain sodium homeostasis, and normal blood pressure. An RPTCs from an individual with ISS and DRD2 SNPs and SLC5A11 SNPS is depicted in the right panel. In ISS individuals with DRD2 SNPs, the lowered expression of D2R and loss of negative regulation of AT1R and APN leads to a loss of Ang III and AT2R signaling and enhanced AT1R sensitivity. Increased sensitivity to Ang II leads to increased sodium reabsorption, as well as the downstream effects of reduced NRF2, CTCF, and SOD2. The decreased expression of NRF2 also leads to decreased expression of SLC5A11 which is further exacerbated by SNPs in SLC5A11 that also reduce expression.
DRD2 variants rs6276 and rs6277 reduce D2R expression/activity [57], reduced D2R expression/activity is associated with hypertension and related diseases, such as obesity and metabolic syndrome [86,87,88]. The long isoform of D2R is the isoform expressed in the kidney [71]. The D2R has been shown to inhibit renal sodium transport and contribute to the maintenance of normal fluid and electrolyte balance and BP [54,56,79,80,86,89,90]. D2R is expressed in RPT and other nephron segments in rats [71,86], mice [91], and humans [92]. D2R expression in the medullary collecting duct is species-specific [86,93].
One mechanism by which DRD2 variants rs6276 and rs6277 impair D2R function is via a decrease in D2R expression [57,74]. The overall effect of dopamine receptors on renal sodium transport is modulated by other receptors and hormones [90,93], including those related to the renin-angiotensin-aldosterone system [45,54,94]. The ability of Ang II to stimulate Na+, K+/ATPase activity in rat RPTs is impaired [54], by the D2-like receptor agonist, bromocriptine (D2R = D3R > D4R) [54,95]. Our studies show that either Ang II (10 nmol/L) or Ang I (10 nmol/L) increases luminal sodium transport to a greater extent in uRPTCs from individuals with ISS than uRPTCs from individuals with SR. These effects are probably exerted at the AT1R because they were blocked by the AT1R blocker, losartan. The apparent increase in AT1R activity in uRPTCs in individuals that express ISS is due to decreased D2R expression in ISS, related to DRD2 variants rs6276 and rs6277, because overexpression of DRD2 long abrogates the greater increase in luminal sodium transport induced by Ang II in these uRPTCs from individuals with ISS.
APA and APN convert Ang II to Ang III and Ang III to Ang IV, respectively, with the natriuretic effect of Ang III opposing the antinatriuretic effect of Ang II [46,82,96]. Ang II and Ang III can bind to both the AT1R and the AT2R, but the affinity of Ang II for AT1R is higher than for AT2R while the affinity of Ang III for AT2R is higher than for AT1R. Ang IV binds to the Ang IV Receptor, also known as IRAP and inhibits protease activity [52]. In uRPTCs incubated in normal sodium concentration (140 mM sodium as NaCl), APN expression which is already higher in uRPTCs from individuals with ISS than those with SR is increased to a greater extent by low sodium concentration (90 mM sodium as NaCl) in individuals with ISS than SR. Ang IV has been reported to decrease renal sodium transport [97] and increases sodium excretion in some studies but has no effect on sodium excretion in other studies [98]. Ang IV may have effects on the renal vasculature and renal blood flow that are biphasic with the vasoconstrictor and pressor responses being mediated through AT1R while the prolonged vasodilator activity being through IRAP [99]. IRAP knockout mice do not have altered blood pressure, but do have increased vasopressin sensitivity during pregnancy [53]. However, we have reported that Ang III increases renal sodium excretion by inhibiting luminal NHE3 and basolateral Na+/K+-ATPase activity in Wistar-Kyoto but not spontaneously hypertensive rats [96]. Thus, the increased activity of Ang II in individuals with ISS can be related to a decreased counter-regulation of Ang III via AT2R. We hypothesize that this is due to decreased stability of Ang III in individuals with ISS in low sodium concentration caused by higher APN expression/activity.
In addition to the contribution of the D2R in the maintenance of homeostasis by the regulation of renal sodium transport and BP, another protective action of the D2R via the suppression of reactive oxygen species (ROS) production and inflammation [44,59,60,72,79,80,89,90,91,92,93,100,101,102]. This occurs, in part, through heterodimerization with receptors and proteins such as DJ1 which prevents the degradation of the antioxidant NRF2 [78]. We have shown that NRF2 is lower in individuals with ISS than individuals with SR under low, but not high, sodium concentration, which could be at least one mechanism why a decrease in the expression of D2R increases inflammation [57,80]. uRPTCs from ISS individuals have reduced NRF2 expression, as well as reduced expression of several downstream genes of NRF2, such as superoxide dismutase (SOD2), and CTCF [101]. CTCF stimulates the D2R so that the reduced expression of CTCF in ISS may be part of the mechanism that decreases D2R expression.
Epigenetics may also be involved in the etiology of ISS. The epigenetic regulation of gene transcription and translation can be influenced by diet [102,103,104,105]. For example, an increase or decrease in oxidative stress caused by diet can provoke changes in epigenetics [106,107]. Thus, increased salt intake can increase oxidative stress and oxidative stress can influence epigenetics [108,109,110,111]. The renal D2R and D5R receptors regulate several redox enzymes [44,59,79,80,89,90,93,111,112]. We have previously reported that miR-124 expression is increased in urinary exosomes of individuals with SS [33]. miR-124 can regulate c-Myc, which we have reported to increase GRK4 expression which impairs D1R function [113]. C-myc is negatively regulated by CTCF [114], a conserved transcriptional repressor found in RPTCs that binds to a region of the chromosome 100 bases away from the DRD2 variant rs6276. We, then, studied miR-485-5p, which lies in the 3″ UTR of the DRD2. miR-9-5p, miR-485-5p, and miR-137 regulate schizophrenia risk genes and miR-9-5p targets the DRD2, a drug-target in schizophrenia [74,75,76]. In this report, we demonstrated that miR-485-5p only binds to the rs6276 D2R SNP, thus reducing D2R expression. We used a miRNA mimic that behaves just like the endogenous mir-485-5p miRNA and found it to reduce the expression of D2R, but only in the uRPTCs obtained from individuals with ISS. When transfected with a miRNA blocker, the endogenous miRNA is inhibited and the expression of D2R returns to normal. We have reported 14 additional micro RNAs to be associated with ISS [33]. Among the 14 studied miRNAs is miRNA 30c-1-3p which is pro-inflammatory [115]; its role in the inflammation that occurs with decreased D2R expression [44,57,59,60,89,93,100,101,102] remains to be determined.
SNPs involved with aberrant blood pressure regulation, especially those involved with sodium balance, could contribute to either the etiology of ISS or SS when compared to SR. Ji and others have discovered SNPs that confer protection against hypertension [116]. We recently showed that one subunit of the Na+ channel (alpha ENaC) is involved in the etiology of ISS [117]. Here, we find that the RPT apical Na+/myoinositol cotransporter 2 (aka SMIT2, SLC5A11, and SGLT6) containing homozygous SNP at rs11074656 is decreased in individuals with ISS [47]. In addition to the SNP-dependent decrease in expression of SLC5A11, the reduced expression of SLC5A11 in low salt conditions in ISS may be due to SLC5A11 being a known positively regulated specific target of the transcription factor, NRF2 [118]. Similarly, the increased expression of SLC5A11 in high salt conditions may be related to our previously published pathway of high salt induction of HNF4A in hRPTCs [72], and HNF4A enhances the expression of SLC5A11 [119].

5. Conclusions

We demonstrated that DRD2 variants rs6276 and rs6277, which decrease D2R expression and function, may be important in pathogenesis of ISS, a state in which BP is increased by a low salt diet. The number of DRD2 rs6276/rs6277 is associated with an increase in BP when sodium intake is changed from high salt (300 mmol sodium/day) diet to a low salt diet (10 mmol sodium/day). The presence of DRD2 rs6276/rs6277 decreases the RPTC plasma membrane expression D2R and its affinity to D2-like receptor antagonist. The decreased expression of D2R in cells expressing DRD2 rs6276/rs6277 can be related to increased binding of miR-485-5p miRNA at rs6276. The presence of DRD2 rs6276/rs6277, by themselves, does not affect uRPTC luminal sodium transport, but is augmented by Ang II, via the AT1R. The expressions of the antioxidant proteins NRF2 and SOD2 are decreased in uRPTCs with DRD2 rs6276/rs6277 incubated in low (90 mmol/L NaCl) but not normal (140 mmol/L NaCl) or high salt (190 mmol/L NaCl) concentration. We conclude that the elevated BP found in individuals with ISS in response to a low salt intake could be caused by polymorphisms in DRD2 that decrease D2R expression and function, resulting in increased luminal sodium transport in the RPT caused by increased sensitivity to Ang II, via AT1R. A decreased concentration of Ang III caused by an increase in APN expression may also play a role in the ISS phenotype. We also found another SNP, SLC5A11 rs11074656, that is also positively associated with ISS. Given that ISS and SS constitute ~33% of the US population and both are associated with increased morbidity and mortality [110,111] then there is a need for diagnostic markers for these conditions. Surrogate markers obtained from random urine specimens may be used in the future to determine each individual’s personal salt index [20,26,33,40,44] that might encourage dietary adjustments leading to improved population health.

Author Contributions

Conceptualization, J.J.G. and R.A.F.; methodology, J.J.G.; validation, J.J.G., R.M.C., P.A.J. and R.A.F.; formal analysis, J.J.G., P.X. and K.A.S.; investigation, J.J.G. and P.X.; resources, R.A.F.; data curation, J.J.G. and P.X.; writing—original draft preparation, J.J.G., P.X. and R.A.F.; writing—review and editing, P.X., K.A.S., W.Y., R.M.C. and P.A.J.; visualization, J.J.G. and P.X.; supervision, R.A.F.; project administration, R.A.F.; funding acquisition, R.A.F. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Heart, Lung, and Blood Institute (NHLBI) P01 HL074940. Carey is Principal Investigator of R01 HL128189 and Project Director for Program Project Grant (P01 HL074940). Jose is Principal Investigator of R01 DK039308 and R01 DK119652 and Project Director of P01 HL074940.

Institutional Review Board Statement

The study was conducted in accordance with the Decla-ration of Helsinki, and approved by the Institutional Review Board of University of Virginia (protocol code HSR# 13310, 8/24/23 and HSR # 11494, 17 June 2021).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

Not applicable.

Acknowledgments

We thank Stephen Marshall, Chase Carson, and Qing Zhang for their support and helpful discussions during the performance of these studies.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

Ang I angiotensin I
Ang II angiotensin II
Ang III angiotensin III
Ang IV angiotensin IV
APN amino peptidase N
AT1R angiotensin II type 1 receptor
AT2R angiotensin II type 2 receptor
BP blood pressure
D2R dopamine type 2 receptor
DRD2dopamine type 2 receptor gene
GRK4 G protein-coupled receptor kinase type 4
ISS inverse salt sensitivity
MAP mean arterial pressure
RAS renin-angiotensin system
RPTC renal proximal tubule cell
SR Salt resistance
SS Salt sensitivity
uRPTC urine-derived renal proximal tubule cell

References

  1. Whelton, P.K.; Carey, R.M.; Aronow, W.S.; Casey, D.E.; Collins, K.J.; Dennison Himmelfarb, C.; DePalma, S.M.; Gidding, S.; Jamerson, K.A.; Jones, D.W.; et al. 2017 ACC/AHA/AAPA/ABC/ACPM/AGS/APHA/ASH/ASPC/ NMA/PCNA guideline for the prevention, detection, evaluation, and management of high blood pressure in adults: A report of the AMERICAN COLLEGE OF CARDIOLOGY/AMERICAN HEART ASSOCIATION task force on clinical practice guidelines. Hypertension 2018, 71, e13–e115. [Google Scholar] [PubMed]
  2. He, F.J.; Tan, M.; Ma, Y.; MacGregor, G.A. Salt reduction to prevent hypertension and cardiovascular disease: JACC state-of-the-art review. J. Am. Coll. Cardiol. 2020, 75, 632–647. [Google Scholar] [CrossRef] [PubMed]
  3. Cappuccio, F.P.; Beer, M.; Strazzullo, P.; Network, E.S.A. Population dietary salt reduction and the risk of cardiovascular disease. A scientific statement from the European Salt Action Network. Nutr. Metab. Cardiovasc. Dis. 2018, 29, 107–114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Carey, R.M.; Schoeffel, C.D.; Gildea, J.J.; Jones, J.E.; McGrath, H.E.; Gordon, L.N.; Park, M.J.; Sobota, R.S.; Underwood, P.C.; Williams, J.; et al. Salt sensitivity of blood pressure is associated with polymorphisms in the sodium-bicarbonate cotransporter. Hypertension 2012, 60, 1359–1366. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Elijovich, F.; Weinberger, M.H.; Anderson, C.A.; Appel, L.J.; Bursztyn, M.; Cook, N.R.; Dart, R.A.; Newton-Cheh, C.H.; Sacks, F.M.; Laffer, C.L. Hypertension AHAPaPECotCo, Biology CoFGaT, Council aS. Salt sensitivity of blood pressure: A scientific statement from the American Heart Association. Hypertension 2016, 68, e7–e46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Rucker, A.J.; Rudemiller, N.P.; Crowley, S.D. Salt, hypertension, and immunity. Annu. Rev. Physiol. 2018, 80, 283–307. [Google Scholar] [CrossRef] [PubMed]
  7. Lastra, G.; Dhuper, S.; Johnson, M.S.; Sowers, J.R. Salt, aldosterone, and insulin resistance: Impact on the cardiovascular system. Nat. Rev. Cardiol. 2010, 7, 577–584. [Google Scholar] [CrossRef]
  8. Kurtz, T.W.; DiCarlo, S.E.; Pravenec, M.; Morris, R.C. The American Heart Association scientific statement on salt sensitivity of blood pressure: Prompting consideration of alternative conceptual frameworks for the pathogenesis of salt sensitivity? J. Hypertens. 2017, 35, 2214–2225. [Google Scholar] [CrossRef]
  9. Skrabal, F.; Herholz, H.; Neumayr, M.; Hamberger, L.; Ledochowski, M.; Sporer, H.; Hortnagl, H.; Schwarz, S.; Schonitzer, D. Salt sensitivity in humans is linked to enhanced sympathetic responsiveness and to enhanced proximal tubular reabsorption. Hypertension 1984, 6, 152–158. [Google Scholar] [CrossRef] [Green Version]
  10. Majid, D.S.; Kopkan, L. Nitric oxide and superoxide interactions in the kidney and their implication in the development of salt-sensitive hypertension. Clin. Exp. Pharmacol. Physiol. 2007, 34, 946–952. [Google Scholar] [CrossRef]
  11. Sigmund, C.D.; Carey, R.M.; Appel, L.J.; Arnett, D.K.; Bosworth, H.B.; Cushman, W.C.; Galis, Z.S.; Green Parker, M.; Hall, J.E.; Harrison, D.G.; et al. Report of the National Heart, Lung, and Blood Institute working group on hypertension: Barriers to translation. Hypertension 2020, 75, 902–917. [Google Scholar] [CrossRef] [PubMed]
  12. Raizada, M.K.; Joe, B.; Bryan, N.S.; Chang, E.B.; Dewhirst, F.E.; Borisy, G.G.; Galis, Z.S.; Henderson, W.; Jose, P.A.; Ketchum, C.J.; et al. Report of the National Heart, Lung, and Blood Institute working group on the role of microbiota in blood pressure regulation: Current status and future directions. Hypertension 2017, 70, 479–485. [Google Scholar] [CrossRef] [PubMed]
  13. Weinberger, M.H.; Fineberg, N.S.; Fineberg, S.E.; Weinberger, M. Salt sensitivity, pulse pressure, and death in normal and hypertensive humans. Hypertension 2001, 37, 429–432. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Alderman, M.H.; Cohen, H.W. Dietary sodium intake and cardiovascular mortality: Controversy resolved? Curr. Hypertens. Rep. 2012, 14, 193–201. [Google Scholar] [CrossRef] [PubMed]
  15. O’Donnell, M.; Mente, A.; Alderman, M.H.; Brady, A.J.B.; Diaz, R.; Gupta, R.; López-Jaramillo, P.; Luft, F.C.; Lüscher, T.F.; Mancia, G.; et al. Salt and cardiovascular disease: Insufficient evidence to recommend low sodium intake. Eur. Heart. J. 2020, 41, 3363–3373. [Google Scholar] [CrossRef] [PubMed]
  16. Asayama, K.; Stolarz-Skrzypek, K.; Persu, A.; Staessen, J.A. Systematic review of health outcomes in relation to salt intake highlights the widening divide between guidelines and the evidence. Am. J. Hypertens. 2014, 27, 1138–1142. [Google Scholar] [CrossRef] [Green Version]
  17. Stolarz-Skrzypek, K.; Kuznetsova, T.; Thijs, L.; Tikhonoff, V.; Seidlerová, J.; Richart, T.; Jin, Y.; Olszanecka, A.; Malyutina, S.; Casiglia, E.; et al. Fatal and nonfatal outcomes, incidence of hypertension, and blood pressure changes in relation to urinary sodium excretion. JAMA 2011, 305, 1777–1785. [Google Scholar] [CrossRef] [Green Version]
  18. O’Donnell, M.; Mente, A.; Rangarajan, S.; McQueen, M.J.; Wang, X.; Liu, L.; Yan, H.; Lee, S.F.; Mony, P.; Devanath, A.; et al. Urinary sodium and potassium excretion, mortality, and cardiovascular events. N. Engl. J. Med. 2014, 371, 612–623. [Google Scholar] [CrossRef] [Green Version]
  19. Strom, B.L.; Anderson, C.A.; Ix, J.H. Sodium reduction in populations: Insights from the Institute of Medicine committee. JAMA 2013, 310, 31–32. [Google Scholar] [CrossRef]
  20. Gildea, J.J.; Lahiff, D.T.; Van Sciver, R.E.; Weiss, R.S.; Shah, N.; McGrath, H.E.; Schoeffel, C.D.; Jose, P.A.; Carey, R.M.; Felder, R.A. A linear relationship between the ex-vivo sodium mediated expression of two sodium regulatory pathways as a surrogate marker of salt sensitivity of blood pressure in exfoliated human renal proximal tubule cells: The virtual renal biopsy. Clin. Chim. Acta 2013, 421, 236–242. [Google Scholar] [CrossRef]
  21. Graudal, N.; Jürgens, G. Conflicting evidence on health effects associated with salt reduction calls for a redesign of the salt dietary guidelines. Prog. Cardiovasc. Dis. 2018, 61, 20–26. [Google Scholar] [CrossRef] [PubMed]
  22. Alderman, M.H.; Blumenfeld, J.D. Hypertension: Evolving from standardized to individualized care. J. Hypertens. 2020, 38, 1251–1254. [Google Scholar] [CrossRef] [PubMed]
  23. Montasser, M.E.; Douglas, J.A.; Roy-Gagnon, M.H.; Van Hout, C.V.; Weir, M.R.; Vogel, R.; Parsa, A.; Steinle, N.I.; Snitker, S.; Brereton, N.H.; et al. Determinants of blood pressure response to low-salt intake in a healthy adult population. J. Clin. Hypertens. 2011, 13, 795–800. [Google Scholar] [CrossRef] [Green Version]
  24. Overlack, A.; Ruppert, M.; Kolloch, R.; Göbel, B.; Kraft, K.; Diehl, J.; Schmitt, W.; Stumpe, K.O. Divergent hemodynamic and hormonal responses to varying salt intake in normotensive subjects. Hypertension 1993, 22, 331–338. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Overlack, A.; Ruppert, M.; Kolloch, R.; Kraft, K.; Stumpe, K.O. Age is a major determinant of the divergent blood pressure responses to varying salt intake in essential hypertension. Am. J. Hypertens. 1995, 8, 829–836. [Google Scholar] [CrossRef]
  26. Cuka, E.; Simonini, M.; Lanzani, C.; Zagato, L.; Citterio, L.; Messaggio, E.; Faienza, S.; Brioni, E.; Hamlyn, J.M.; Manunta, P. Inverse salt sensitivity: An independent risk factor for cardiovascular damage in essential hypertension. J. Hypertens. 2022, 40, 1504–1512. [Google Scholar] [CrossRef] [PubMed]
  27. Bihorac, A.; Tezcan, H.; Ozener, C.; Oktay, A.; Akoglu, E. Association between salt sensitivity and target organ damage in essential hypertension. Am. J. Hypertens. 2000, 13, 864–872. [Google Scholar] [CrossRef] [Green Version]
  28. Felder, R.A.; White, M.J.; Williams, S.M.; Jose, P.A. Diagnostic tools for hypertension and salt sensitivity testing. Curr. Opin. Nephrol. Hypertens. 2013, 22, 65–76. [Google Scholar] [CrossRef] [Green Version]
  29. Iatrino, R.; Manunta, P.; Zagato, L. Salt Sensitivity: Challenging and Controversial Phenotype of Primary Hypertension. Curr. Hypertens. Rep. 2016, 18, 70. [Google Scholar] [CrossRef]
  30. Institute of Medicine (US) Committee on Strategies to Reduce Sodium Intake. Strategies to Reduce Sodium Intake in the United States; Henney, J.E., Taylor, C.L., Boon, C.S., Eds.; National Academies Press: Washington, DC, USA, 2010.
  31. O’Donnell, M.; Mente, A.; Rangarajan, S.; McQueen, M.J.; O’Leary, N.; Yin, L.; Liu, X.; Swaminathan, S.; Khatib, R.; Rosengren, A.; et al. Joint association of urinary sodium and potassium excretion with cardiovascular events and mortality: Prospective cohort study. BMJ 2019, 364, l772. [Google Scholar] [CrossRef]
  32. Castiglioni, P.; Parati, G.; Lazzeroni, D.; Bini, M.; Faini, A.; Brambilla, L.; Brambilla, V.; Coruzzi, P. Hemodynamic and autonomic response to different salt intakes in normotensive individuals. J. Am. Heart. Assoc. 2016, 5, e003736. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Gildea, J.J.; Carlson, J.M.; Schoeffel, C.D.; Carey, R.M.; Felder, R.A. Urinary exosome miRNome analysis and its applications to salt sensitivity of blood pressure. Clin. Biochem. 2013, 46, 1131–1134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Barros, E.R.; Carvajal, C.A. Urinary Exosomes and Their Cargo: Potential Biomarkers for Mineralocorticoid Arterial Hypertension? Front. Endocrinol. 2017, 8, 23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Tan, P.P.S.; Hall, D.; Chilian, W.M.; Chia, Y.C.; Mohd Zain, S.; Lim, H.M.; Kumar, D.N.; Ching, S.M.; Low, T.Y.; Md Noh, M.F.; et al. Exosomal microRNAs in the development of essential hypertension and its potential as biomarkers. Am. J. Physiol. Heart Circ. Physiol. 2021, 320, H1486–H1497. [Google Scholar] [CrossRef] [PubMed]
  36. Manosroi, W.; Williams, G.H. Genetics of human primary hypertension: Focus on hormonal mechanisms. Endocr. Rev. 2019, 40, 825–856. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Rayner, B.; Ramesar, R. The importance of G protein-coupled receptor kinase 4 (GRK4) in pathogenesis of salt sensitivity, salt sensitive hypertension and response to antihypertensive treatment. Int. J. Mol. Sci. 2015, 16, 5741–5749. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Jeong, S.; Kim, J.Y.; Cho, Y.; Koh, S.B.; Kim, N.; Choi, J.R. Genetically, dietary sodium intake is causally associated with salt-sensitive hypertension risk in a community-based cohort study: A mendelian randomization approach. Curr. Hypertens. Rep. 2020, 22, 45. [Google Scholar] [CrossRef]
  39. Lee, M.; Kim, M.K.; Kim, S.M.; Park, H.; Park, C.G.; Park, H.K. Gender-based differences on the association between salt-sensitive genes and obesity in Korean children aged between 8 and 9 years. PLoS ONE 2015, 10, e0120111. [Google Scholar] [CrossRef] [Green Version]
  40. Sanada, H.; Yatabe, J.; Midorikawa, S.; Hashimoto, S.; Watanabe, T.; Moore, J.H.; Ritchie, M.D.; Williams, S.M.; Pezzullo, J.C.; Sasaki, M.; et al. Single nucleotide polymorphisms for diagnosis of salt-sensitive hypertension. Clin. Chem. 2006, 52, 352–360. [Google Scholar] [CrossRef] [Green Version]
  41. Bengra, C.; Mifflin, T.E.; Khripin, Y.; Manunta, P.; Williams, S.M.; Jose, P.A.; Felder, R.A. Genotyping of essential hypertension single-nucleotide polymorphisms by a homogeneous PCR method with universal energy transfer primers. Clin. Chem. 2002, 48, 2131–2140. [Google Scholar] [CrossRef] [Green Version]
  42. Wang, Z.; Zeng, C.; Villar, V.A.; Chen, S.Y.; Konkalmatt, P.; Wang, X.; Asico, L.D.; Jones, J.E.; Yang, Y.; Sanada, H.; et al. Human GRK4γ142V Variant Promotes Angiotensin II Type I Receptor-Mediated Hypertension via Renal Histone Deacetylase Type 1 Inhibition. Hypertension 2016, 67, 325–334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Diao, Z.; Asico, L.D.; Villar, V.A.M.; Zheng, X.; Cuevas, S.; Armando, I.; Jose, P.A.; Wang, X. Increased renal oxidative stress in salt-sensitive human GRK4gamma486V transgenic mice. Free Radic. Biol. Med. 2017, 106, 80–90. [Google Scholar] [CrossRef] [Green Version]
  44. Armando, I.; Konkalmatt, P.; Felder, R.A.; Jose, P.A. The renal dopaminergic system: Novel diagnostic and therapeutic approaches in hypertension and kidney disease. Transl. Res. 2015, 165, 505–511. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Harris, R.C.; Zhang, M.Z. Dopamine, the kidney, and hypertension. Curr. Hypertens. Rep. 2012, 14, 138–143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Padia, S.H.; Kemp, B.A.; Howell, N.L.; Fournie-Zaluski, M.C.; Roques, B.P.; Carey, R.M. Conversion of renal angiotensin II to angiotensin III is critical for AT2 receptor-mediated natriuresis in rats. Hypertension 2008, 51, 460–465. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Felder, R.A.; Gildea, J.J.; Xu, P.; Yue, W.; Armando, I.; Carey, R.M.; Jose, P.A. Inverse Salt Sensitivity of Blood Pressure: Mechanisms and Potential Relevance for Prevention of Cardiovascular Disease. Curr. Hypertens. Rep. 2022, 9, 361–374. [Google Scholar] [CrossRef]
  48. Li, X.; Campbell, D.; Ohishi, M.; Yuan, S.; Zhuo, J. AT1 receptor-activated signaling mediates angiotensin IV-induced renal cortical vasoconstriction in rats. Am. J. Physiol. Renal. Physiol. 2006, 290, F1024–F1033. [Google Scholar] [CrossRef] [Green Version]
  49. Danziger, R.S. Aminopeptidase N in arterial hypertension. Heart Fail. Rev. 2008, 13, 293–298. [Google Scholar] [CrossRef]
  50. Padia, S.H.; Howell, N.L.; Kemp, B.A.; Fournie-Zaluski, M.C.; Roques, B.P.; Carey, R.M. Intrarenal aminopeptidase N inhibition restores defective angiontesin II type 2-mediated natriuresis in spontaneously hypertensive rats. Hypertension 2010, 55, 474–480. [Google Scholar] [CrossRef] [Green Version]
  51. Kemp, B.A.; Howell, N.L.; Gildea, J.J.; Keller, S.R.; Carey, R.M. Identification of a Primary Renal AT2 Receptor Defect in Spontaneously Hypertensive Rats. Circ. Res. 2020, 126, 644–659. [Google Scholar] [CrossRef]
  52. Lew, R.A.; Mustafa, T.; Ye, S.; McDowall, S.G.; Chai, S.Y.; Albiston, A.L. Angiotensin AT4 ligands are potent, competitive inhibitors of insulin regulated aminopeptidase (IRAP). J. Neurochem. 2003, 86, 344–350. [Google Scholar] [CrossRef] [PubMed]
  53. Walton, S.L.; Mirabito Colafella, K.M.; Ansari, A.; Chai, S.Y.; Denton, K.M. Insulin-regulated aminopeptidase deficiency impairs cardiovascular adaptations and placental development during pregnancy. Clin. Sci. 2020, 134, 3213–3228. [Google Scholar] [CrossRef] [PubMed]
  54. Hussain, T.; Abdul-Wahab, R.; Kotak, D.K.; Lokhandwala, M.F. Bromocriptine regulates angiotensin II response on sodium pump in proximal tubules. Hypertension 1998, 32, 1054–1059. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Sarkar, C.; Ganju, R.K.; Pompili, V.J.; Chakroborty, D. Enhanced peripheral dopamine impairs post-ischemic healing by suppressing angiotensin receptor type 1 expression in endothelial cells and inhibiting angiogenesis. Angiogenesis 2017, 20, 97–107. [Google Scholar] [CrossRef] [PubMed]
  56. Choi, M.R.; Kouyoumdzian, N.M.; Rukavina Mikusic, N.L.; Kravetz, M.C.; Rosón, M.I.; Rodríguez Fermepin, M.; Fernández, B.E. Renal dopaminergic system: Pathophysiological implications and clinical perspectives. World J. Nephrol. 2015, 4, 196–212. [Google Scholar] [CrossRef] [PubMed]
  57. Jiang, X.; Konkalmatt, P.; Yang, Y.; Gildea, J.J.; Jones, J.E.; Cuevas, S.; Felder, R.A.; Jose, P.A.; Armando, I. Single-nucleotide polymorphisms of the dopamine D2 receptor increase inflammation and fibrosis in human renal proximal tubule cells. Hypertension 2014, 63, e74–e80. [Google Scholar] [CrossRef] [PubMed]
  58. Konkalmatt, P.R.; Asico, L.D.; Zhang, Y.; Yang, Y.; Drachenberg, C.; Zheng, X.; Han, F.; Jose, P.A.; Armando, I. Renal rescue of dopamine D2 receptor function reverses renal injury and high blood pressure. JCI Insight 2016, 1, e85888. [Google Scholar] [CrossRef]
  59. Zhang, Y.; Cuevas, S.; Asico, L.D.; Escano, C.; Yang, Y.; Pascua, A.M.; Wang, X.; Jones, J.E.; Grandy, D.; Eisner, G.; et al. Deficient dopamine D2 receptor function causes renal inflammation independently of high blood pressure. PLoS ONE 2012, 7, e38745. [Google Scholar] [CrossRef] [Green Version]
  60. Zhang, Y.; Jiang, X.; Qin, C.; Cuevas, S.; Jose, P.A.; Armando, I. Dopamine D2 receptors’ effects on renal inflammation are mediated by regulation of PP2A function. Am. J. Physiol. Renal. Physiol. 2016, 310, F128–F134. [Google Scholar] [CrossRef] [Green Version]
  61. Barrese, V.; Stott, J.B.; Baldwin, S.N.; Mondejar-Parreño, G.; Greenwood, I.A. SMIT (Sodium-Myo-Inositol Transporter) 1 Regulates Arterial Contractility Through the Modulation of Vascular Kv7 Channels. Arterioscler. Thromb. Vasc. Biol. 2020, 40, 2468–2480. [Google Scholar] [CrossRef]
  62. Lahjouji, K.; Aouameur, R.; Bissonnette, P.; Coady, M.J.; Bichet, D.G.; Lapointe, J.Y. Expression and functionality of the Na+/myo-inositol cotransporter SMIT2 in rabbit kidney. Biochim. Biophys. Acta 2007, 1768, 1154–1159. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Gildea, J.J.; McGrath, H.E.; Van Sciver, R.E.; Wang, D.B.; Felder, R.A. Isolation, growth, and characterization of human renal epithelial cells using traditional and 3D methods. Methods Mol. Biol. 2013, 945, 329–345. [Google Scholar] [PubMed]
  64. Gildea, J.J.; Xu, P.; Kemp, B.A.; Carey, R.M.; Jose, P.A.; Felder, R.A. The dopamine D1 receptor and angiotensin II type-2 receptor are required for inhibition of sodium transport through a protein phosphatase 2A pathway. Hypertension 2019, 73, 1258–1265. [Google Scholar] [CrossRef]
  65. Rosić, N.K.; Standaert, M.L.; Pollet, R.J. The mechanism of insulin stimulation of (Na+,K+)-ATPase transport activity in muscle. J. Biol. Chem. 1985, 260, 6206–6212. [Google Scholar] [CrossRef]
  66. Stojanovic, T.; Orlova, M.; Sialana, F.J.; Höger, H.; Stuchlik, S.; Milenkovic, I.; Aradska, J.; Lubec, G. Validation of dopamine receptor Drd1 and Drd2 antibodies using receptor deficient mice. Amino Acids 2017, 49, 1101–1109. [Google Scholar] [CrossRef] [PubMed]
  67. Newton, C.L.; Wood, M.D.; Strange, P.G. Examining the Effects of Sodium Ions on the Binding of Antagonists to Dopamine D2 and D3 Receptors. PLoS ONE 2016, 11, e0158808. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Seeman, P.; Van Tol, H.H. Dopamine receptor pharmacology. Curr. Opin. Neurol. Neurosurg. 1993, 6, 602–608. [Google Scholar] [CrossRef]
  69. 741742 L. Guide to Pharmacology. 2021. Available online: https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=3302 (accessed on 13 October 2021).
  70. Kling-Petersen, T.; Ljung, E.; Svensson, K. Effects on locomotor activity after local application of D3 preferring compounds in discrete areas of the rat brain. J. Neural. Transm. Gen. Sect. 1995, 102, 209–220. [Google Scholar] [CrossRef]
  71. Gao, D.Q.; Canessa, L.M.; Mouradian, M.M.; Jose, P.A. Expression of the D2 subfamily of dopamine receptor genes in kidney. Am. J. Physiol. 1994, 266, F646–F650. [Google Scholar] [CrossRef]
  72. Gildea, J.J.; Xu, P.; Kemp, B.A.; Carlson, J.M.; Tran, H.T.; Bigler Wang, D.; Langouët-Astrié, C.J.; McGrath, H.E.; Carey, R.M.; Jose, P.A.; et al. Sodium bicarbonate cotransporter NBCe2 gene variants increase sodium and bicarbonate transport in human renal proximal tubule cells. PLoS ONE 2018, 13, e0189464. [Google Scholar] [CrossRef]
  73. Diarra, A.; Sheldon, C.; Church, J. In situ calibration and [H+] sensitivity of the fluorescent Na+ indicator SBFI. Am. J. Physiol. Cell Physiol. 2001, 280, C1623–C1633. [Google Scholar] [CrossRef] [PubMed]
  74. Crocco, P.; Montesanto, A.; Passarino, G.; Rose, G. Polymorphisms falling within putative miRNA target sites in the 3’UTR region of SIRT2 and DRD2 genes are correlated with human longevity. J. Gerontol. A Biol. Sci. Med. Sci. 2016, 71, 586–592. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Hauberg, M.E.; Roussos, P.; Grove, J.; Børglum, A.D.; Mattheisen, M.; Schizophrenia Working Group of the Psychiatric Genomics Consortium. Analyzing the role of microRNAs in schizophrenia in the context of common genetic risk variants. JAMA Psychiatry 2016, 73, 369–377. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Maffioletti, E.; Tardito, D.; Gennarelli, M.; Bocchio-Chiavetto, L. Micro spies from the brain to the periphery: New clues from studies on micrornas in neuropsychiatric disorders. Front. Cell. Neurosci. 2014, 8, 75. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Xu, P.; Gildea, J.J.; Zhang, C.; Konkalmatt, P.; Cuevas, S.; Bigler Wang, D.; Tran, H.T.; Jose, P.A.; Felder, R.A. Stomach gastrin is regulated by sodium via PPAR-α and dopamine D1 receptor. J. Mol. Endocrinol. 2020, 64, 53–65. [Google Scholar] [CrossRef]
  78. Luo, Y.; Hitz, B.C.; Gabdank, I.; Hilton, J.A.; Kagda, M.S.; Lam, B.; Myers, Z.; Sud, P.; Jou, J.; Lin, K.; et al. New developments on the Encyclopedia of DNA Elements (ENCODE) data portal. Nucleic Acids Res. 2020, 48, D882–D889. [Google Scholar] [CrossRef]
  79. Yang, Y.; Cuevas, S.; Yang, S.; Villar, V.A.; Escano, C.; Asico, L.; Yu, P.; Jiang, X.; Weinman, E.J.; Armando, I.; et al. Sestrin2 decreases renal oxidative stress, lowers blood pressure, and mediates dopamine D2 receptor-induced inhibition of reactive oxygen species production. Hypertension 2014, 64, 825–832. [Google Scholar] [CrossRef] [Green Version]
  80. Cuevas, S.; Yang, Y.; Konkalmatt, P.; Asico, L.D.; Feranil, J.; Jones, J.; Villar, V.A.; Armando, I.; Jose, P.A. Role of nuclear factor erythroid 2-related factor 2 in the oxidative stress-dependent hypertension associated with the depletion of DJ-1. Hypertension 2015, 65, 1251–1257. [Google Scholar] [CrossRef]
  81. Ryoo, I.G.; Kwak, M.K. Regulatory crosstalk between the oxidative stress-related transcription factor Nfe2l2/Nrf2 and mitochondria. Toxicol. Appl. Pharmacol. 2018, 359, 24–33. [Google Scholar] [CrossRef]
  82. Padia, S.H.; Kemp, B.A.; Howell, N.L.; Gildea, J.J.; Keller, S.R.; Carey, R.M. Intrarenal angiotensin III infusion induces natriuresis and angiotensin type 2 receptor translocation in Wistar-Kyoto but not in spontaneously hypertensive rats. Hypertension 2009, 53, 338–343. [Google Scholar] [CrossRef]
  83. Guirguis-Blake, J.M.; Evans, C.V.; Webber, E.M.; Coppola, E.L.; Perdue, L.A.; Weyrich, M.S. Screening for Hypertension in adults: Updated Evidence Report and Systematic Review for the US Preventive Services Task Force; Report No.: 20–05265-EF-1; Agency for Healthcare Research and Quality: Rockville, MD, USA, 2021.
  84. MacFadyen, R.J.; Lees, K.R.; Reid, J.L. Responses to low dose intravenous perindoprilat infusion in salt deplete/salt replete normotensive volunteers. Br. J. Clin. Pharmacol. 1994, 38, 329–334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Dimsdale, J.E.; Ziegler, M.; Mills, P.; Berry, C. Prediction of salt sensitivity. Am. J. Hypertens. 1990, 3, 429–435. [Google Scholar] [CrossRef] [PubMed]
  86. Shin, Y.; Kumar, U.; Patel, Y.; Patel, S.C.; Sidhu, A. Differential expression of D2-like dopamine receptors in the kidney of the spontaneously hypertensive rat. J. Hypertens. 2003, 21, 199–207. [Google Scholar] [CrossRef] [PubMed]
  87. Fang, Y.J.; Thomas, G.N.; Xu, Z.L.; Fang, J.Q.; Critchley, J.A.; Tomlinson, B. An affected pedigree member analysis of linkage between the dopamine D2 receptor gene Taq1 polymorphism and obesity and hypertension. Int. J. Cardiol. 2005, 102, 111–116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Thompson, J.; Thomas, N.; Singleton, A.; Piggott, M.; Lloyd, S.; Perry, E.K.; Morris, C.M.; Perry, R.H.; Ferrier, I.N.; Court, J.A. D2 dopamine receptor gene (DRD2) Taq1 a polymorphism: Reduced dopamine D2 receptor binding in the human striatum associated with the A1 allele. Pharmacogenetics 1997, 7, 479–484. [Google Scholar] [CrossRef]
  89. Qaddumi, W.N.; Jose, P.A. The role of the renal dopaminergic system and oxidative stress in the pathogenesis of hypertension. Biomedicines 2021, 9, 139. [Google Scholar] [CrossRef]
  90. Armando, I.; Asico, L.D.; Wang, X.; Jones, J.E.; Serrão, M.P.; Cuevas, S.; Grandy, D.K.; Soares-da-Silva, P.; Jose, P.A. Antihypertensive effect of etamicastat in dopamine D2 receptor-deficient mice. Hypertens. Res. 2018, 41, 489–498. [Google Scholar] [CrossRef]
  91. Han, F.; Konkalmatt, P.; Mokashi, C.; Kumar, M.; Zhang, Y.; Ko, A.; Farino, Z.J.; Asico, L.D.; Xu, G.; Gildea, J.; et al. Dopamine D2 receptor modulates Wnt expression and control of cell proliferation. Sci. Rep. 2019, 9, 16861. [Google Scholar] [CrossRef] [Green Version]
  92. Han, F.; Konkalmatt, P.; Chen, J.; Gildea, J.; Felder, R.A.; Jose, P.A.; Armando, I. miR-217 mediates the protective effects of the dopamine D2 receptor on fibrosis in human renal proximal tubule cells. Hypertension 2015, 65, 1118–1125. [Google Scholar] [CrossRef] [Green Version]
  93. Armando, I.; Villar, V.A.; Jose, P.A. Dopamine and renal function and blood pressure regulation. Compr. Physiol. 2011, 1, 1075–1117. [Google Scholar]
  94. Zhang, M.Z.; Yao, B.; Fang, X.; Wang, S.; Smith, J.P.; Harris, R.C. Intrarenal dopaminergic system regulates renin expression. Hypertension 2009, 53, 564–570. [Google Scholar] [CrossRef] [PubMed]
  95. Bromocriptine. Guide to Pharmacology. 2021. Available online: https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=35 (accessed on 13 October 2021).
  96. Kemp, B.A.; Howell, N.L.; Keller, S.R.; Gildea, J.J.; Shao, W.; Navar, L.G.; Carey, R.M. Defective renal angiotensin III and AT receptor signaling in prehypertensive spontaneously hypertensive rats. J. Am. Heart Assoc. 2019, 8, e012016. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Kotlo, K.; Shukla, S.; Tawar, U.; Skidgel, R.A.; Danziger, R.S. Aminopeptidase N reduces basolateral Na+ -K+ -ATPase in proximal tubule cells. Am. J. Physiol. Renal. Physiol. 2007, 293, F1047–F1053. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Yang, R.; Smolders, I.; Dupont, A.G. Blood pressure and renal hemodynamic effects of angiotensin fragments. Hypertens. Res. 2011, 34, 674–683. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Handa, R.K. Biphasic actions of angiotensin IV on renal blood flow in the rat. Regul. Pept. 2006, 136, 23–29. [Google Scholar] [CrossRef] [PubMed]
  100. Cuevas, S.; Villar, V.A.; Jose, P.A.; Armando, I. Renal dopamine receptors, oxidative stress, and hypertension. Int. J. Mol. Sci. 2013, 14, 17553–17572. [Google Scholar] [CrossRef] [Green Version]
  101. Rukavina Mikusic, N.L.; Silva, M.G.; Mazzitelli, L.R.; Santos, R.A.S.; Gómez, K.A.; Grecco, H.E.; Gironacci, M.M. Interaction between the angiotensin-(1–7) mas receptor and the dopamine D2 receptor: Implications in inflammation. Hypertension 2021, 77, 1659–1669. [Google Scholar] [CrossRef]
  102. Han, X.; Li, B.; Ye, X.; Mulatibieke, T.; Wu, J.; Dai, J.; Wu, D.; Ni, J.; Zhang, R.; Xue, J.; et al. Dopamine D2 receptor signalling controls inflammation in acute pancreatitis via a PP2A-dependent Akt/NF-κB signalling pathway. Br. J. Pharmacol. 2017, 174, 4751–4770. [Google Scholar] [CrossRef] [Green Version]
  103. Udali, S.; Guarini, P.; Moruzzi, S.; Choi, S.W.; Friso, S. Cardiovascular epigenetics: From DNA methylation to microRNAs. Mol. Asp. Med. 2013, 34, 883–901. [Google Scholar] [CrossRef]
  104. Chen, L.J.; Wei, S.Y.; Chiu, J.J. Mechanical regulation of epigenetics in vascular biology and pathobiology. J. Cell Mol. Med. 2013, 17, 437–448. [Google Scholar] [CrossRef]
  105. Findeisen, H.M.; Kahles, F.K.; Bruemmer, D. Epigenetic regulation of vascular smooth muscle cell function in atherosclerosis. Curr. Atheroscler. Rep. 2013, 15, 319. [Google Scholar] [CrossRef]
  106. Wang, J.; Yin, N.; Deng, Y.; Wei, Y.; Huang, Y.; Pu, X.; Li, L.; Zheng, Y.; Guo, J.; Yu, J.; et al. Ascorbic acid protects against hypertension through downregulation of ACE1 gene expression mediated by histone deacetylation in prenatal inflammation-induced offspring. Sci. Rep. 2016, 6, 39469. [Google Scholar] [CrossRef] [PubMed]
  107. Schwenk, R.W.; Vogel, H.; Schürmann, A. Genetic and epigenetic control of metabolic health. Mol. Metab. 2013, 2, 337–347. [Google Scholar] [CrossRef] [PubMed]
  108. Tammen, S.A.; Friso, S.; Choi, S.W. Epigenetics: The link between nature and nurture. Mol. Asp. Med. 2013, 34, 753–764. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Banday, A.A.; Fazili, F.R.; Lokhandwala, M.F. Oxidative stress causes renal dopamine D1 receptor dysfunction and hypertension via mechanisms that involve nuclear factor-kappab and protein kinase C. J. Am. Soc. Nephrol. 2007, 18, 1446–1457. [Google Scholar] [CrossRef] [Green Version]
  110. Kawarazaki, W.; Fujita, T. Kidney and epigenetic mechanisms of salt-sensitive hypertension. Nat. Rev. Nephrol. 2021, 17, 350–363. [Google Scholar] [CrossRef]
  111. Cuevas, S.; Villar, V.A.M.; Jose, P.A. Genetic polymorphisms associated with reactive oxygen species and blood pressure regulation. Pharm. J. 2019, 19, 315–336. [Google Scholar] [CrossRef]
  112. Lee, H.; Jiang, X.; Perwaiz, I.; Yu, P.; Wang, J.; Wang, Y.; Hüttemann, M.; Felder, R.A.; Sibley, D.R.; Polster, B.M.; et al. Dopamine D(5) receptor-mediated decreases in mitochondrial reactive oxygen species production are cAMP and autophagy dependent. Hypertens. Res. 2021, 44, 628–641. [Google Scholar] [CrossRef]
  113. Gildea, J.J.; Tran, H.T.; Van Sciver, R.E.; Bigler Wang, D.; Carlson, J.M.; Felder, R.A. A novel role for c-Myc in G protein-coupled receptor kinase 4 (GRK4) transcriptional regulation in human kidney proximal tubule cells. Hypertension 2013, 61, 1021–1027. [Google Scholar] [CrossRef] [Green Version]
  114. Filippova, G.N.; Fagerlie, S.; Klenova, E.M.; Myers, C.; Dehner, Y.; Goodwin, G.; Neiman, P.E.; Collins, S.J.; Lobanenkov, V.V. An exceptionally conserved transcriptional repressor, CTCF, employs different combinations of zinc fingers to bind diverged promoter sequences of avian and mammalian c-myc oncogenes. Mol. Cell. Biol. 1996, 16, 2802–2813. [Google Scholar] [CrossRef] [Green Version]
  115. Wu, M.; Liang, G.; Duan, H.; Yang, X.; Qin, G.; Sang, N. Synergistic effects of sulfur dioxide and polycyclic aromatic hydrocarbons on pulmonary pro-fibrosis via mir-30c-1–3p/ transforming growth factor β type ii receptor axis. Chemosphere 2019, 219, 268–276. [Google Scholar] [CrossRef] [PubMed]
  116. Ji, W.; Foo, J.N.; O’Roak, B.J.; Zhao, H.; Larson, M.G.; Simon, D.B.; Newton-Cheh, C.; State, M.W.; Levy, D.; Lifton, R.P. Rare independent mutations in renal salt handling genes contribute to blood pressure variation. Nat. Genet. 2008, 40, 592–599. [Google Scholar] [CrossRef] [Green Version]
  117. Xu, P.; Sudarikova, A.V.; Ilatovskaya, D.V.; Gildea, J.J.; Akhter, M.; Carey, R.M.; Yue, W.; Jose, P.A.; Felder, R.A. Epithelial Sodium Channel Alpha Subunit (αENaC) Is Associated with Inverse Salt Sensitivity of Blood Pressure. Biomedicines 2022, 10, 981. [Google Scholar] [CrossRef] [PubMed]
  118. Horniblow, R.D.; Pathak, P.; Balacco, D.L.; Acharjee, A.; Lles, E.; Gkoutos, G.; Beggs, A.D.; Tselepis, C. Iron-mediated epigenetic activation of NRF2 targets. J. Nutr. Biochem. 2022, 101, 108929. [Google Scholar] [CrossRef] [PubMed]
  119. Dhillon, P.; Park, J.; Hurtado Del Pozo, C.; Li, L.; Doke, T.; Huang, S.; Zhao, J.; Kang, H.M.; Shrestra, R.; Balzer, M.S.; et al. The Nuclear Receptor ESRRA Protects from Kidney Disease by Coupling Metabolism and Differentiation. Cell Metab. 2021, 33, 379–394.e8. [Google Scholar] [CrossRef]
Figure 1. D1R and AT2R recruitment in urine-derived renal proximal tubule cells (uRPTCs) from salt-resistant (SR) and inverse salt-sensitive (ISS) subjects. (A,B) Under 1-h 10 µM monensin (MON) treatment, there are more D1Rs (A) and AT2Rs (B) recruited to the cell membrane in ISS uRPTCs, but not in SR uRPTCs (D1R: MON/VEH: SR, 1.022 ± 0.038, n = 6; ISS, 1.635 ± 0.178, n = 6; t-test, ** p < 0.01; AT2R: MON/VEH: SR, 0.965 ± 0.04, n = 6; ISS, 1.434 ± 0.107, n = 6; t-test, ** p < 0.01). (C) Correlation between the degrees of salt sensitivity (SS), expressed as a change in MAP (ΔMAP, High Salt–Low Salt) in response to increased dietary sodium and FEN-stimulated AT2R membrane recruitment in directly evaluated uRPTCs. The SS index of each SS class is depicted on the X axis expressed as the mean ΔMAP vs. AT2R 647/488 (representing AT2R recruitment from the cytosol to the plasma membrane). A higher ratio of FEN-induced plasma membrane AT2R expression is observed in ISS individuals compared with SR individuals (AT2R: FEN/VEH: ISS, 1.51 ± 0.13, n = 4, mean ΔMAP = −12.5 mm Hg; SR, 0.955 ± 0.04, n = 4, mean ΔMAP = −1.38; SS, 1.148 ± 0.082, n = 3, mean ΔMAP = +14.9 mm Hg; one-way ANOVA, Holm-Sidak post hoc test ** p < 0.01).
Figure 1. D1R and AT2R recruitment in urine-derived renal proximal tubule cells (uRPTCs) from salt-resistant (SR) and inverse salt-sensitive (ISS) subjects. (A,B) Under 1-h 10 µM monensin (MON) treatment, there are more D1Rs (A) and AT2Rs (B) recruited to the cell membrane in ISS uRPTCs, but not in SR uRPTCs (D1R: MON/VEH: SR, 1.022 ± 0.038, n = 6; ISS, 1.635 ± 0.178, n = 6; t-test, ** p < 0.01; AT2R: MON/VEH: SR, 0.965 ± 0.04, n = 6; ISS, 1.434 ± 0.107, n = 6; t-test, ** p < 0.01). (C) Correlation between the degrees of salt sensitivity (SS), expressed as a change in MAP (ΔMAP, High Salt–Low Salt) in response to increased dietary sodium and FEN-stimulated AT2R membrane recruitment in directly evaluated uRPTCs. The SS index of each SS class is depicted on the X axis expressed as the mean ΔMAP vs. AT2R 647/488 (representing AT2R recruitment from the cytosol to the plasma membrane). A higher ratio of FEN-induced plasma membrane AT2R expression is observed in ISS individuals compared with SR individuals (AT2R: FEN/VEH: ISS, 1.51 ± 0.13, n = 4, mean ΔMAP = −12.5 mm Hg; SR, 0.955 ± 0.04, n = 4, mean ΔMAP = −1.38; SS, 1.148 ± 0.082, n = 3, mean ΔMAP = +14.9 mm Hg; one-way ANOVA, Holm-Sidak post hoc test ** p < 0.01).
Biomedicines 10 02811 g001
Figure 2. D2R immunostaining in human kidney and renal proximal tubule cells. (A) D2R is stained red in the kidney. LTA, Lotus tetragonolobus, a proximal tubule marker. (B) Plasma membrane expression of D2R in renal proximal tubule cells. Nuclei are stained blue. Scale bar = 10 μm (C) Plasma membrane D2R expression, using an extracellular epitope-specific anti-D2R polyclonal antibody directly labeled with Alexa 647, is lower in fixed uRPTCs from individuals with ISS than individuals with SR (SR, 1.07 ± 0.02; vs. ISS, 0.81 ± 0.02; **** p < 0.001, n = 54; t-test). ISS = inverse salt-sensitive; SR = salt-resistant.
Figure 2. D2R immunostaining in human kidney and renal proximal tubule cells. (A) D2R is stained red in the kidney. LTA, Lotus tetragonolobus, a proximal tubule marker. (B) Plasma membrane expression of D2R in renal proximal tubule cells. Nuclei are stained blue. Scale bar = 10 μm (C) Plasma membrane D2R expression, using an extracellular epitope-specific anti-D2R polyclonal antibody directly labeled with Alexa 647, is lower in fixed uRPTCs from individuals with ISS than individuals with SR (SR, 1.07 ± 0.02; vs. ISS, 0.81 ± 0.02; **** p < 0.001, n = 54; t-test). ISS = inverse salt-sensitive; SR = salt-resistant.
Biomedicines 10 02811 g002
Figure 3. CTCF, NRF2 and SOD expression under salt treatment by In-Cell Western. SR and ISS cells were treated with low salt (90 mM), normal salt (140 mM) and high salt (190 mM) overnight. (A) CCCTC-binding factor (CTCF) expression decreased only in uRPTCs from ISS subjects incubated under low salt concentration (SR low salt, 1.02 ± 0.03 vs. ISS low salt, 0.83 ± 0.06; ISS low salt vs. ISS normal salt, 1 ± 0.04; ISS low salt vs. ISS high salt, 0.98 ± 0.03, * p < 0.05, ** p < 0.01, n = 9/group, two-way ANOVA, Holm-Sidak post hoc test). (B) Nuclear factor erythroid 2-related factor 2 (NRF2) expression significantly decreased only in ISS uRPTCs under low salt concentration (SR low salt, 0.98 ± 0.03; vs. ISS low salt, 0.87 ± 0.03; * p < 0.05, n = 18/group; ISS low salt vs. ISS normal salt, 1 ± 0.02; ISS low salt vs. ISS high salt, 0.96 ± 0.02, * p < 0.05, ** p < 0.01, n = 18/group, Two-way ANOVA, Holm-Sidak post hoc test). (C) SOD2 was significantly decreased only in ISS uRPTCs under low salt concentration (SR low salt, 1.15 ± 0.19; vs. ISS low salt, 0.58 ± 0.11; ISS low salt vs. normal salt 1 ± 0.08; ISS low salt vs. ISS high salt 1.02 ± 0.16, * p < 0.05, n = 18/group, ISS low salt vs. ISS normal salt, one-way ANOVA, Holm-Sidak post hoc test). ISS = inverse salt-sensitive; SR = salt-resistant.
Figure 3. CTCF, NRF2 and SOD expression under salt treatment by In-Cell Western. SR and ISS cells were treated with low salt (90 mM), normal salt (140 mM) and high salt (190 mM) overnight. (A) CCCTC-binding factor (CTCF) expression decreased only in uRPTCs from ISS subjects incubated under low salt concentration (SR low salt, 1.02 ± 0.03 vs. ISS low salt, 0.83 ± 0.06; ISS low salt vs. ISS normal salt, 1 ± 0.04; ISS low salt vs. ISS high salt, 0.98 ± 0.03, * p < 0.05, ** p < 0.01, n = 9/group, two-way ANOVA, Holm-Sidak post hoc test). (B) Nuclear factor erythroid 2-related factor 2 (NRF2) expression significantly decreased only in ISS uRPTCs under low salt concentration (SR low salt, 0.98 ± 0.03; vs. ISS low salt, 0.87 ± 0.03; * p < 0.05, n = 18/group; ISS low salt vs. ISS normal salt, 1 ± 0.02; ISS low salt vs. ISS high salt, 0.96 ± 0.02, * p < 0.05, ** p < 0.01, n = 18/group, Two-way ANOVA, Holm-Sidak post hoc test). (C) SOD2 was significantly decreased only in ISS uRPTCs under low salt concentration (SR low salt, 1.15 ± 0.19; vs. ISS low salt, 0.58 ± 0.11; ISS low salt vs. normal salt 1 ± 0.08; ISS low salt vs. ISS high salt 1.02 ± 0.16, * p < 0.05, n = 18/group, ISS low salt vs. ISS normal salt, one-way ANOVA, Holm-Sidak post hoc test). ISS = inverse salt-sensitive; SR = salt-resistant.
Biomedicines 10 02811 g003
Figure 4. Sodium influx in SR and ISS cells under ANG II treatment. Ang II increases luminal sodium transport to a greater extent in uRPTCs from individuals with ISS than those from individuals with SR. uRPTCs, incubated in low salt concentration (90 mM Sodium as NaCl, osmolality adjusted to 290 mOsm/L with mannitol) for 2 h, loaded with sodium-sensitive ion indicator SBFI (Ex 340/Em 510), in the presence of ouabain to inhibit basolateral sodium exit, were studied with and without 10 nmol/L Ang II. There is a robust increase in sodium influx with Ang II stimulation in uRPTCs from both SR (15426 ± 900 RFU SR vehicle vs. 20,357 ± 667 RFU SR Ang II, n = 15, * p < 0.01) and ISS (15571 ± 904 RFU ISS vehicle vs. 26,942 ± 815 RFU ISS Ang II, n = 15, one-way ANOVA, Holm-Sidak test, ‡ p < 0.001) individuals. However, the Ang II-mediated increase in sodium influx is greater in individuals with ISS than individuals with SR (20357 ± 667 RFU SR Ang II vs. 26,942 ± 815 RFU ISS Ang II, n = 15, one-way ANOVA, Holm-Sidak test, † p < 0.001). ISS = inverse salt-sensitive; SR = salt-resistant.
Figure 4. Sodium influx in SR and ISS cells under ANG II treatment. Ang II increases luminal sodium transport to a greater extent in uRPTCs from individuals with ISS than those from individuals with SR. uRPTCs, incubated in low salt concentration (90 mM Sodium as NaCl, osmolality adjusted to 290 mOsm/L with mannitol) for 2 h, loaded with sodium-sensitive ion indicator SBFI (Ex 340/Em 510), in the presence of ouabain to inhibit basolateral sodium exit, were studied with and without 10 nmol/L Ang II. There is a robust increase in sodium influx with Ang II stimulation in uRPTCs from both SR (15426 ± 900 RFU SR vehicle vs. 20,357 ± 667 RFU SR Ang II, n = 15, * p < 0.01) and ISS (15571 ± 904 RFU ISS vehicle vs. 26,942 ± 815 RFU ISS Ang II, n = 15, one-way ANOVA, Holm-Sidak test, ‡ p < 0.001) individuals. However, the Ang II-mediated increase in sodium influx is greater in individuals with ISS than individuals with SR (20357 ± 667 RFU SR Ang II vs. 26,942 ± 815 RFU ISS Ang II, n = 15, one-way ANOVA, Holm-Sidak test, † p < 0.001). ISS = inverse salt-sensitive; SR = salt-resistant.
Biomedicines 10 02811 g004
Figure 5. D2R cell surface expression when knocked down, overexpressed, or blocked in cells from subjects with SR or ISS. (A) siRNA decreases D2R cell surface expression in uRPTCs from both SR and ISS subjects (37.1% ± 2.0% reduction in SR DRD2 siRNA vs. SR control, n = 5, and 42.7% ± 6.0% reduction in ISS DRD2 siRNA vs. ISS control, n = 5, *, †, p < 0.01, Two-way ANOVA, Tukey’s test). The overexpression of D2R is relatively greater in uRPTCs from ISS than SR (22.5% ± 0.9% increase for ISS D2R overexpression over SR control siRNA, n = 5, ‡ p < 0.01, Two-way ANOVA, Tukey’s test). (B) mir-485-5p is involved in the reduced expression of the D2R in uRPTCs from individuals with ISS and DRD2 rs6276 because mir-485 blocker normalizes DRD2 expression in these cells (52619 ± 2001 RFU ISS control miRNA vs. 69,496 ± 2108 RFU ISS miRNA blocker and vs. 30,434 ± 1824 RFU ISS miRNA mimic, n = 9, Two-way ANOVA, Tukey’s test, *, †, ‡, p < 0.01); there is no effect in uRPTCs from individuals with SR. Mir-485 mimic decreases spiperone binding in uRPTCs from individuals with ISS and DRD2 rs6276; there is no effect in uRPTCs from individuals with SR. ISS = inverse salt-sensitive; SR = salt-resistant.
Figure 5. D2R cell surface expression when knocked down, overexpressed, or blocked in cells from subjects with SR or ISS. (A) siRNA decreases D2R cell surface expression in uRPTCs from both SR and ISS subjects (37.1% ± 2.0% reduction in SR DRD2 siRNA vs. SR control, n = 5, and 42.7% ± 6.0% reduction in ISS DRD2 siRNA vs. ISS control, n = 5, *, †, p < 0.01, Two-way ANOVA, Tukey’s test). The overexpression of D2R is relatively greater in uRPTCs from ISS than SR (22.5% ± 0.9% increase for ISS D2R overexpression over SR control siRNA, n = 5, ‡ p < 0.01, Two-way ANOVA, Tukey’s test). (B) mir-485-5p is involved in the reduced expression of the D2R in uRPTCs from individuals with ISS and DRD2 rs6276 because mir-485 blocker normalizes DRD2 expression in these cells (52619 ± 2001 RFU ISS control miRNA vs. 69,496 ± 2108 RFU ISS miRNA blocker and vs. 30,434 ± 1824 RFU ISS miRNA mimic, n = 9, Two-way ANOVA, Tukey’s test, *, †, ‡, p < 0.01); there is no effect in uRPTCs from individuals with SR. Mir-485 mimic decreases spiperone binding in uRPTCs from individuals with ISS and DRD2 rs6276; there is no effect in uRPTCs from individuals with SR. ISS = inverse salt-sensitive; SR = salt-resistant.
Biomedicines 10 02811 g005
Figure 6. pH recovery in SR and ISS cells under Ang II and Ang I treatments. SR and ISS uRPTCs were plated in 96-well plates incubated in 90 mmol/L NaCl, osmolality adjusted to 290 with mannitol, labeled with SNARF1 pH indicator, acidified with ammonium chloride (20 mmol/L, 10 min), switched to sodium-free media (290 osmolality/mannitol) with or without angiotensin II (Ang II, (A)) or angiotensin I (Ang I, (B)) (10 nmol/L), for 5 min with or without the addition of the AT1R antagonist, losartan (LOS, 1 μmol/L, 5 min). pH was measured in a microplate fluorometer using a custom single excitation dual emission filter set every 10 s with pH recovery initiated by low sodium buffer (90 mmol/L Sodium as NaCl) and pH recovery measured as pH units/sec. (A) Ang II increases pH recovery in low sodium concentration only in ISS uRPTCs (ISS Ang II 0.0044 ± 0.0003 vs. ISS VEH 0.0031 ± 0.0002, n = 6/group, Two-way ANOVA, Tukey’s test, * p < 0.05) and the AT1R antagonist, losartan, LOS completely blocks the response (ISS Ang II 0.0044 ± 0.0003 vs. ISS Ang II LOS 0.0029 ± 0.0004, n = 6/group, Two-way ANOVA, Tukey’s test, ‡ p < 0.05). Stable transfection of DRD2 (wD2L) completely blocks the low sodium Ang II-dependent increase in pH in uRPTCs from individuals with ISS (ISS Ang II 0.0044 ± 0.0003 vs. ISS Ang II w D2R 0.0024 ± 0.0003, n = 6/group, Two-way ANOVA, Holm-Sidak test, † p < 0.05). (B) The Ang II precursor peptide, Ang I, increases pH recovery in low sodium, only in ISS uRPTCs (ISS Ang I 0.0033 ± 0.0002 vs. ISS VEH 0.0023 ± 0.0001, n = 6/group, Two-way ANOVA, Tukey’s test, * p < 0.05) and LOS completely blocks the response (ISS Ang I 0.0033 ± 0.0002 vs. ISS Ang I LOS 0.002 ± 0.0002, n = 6/group, Two-way ANOVA, Tukey’s test, ‡ p < 0.05). Stable transfection of DRD2 (wD2L) completely blocks the low sodium Ang I-dependent increase in pH in uRPTCs from individuals with ISS (ISS Ang I 0.0033 ± 0.0002 vs. ISS Ang I w D2R 0.0022 ± 0.0002, n = 6/group, Two-way ANOVA, Holm-Sidak test, † p < 0.05). wo (non DRD2-transfected uRPTCs), wD2L (DRD2 Long-transfected uRPTCs). ISS = inverse salt-sensitive; SR = salt-resistant.
Figure 6. pH recovery in SR and ISS cells under Ang II and Ang I treatments. SR and ISS uRPTCs were plated in 96-well plates incubated in 90 mmol/L NaCl, osmolality adjusted to 290 with mannitol, labeled with SNARF1 pH indicator, acidified with ammonium chloride (20 mmol/L, 10 min), switched to sodium-free media (290 osmolality/mannitol) with or without angiotensin II (Ang II, (A)) or angiotensin I (Ang I, (B)) (10 nmol/L), for 5 min with or without the addition of the AT1R antagonist, losartan (LOS, 1 μmol/L, 5 min). pH was measured in a microplate fluorometer using a custom single excitation dual emission filter set every 10 s with pH recovery initiated by low sodium buffer (90 mmol/L Sodium as NaCl) and pH recovery measured as pH units/sec. (A) Ang II increases pH recovery in low sodium concentration only in ISS uRPTCs (ISS Ang II 0.0044 ± 0.0003 vs. ISS VEH 0.0031 ± 0.0002, n = 6/group, Two-way ANOVA, Tukey’s test, * p < 0.05) and the AT1R antagonist, losartan, LOS completely blocks the response (ISS Ang II 0.0044 ± 0.0003 vs. ISS Ang II LOS 0.0029 ± 0.0004, n = 6/group, Two-way ANOVA, Tukey’s test, ‡ p < 0.05). Stable transfection of DRD2 (wD2L) completely blocks the low sodium Ang II-dependent increase in pH in uRPTCs from individuals with ISS (ISS Ang II 0.0044 ± 0.0003 vs. ISS Ang II w D2R 0.0024 ± 0.0003, n = 6/group, Two-way ANOVA, Holm-Sidak test, † p < 0.05). (B) The Ang II precursor peptide, Ang I, increases pH recovery in low sodium, only in ISS uRPTCs (ISS Ang I 0.0033 ± 0.0002 vs. ISS VEH 0.0023 ± 0.0001, n = 6/group, Two-way ANOVA, Tukey’s test, * p < 0.05) and LOS completely blocks the response (ISS Ang I 0.0033 ± 0.0002 vs. ISS Ang I LOS 0.002 ± 0.0002, n = 6/group, Two-way ANOVA, Tukey’s test, ‡ p < 0.05). Stable transfection of DRD2 (wD2L) completely blocks the low sodium Ang I-dependent increase in pH in uRPTCs from individuals with ISS (ISS Ang I 0.0033 ± 0.0002 vs. ISS Ang I w D2R 0.0022 ± 0.0002, n = 6/group, Two-way ANOVA, Holm-Sidak test, † p < 0.05). wo (non DRD2-transfected uRPTCs), wD2L (DRD2 Long-transfected uRPTCs). ISS = inverse salt-sensitive; SR = salt-resistant.
Biomedicines 10 02811 g006
Figure 7. Plasma membrane Aminopeptidase N (APN) expression in SR and ISS cells under low salt treatment. In-Cell immunofluorescence staining in 96 well plates using an extracellular epitope specific antibody to APN shows that ISS cells have a higher plasma membrane APN expression than SR cells (Normal salt: ISS 0.49 ± 0.02 vs. SR 0.32 ± 0.03, * p < 0.01, n = 18/group, Two-way ANOVA, Tukey’s test). ISS uRPTCs have a further increase in APN expression when cultured under low salt concentration vs. the same cells cultured under normal salt concentration (ISS low salt 0.61 ± 0.03 vs. ISS normal salt 0.49 ± 0.02 † p < 0.05, n = 18/group, Two-way ANOVA, Holm-Sidak post hoc test). ISS = inverse salt-sensitive; SR = salt-resistant.
Figure 7. Plasma membrane Aminopeptidase N (APN) expression in SR and ISS cells under low salt treatment. In-Cell immunofluorescence staining in 96 well plates using an extracellular epitope specific antibody to APN shows that ISS cells have a higher plasma membrane APN expression than SR cells (Normal salt: ISS 0.49 ± 0.02 vs. SR 0.32 ± 0.03, * p < 0.01, n = 18/group, Two-way ANOVA, Tukey’s test). ISS uRPTCs have a further increase in APN expression when cultured under low salt concentration vs. the same cells cultured under normal salt concentration (ISS low salt 0.61 ± 0.03 vs. ISS normal salt 0.49 ± 0.02 † p < 0.05, n = 18/group, Two-way ANOVA, Holm-Sidak post hoc test). ISS = inverse salt-sensitive; SR = salt-resistant.
Biomedicines 10 02811 g007
Figure 8. Solute Carrier Family 5 Member 11 (SLC5A11) and DRD2 SNPs. (A) There is a linear relationship between the number of SNPs in DRD2 (rs6276 and rs6277) and SLC5A11 (rs11074656) and the magnitude of change in MAP (ΔMAP) when transitioning between a high and low salt diet (Y = 0.4996X − 2.396, R2 = 0.62, p < 0.05). (B) The cohort was divided into 3 groups, those with 1 or less, those with 2 to 4 and those with 5–6 DRD2 and SLC5A11 SNPs. ΔMAP is significantly greater in those with 5–6 SNPs than those with 1 or no SNPs (0–1: −2.12 ± 0.81, n = 58; 2–4: −1.14 ± 0.52, n = 190; 5–6: 0.98 ± 0.88, n = 54, * p < 0.05, one-way ANOVA, Tukey’s test). (C) Real time RT-PCR shows a significantly lower SLC5A11 expression in cell lines with SNPs than in cell lines with no SNPs, i.e., wild-type (WT) cell lines (WT, 1.25 ± 0.06, n = 2; heterozygous, 0.64 ± 0.15, n = 4; SNP, 0.44 ± 0.16, n = 3; WT vs. SNP, * p < 0.05, one-way ANOVA, Tukey’s test).
Figure 8. Solute Carrier Family 5 Member 11 (SLC5A11) and DRD2 SNPs. (A) There is a linear relationship between the number of SNPs in DRD2 (rs6276 and rs6277) and SLC5A11 (rs11074656) and the magnitude of change in MAP (ΔMAP) when transitioning between a high and low salt diet (Y = 0.4996X − 2.396, R2 = 0.62, p < 0.05). (B) The cohort was divided into 3 groups, those with 1 or less, those with 2 to 4 and those with 5–6 DRD2 and SLC5A11 SNPs. ΔMAP is significantly greater in those with 5–6 SNPs than those with 1 or no SNPs (0–1: −2.12 ± 0.81, n = 58; 2–4: −1.14 ± 0.52, n = 190; 5–6: 0.98 ± 0.88, n = 54, * p < 0.05, one-way ANOVA, Tukey’s test). (C) Real time RT-PCR shows a significantly lower SLC5A11 expression in cell lines with SNPs than in cell lines with no SNPs, i.e., wild-type (WT) cell lines (WT, 1.25 ± 0.06, n = 2; heterozygous, 0.64 ± 0.15, n = 4; SNP, 0.44 ± 0.16, n = 3; WT vs. SNP, * p < 0.05, one-way ANOVA, Tukey’s test).
Biomedicines 10 02811 g008
Figure 9. A model comparing SR and ISS under low salt condition. In low salt conditions juxtaglomerular cells increase renin and hepatocytes produce more angiotensinogen, delivering increased angiotensin I (Ang I) to the renal proximal tubule where angiotensin converting enzyme, ACE1, on the surface of the proximal tubule cell converts Ang I to Ang II. In ISS, angiotensin type 1 receptor (AT1R) is sensitized due to the decreased negative regulation by D2R. Increased AT1R signaling leads to increase sodium transport. ISS in low salt increases the expression of APN even more, causing a reduction of Ang III, the preferred agonist of AT2R, and further sensitizing the AT1R by reducing the negative regulation of the AT2R on AT1R signaling. A feed-forward system is then established by increased AT1R signaling leading to increased renin, angiotensinogen, ACE1, and lowering APA, in order to raise Ang II and lower Ang III peptide levels. The increased AT1R signaling lowers NRF2 and SOD2 levels leading to increased free radical damage to the cell, as well as decreased positive epigenetic regulation of DRD2 via CTCF. Green lines represent positive cell signaling pathways with thicker green lines representing enhanced positive signaling; Red lines represent negative cell signaling pathways and thinner red lines representing decreased negative regulation; Dotted circles around proteins represent decreased protein expression.
Figure 9. A model comparing SR and ISS under low salt condition. In low salt conditions juxtaglomerular cells increase renin and hepatocytes produce more angiotensinogen, delivering increased angiotensin I (Ang I) to the renal proximal tubule where angiotensin converting enzyme, ACE1, on the surface of the proximal tubule cell converts Ang I to Ang II. In ISS, angiotensin type 1 receptor (AT1R) is sensitized due to the decreased negative regulation by D2R. Increased AT1R signaling leads to increase sodium transport. ISS in low salt increases the expression of APN even more, causing a reduction of Ang III, the preferred agonist of AT2R, and further sensitizing the AT1R by reducing the negative regulation of the AT2R on AT1R signaling. A feed-forward system is then established by increased AT1R signaling leading to increased renin, angiotensinogen, ACE1, and lowering APA, in order to raise Ang II and lower Ang III peptide levels. The increased AT1R signaling lowers NRF2 and SOD2 levels leading to increased free radical damage to the cell, as well as decreased positive epigenetic regulation of DRD2 via CTCF. Green lines represent positive cell signaling pathways with thicker green lines representing enhanced positive signaling; Red lines represent negative cell signaling pathways and thinner red lines representing decreased negative regulation; Dotted circles around proteins represent decreased protein expression.
Biomedicines 10 02811 g009
Table 1. Distribution of SLC5A11 rare variant, rs11074656, in different salt sensitivities.
Table 1. Distribution of SLC5A11 rare variant, rs11074656, in different salt sensitivities.
Salt SensitivityWTHeterozygousSNP
ISS9 (29%)19 (61%)3 (10%)
SR111 (51%)83 (38%)24 (11%)
SS20 (38%)23 (43%)10 (19%)
Χ2 = 9.77 p < 0.05.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Gildea, J.J.; Xu, P.; Schiermeyer, K.A.; Yue, W.; Carey, R.M.; Jose, P.A.; Felder, R.A. Inverse Salt Sensitivity of Blood Pressure Is Associated with an Increased Renin-Angiotensin System Activity. Biomedicines 2022, 10, 2811. https://doi.org/10.3390/biomedicines10112811

AMA Style

Gildea JJ, Xu P, Schiermeyer KA, Yue W, Carey RM, Jose PA, Felder RA. Inverse Salt Sensitivity of Blood Pressure Is Associated with an Increased Renin-Angiotensin System Activity. Biomedicines. 2022; 10(11):2811. https://doi.org/10.3390/biomedicines10112811

Chicago/Turabian Style

Gildea, John J., Peng Xu, Katie A. Schiermeyer, Wei Yue, Robert M. Carey, Pedro A. Jose, and Robin A. Felder. 2022. "Inverse Salt Sensitivity of Blood Pressure Is Associated with an Increased Renin-Angiotensin System Activity" Biomedicines 10, no. 11: 2811. https://doi.org/10.3390/biomedicines10112811

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop