Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
J Am Soc Nephrol ; 34(7): 1279-1291, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37022120

ABSTRACT

SIGNIFICANCE STATEMENT: Although gene expression changes have been characterized in human diabetic kidney disease (DKD), unbiased tissue proteomics information for this condition is lacking. The authors conducted an unbiased aptamer-based proteomic analysis of samples from patients with DKD and healthy controls, identifying proteins with levels that associate with kidney function (eGFR) or fibrosis, after adjusting for key covariates. Overall, tissue gene expression only modestly correlated with tissue protein levels. Kidney protein and RNA levels of matrix metalloproteinase 7 (MMP7) strongly correlated with fibrosis and with eGFR. Single-cell RNA sequencing indicated that kidney tubule cells are an important source of MMP7. Furthermore, plasma MMP7 levels predicted future kidney function decline. These findings identify kidney tissue MMP7 as a biomarker of fibrosis and blood MMP7 as a biomarker for future kidney function decline. BACKGROUND: Diabetic kidney disease (DKD) is responsible for close to half of all ESKD cases. Although unbiased gene expression changes have been extensively characterized in human kidney tissue samples, unbiased protein-level information is not available. METHODS: We collected human kidney samples from 23 individuals with DKD and ten healthy controls, gathered associated clinical and demographics information, and implemented histologic analysis. We performed unbiased proteomics using the SomaScan platform and quantified the level of 1305 proteins and analyzed gene expression levels by bulk RNA and single-cell RNA sequencing (scRNA-seq). We validated protein levels in a separate cohort of kidney tissue samples as well as in 11,030 blood samples. RESULTS: Globally, human kidney transcript and protein levels showed only modest correlation. Our analysis identified 14 proteins with kidney tissue levels that correlated with eGFR and found that the levels of 152 proteins correlated with interstitial fibrosis. Of the identified proteins, matrix metalloprotease 7 (MMP7) showed the strongest association with both fibrosis and eGFR. The correlation between tissue MMP7 protein expression and kidney function was validated in external datasets. The levels of MMP7 RNA correlated with fibrosis in the primary and validation datasets. Findings from scRNA-seq pointed to proximal tubules, connecting tubules, and principal cells as likely cellular sources of increased tissue MMP7 expression. Furthermore, plasma MMP7 levels correlated not only with kidney function but also associated with prospective kidney function decline. CONCLUSIONS: Our findings, which underscore the value of human kidney tissue proteomics analysis, identify kidney tissue MMP7 as a diagnostic marker of kidney fibrosis and blood MMP7 as a biomarker for future kidney function decline.


Subject(s)
Diabetic Nephropathies , Matrix Metalloproteinase 7 , Humans , Matrix Metalloproteinase 7/genetics , Matrix Metalloproteinase 7/metabolism , Proteomics , Kidney/metabolism , Biomarkers , Fibrosis , RNA
2.
Kidney Int Rep ; 7(7): 1565-1574, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35812286

ABSTRACT

Introduction: Impaired response to erythropoiesis-stimulating agents (ESAs) is associated with increased mortality in patients with end-stage kidney disease. However, the underlying mechanisms are not fully elucidated. Accumulating data reveal that selenium (Se), a trace element, plays a key role in stress erythropoiesis and erythrocyte homeostasis. We evaluated the relationship between serum Se levels and the response to ESAs in hemodialysis patients. Methods: In this cross-sectional study, we determined serum Se levels in 173 hemodialysis patients. We analyzed the association of serum Se with ESA responsiveness, as defined by ESA resistance index. Results: Of the study participants, 50% had lower Se levels than the population-based reference values. We found that serum Se levels were significantly and inversely correlated with erythropoiesis resistance index (ERI) but not transferrin saturation (TSAT) or ferritin levels. Multiple regression analyses confirmed the association between Se levels and ESA hyporesponsiveness, independently of other known factors, such as iron status, being female, and dialysis vintage (ß = -0.11, P < 0.001). When patients were divided according to Se levels and iron status, both low serum Se (<10.5 µg/dl) and iron deficiency significantly affected the response to ESA. Conversely, serum Se levels were significantly different among groups when patients were divided according to ERI quartiles. The association of low serum Se with ESA hyporesponsiveness persisted after adjustment of confounding variables. Conclusion: Serum Se levels are associated with the response to ESAs and can predict ESA resistance independently of iron status in Japanese hemodialysis patients. These data open the possibility to test whether Se supplementation reduces ESA demand.

3.
Nat Rev Nephrol ; 18(2): 76-77, 2022 02.
Article in English | MEDLINE | ID: mdl-34819631

Subject(s)
Kidney , Humans
4.
Hypertens Res ; 44(12): 1557-1567, 2021 12.
Article in English | MEDLINE | ID: mdl-34326480

ABSTRACT

Pendrin is a Cl-/HCO3- exchanger selectively present in the intercalated cells of the kidney. Although experimental studies have demonstrated that pendrin regulates blood pressure downstream of the renin-angiotensin-aldosterone system, its role in human hypertension remains unclear. Here, we analyzed the quantitative changes in pendrin in urinary extracellular vesicles (uEVs) isolated from a total of 30 patients with primary aldosteronism (PA) and from a rat model of aldosterone excess. Western blot analysis revealed that pendrin is present in dimeric and monomeric forms in uEVs in humans and rats. In a rodent model that received continuous infusion of aldosterone with or without concomitant administration of the selective mineralocorticoid receptor (MR) antagonist esaxerenone, pendrin levels in uEVs, as well as those of epithelial Na+ channel (ENaC) and Na-Cl-cotransporter (NCC), were highly correlated with renal abundance. In patients with PA, pendrin levels in uEVs were reduced by 49% from baseline by adrenalectomy or pharmacological MR blockade. Correlation analysis revealed that the magnitude of pendrin reduction after treatment significantly correlated with the baseline aldosterone-renin ratio (ARR). Finally, a cross-sectional analysis of patients with PA confirmed a significant correlation between the ARR and pendrin levels in uEVs. These data are consistent with experimental studies showing the role of pendrin in aldosterone excess and suggest that pendrin abundance is attenuated by therapeutic interventions in human PA. Our study also indicates that pendrin analysis in uEVs, along with other proteins, can be useful to understand the pathophysiology of hypertensive disorders.


Subject(s)
Chloride-Bicarbonate Antiporters/urine , Extracellular Vesicles , Hyperaldosteronism , Sulfate Transporters/urine , Aldosterone , Animals , Cross-Sectional Studies , Humans , Rats
6.
Intern Med ; 60(18): 2939-2945, 2021 Sep 15.
Article in English | MEDLINE | ID: mdl-33776003

ABSTRACT

A 21-year-old woman was admitted to our hospital because of massive intestinal bleeding. She started hemodialysis due to myeloperoxidase antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) at 18 years of age. Her ANCA titers remained stable; however, her C-reactive protein increased on 5 mg/day prednisolone before admission. Computed tomography angiography revealed a ruptured jejunal arterial aneurysm. Transcatheter arterial embolization, blood transfusion and the reinforcement of steroid therapy resolved her symptoms of AAV. Our case of a young patient with AAV and medium-sized arterial vasculitis is rare and emphasizes that the ANCA titer does not always rise, especially in patients with nonrenal vasculitis flare-ups.


Subject(s)
Aneurysm , Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis , Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/complications , Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/diagnosis , Antibodies, Antineutrophil Cytoplasmic , Female , Gastrointestinal Hemorrhage , Humans , Peroxidase , Renal Dialysis , Young Adult
7.
Article in English | MEDLINE | ID: mdl-32883689

ABSTRACT

INTRODUCTION: Renal tubular injury contributes to the decline in kidney function in patients with diabetes. Cell type-specific DNA methylation patterns have been used to calculate proportions of particular cell types. In this study, we developed a method to detect renal tubular injury in patients with diabetes by detecting exfoliated tubular cells shed into the urine based on tubular cell-specific DNA methylation patterns. RESEARCH DESIGN AND METHODS: We identified DNA methylation patterns specific for human renal proximal tubular cells through compartment-specific methylome analysis. We next determined the methylation levels of proximal tubule-specific loci in urine sediment of patients with diabetes and analyzed correlation with clinical variables. RESULTS: We identified genomic loci in SMTNL2 and G6PC to be selectively unmethylated in human proximal tubular cells. The methylation levels of SMTNL2 and G6PC in urine sediment, deemed to reflect the proportion of exfoliated proximal tubular cells due to injury, correlated well with each other. Methylation levels of SMTNL2 in urine sediment significantly correlated with the annual decline in estimated glomerular filtration rate. Moreover, addition of urinary SMTNL2 methylation to a model containing known risk factors significantly improved discrimination of patients with diabetes with faster estimated glomerular filtration rate decline. CONCLUSIONS: This study demonstrates that patients with diabetes with continual loss in kidney function may be stratified by a specific DNA methylation signature through epigenetic urinalysis and provides further evidence at the level of exfoliated cells in the urine that injury of proximal tubular cells may contribute to pathogenesis of diabetic kidney disease.


Subject(s)
Diabetes Mellitus , Diabetic Nephropathies , DNA/metabolism , Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Diabetic Nephropathies/diagnosis , Diabetic Nephropathies/genetics , Glomerular Filtration Rate , Humans , Kidney/metabolism , Methylation
8.
Int J Mol Sci ; 21(15)2020 Jul 28.
Article in English | MEDLINE | ID: mdl-32731518

ABSTRACT

The kidney and the vasculature play crucial roles in regulating blood pressure. The ubiquitin proteasome system (UPS), a multienzyme process mediating covalent conjugation of the 76-amino acid polypeptide ubiquitin to a substrate protein followed by proteasomal degradation, is involved in multiple cellular processes by regulating protein turnover in various tissues. Increasing evidence demonstrates the roles of UPS in blood pressure regulation. In the kidney, filtered sodium is reabsorbed through diverse sodium transporters and channels along renal tubules, and studies conducted till date have provided insights into the complex molecular network through which ubiquitin ligases modulate sodium transport in different segments. Components of these pathways include ubiquitin ligase neuronal precursor cell-expressed developmentally downregulated 4-2, Cullin-3, and Kelch-like 3. Moreover, accumulating data indicate the roles of UPS in blood vessels, where it modulates nitric oxide bioavailability and vasoconstriction. Cullin-3 not only regulates renal salt reabsorption but also controls vascular tone using different adaptor proteins that target distinct substrates in vascular smooth muscle cells. In endothelial cells, UPS can also contribute to blood pressure regulation by modulating endothelial nitric oxide synthase. In this review, we summarize current knowledge regarding the role of UPS in blood pressure regulation, focusing on renal sodium reabsorption and vascular function.


Subject(s)
Blood Pressure , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Ubiquitin-Protein Ligases/biosynthesis , Ubiquitin/metabolism , Animals , Gene Expression Regulation, Enzymologic , Humans , Kidney/metabolism , Vasoconstriction
9.
J Clin Invest ; 130(8): 4152-4166, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32597829

ABSTRACT

Aging is associated with a high prevalence of hypertension due to elevated susceptibility of BP to dietary salt, but its mechanism is unknown. Serum levels of Klotho, an anti-aging factor, decline with age. We found that high salt (HS) increased BP in aged mice and young heterozygous Klotho-knockout mice and was associated with increased vascular expression of Wnt5a and p-MYPT1, which indicate RhoA activity. Not only the Wnt inhibitor LGK974 and the Wnt5a antagonist Box5 but Klotho supplementation inhibits HS-induced BP elevation, similarly to the Rho kinase inhibitor fasudil, associated with reduced p-MYPT1 expression in both groups of mice. In cultured vascular smooth muscle cells, Wnt5a and angiotensin II (Ang II) increased p-MYPT1 expression but knockdown of Wnt5a with siRNA abolished Ang II-induced upregulation of p-MYPT1, indicating that Wnt5a is indispensable for Ang II-induced Rho/ROCK activation. Notably, Klotho inhibited Wnt5a- and Ang II-induced upregulation of p-MYPT1. Consistently, Klotho supplementation ameliorated HS-induced augmentation of reduced renal blood flow (RBF) response to intra-arterial infusion of Ang II and the thromboxane A2 analog U46619, which activated RhoA in both groups of mice and were associated with the inhibition of BP elevation, suggesting that abnormal response of RBF to Ang II contributes to HS-induced BP elevation. Thus, Klotho deficiency underlies aging-associated salt-sensitive hypertension through vascular non-canonical Wnt5a/RhoA activation.


Subject(s)
Aging , Glucuronidase/deficiency , Hypertension , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Sodium Chloride, Dietary/adverse effects , Wnt-5a Protein/metabolism , Aging/drug effects , Aging/genetics , Aging/metabolism , Aging/pathology , Angiotensin II/genetics , Angiotensin II/metabolism , Animals , Glucuronidase/metabolism , Hypertension/chemically induced , Hypertension/genetics , Hypertension/metabolism , Hypertension/pathology , Klotho Proteins , Male , Mice , Mice, Knockout , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Myosin-Light-Chain Phosphatase/genetics , Myosin-Light-Chain Phosphatase/metabolism , Sodium Chloride, Dietary/pharmacology , Wnt-5a Protein/genetics
10.
Int J Mol Sci ; 21(12)2020 Jun 22.
Article in English | MEDLINE | ID: mdl-32580367

ABSTRACT

Renal inflammation is known to be involved in salt-induced renal damage, leading to end-stage renal disease. This study aims to evaluate the role of inflammation in anti-inflammatory and renoprotective effects of beraprost sodium (BPS), a prostaglandin I2 (PGI2) analog, in Dahl salt-sensitive (DS) rats. Five-week-old male DS rats were fed a normal-salt diet (0.5% NaCl), a high-salt diet (8% NaCl), or a high-salt diet plus BPS treatment for 3 weeks. BPS treatment could inhibit marked proteinuria and renal injury in salt-loaded DS rats with elevated blood pressure, accompanied by renal inflammation suppression. Notably, high salt increased renal expression of active Rac1, followed by increased Sgk1 expressions, a downstream molecule of mineralocorticoid receptor (MR) signal, indicating salt-induced activation of Rac1-MR pathway. However, BPS administration inhibited salt-induced Rac1-MR activation as well as renal inflammation and damage, suggesting that Rac1-MR pathway is involved in anti-inflammatory and renoprotective effects of PGI2. Based upon Rac1 activated by inflammation, moreover, BPS inhibited salt-induced activation of Rac1-MR pathway by renal inflammation suppression, resulting in the attenuation of renal damage in salt-loaded DS rats. Thus, BPS is efficacious for the treatment of salt-induced renal injury.


Subject(s)
Acute Kidney Injury/prevention & control , Epoprostenol/analogs & derivatives , Gene Expression Regulation/drug effects , Inflammation/prevention & control , Receptors, Mineralocorticoid/metabolism , Sodium Chloride/toxicity , rac1 GTP-Binding Protein/metabolism , Acute Kidney Injury/chemically induced , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Animals , Epoprostenol/pharmacology , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Male , Rats , Rats, Sprague-Dawley , Receptors, Mineralocorticoid/genetics , Vasodilator Agents/pharmacology , rac1 GTP-Binding Protein/genetics
11.
J Am Soc Nephrol ; 31(4): 748-764, 2020 04.
Article in English | MEDLINE | ID: mdl-32034107

ABSTRACT

BACKGROUND: Regulation of sodium chloride transport in the aldosterone-sensitive distal nephron is essential for fluid homeostasis and BP control. The chloride-bicarbonate exchanger pendrin in ß-intercalated cells, along with sodium chloride cotransporter (NCC) in distal convoluted tubules, complementarily regulate sodium chloride handling, which is controlled by the renin-angiotensin-aldosterone system. METHODS: Using mice with mineralocorticoid receptor deletion in intercalated cells, we examined the mechanism and roles of pendrin upregulation via mineralocorticoid receptor in two different models of renin-angiotensin-aldosterone system activation. We also used aldosterone-treated NCC knockout mice to examine the role of pendrin regulation in salt-sensitive hypertension. RESULTS: Deletion of mineralocorticoid receptor in intercalated cells suppressed the increase in renal pendrin expression induced by either exogenous angiotensin II infusion or endogenous angiotensin II upregulation via salt restriction. When fed a low-salt diet, intercalated cell-specific mineralocorticoid receptor knockout mice with suppression of pendrin upregulation showed BP reduction that was attenuated by compensatory activation of NCC. In contrast, upregulation of pendrin induced by aldosterone excess combined with a high-salt diet was scarcely affected by deletion of mineralocorticoid receptor in intercalated cells, but depended instead on hypokalemic alkalosis through the activated mineralocorticoid receptor-epithelial sodium channel cascade in principal cells. In aldosterone-treated NCC knockout mice showing upregulation of pendrin, potassium supplementation corrected alkalosis and inhibited the pendrin upregulation, thereby lowering BP. CONCLUSIONS: In conjunction with NCC, the two pathways of pendrin upregulation, induced by angiotensin II through mineralocorticoid receptor activation in intercalated cells and by alkalosis through mineralocorticoid receptor activation in principal cells, play important roles in fluid homeostasis during salt depletion and salt-sensitive hypertension mediated by aldosterone excess.


Subject(s)
Hypertension/etiology , Nephrons/metabolism , Nephrons/pathology , Receptors, Mineralocorticoid/physiology , Sodium Chloride Symporters/physiology , Sulfate Transporters/metabolism , Aldosterone , Animals , Disease Models, Animal , Mice , Mice, Knockout , Renin-Angiotensin System/physiology
12.
Hypertens Res ; 42(12): 1848-1857, 2019 12.
Article in English | MEDLINE | ID: mdl-31541221

ABSTRACT

The currently available data have indicated that dietary salt is directly correlated with blood pressure (BP) and the occurrence of hypertension. However, the salt sensitivity of BP is different in each individual. Genetic factors and environmental factors influence the salt sensitivity of BP. Obesity, stress, and aging are strongly associated with increased BP salt sensitivity. Indeed, a complex and interactive genetic and environmental system can determine an individual's BP salt sensitivity. However, the genetic/epigenetic determinants leading to salt sensitivity of BP are still challenging to identify primarily because lifestyle-related diseases, including hypertension, usually become a medical problem during adulthood, although their causes may be attributed to the earlier stages of ontogeny. The association between distinct developmental periods involves changes in gene expression, which include epigenetic phenomena. The role of epigenetic modification in the development of salt-sensitive hypertension is presently under investigation. Recently, we identified aberrant DNA methylation in the context of prenatally programmed hypertension. In this review, we summarize the existing knowledge regarding the pathophysiological mechanisms of salt-sensitive hypertension. Additionally, we discuss the contribution of epigenetic mechanisms in the development of salt-sensitive hypertension.


Subject(s)
Hypertension/physiopathology , Sodium Chloride, Dietary/adverse effects , Animals , Blood Pressure/drug effects , Epigenesis, Genetic , Humans
13.
Hypertens Res ; 42(4): 514-521, 2019 04.
Article in English | MEDLINE | ID: mdl-30631161

ABSTRACT

Excessive dietary salt intake can counteract the renoprotective effects of renin-angiotensin system (RAS) blockade in hypertensive patients with chronic kidney disease (CKD). In rodents, salt loading induces hypertension and renal damage by activating the mineralocorticoid receptor (MR) independently of plasma aldosterone levels. Thus, high salt-induced resistance to RAS blockade may be mediated by MR activation. To test this, a post hoc analysis of the Eplerenone Combination Versus Conventional Agents to Lower Blood Pressure on Urinary Antialbuminuric Treatment Effect (EVALUATE) trial was conducted. Thus, 304 non-diabetic hypertensive patients on RAS-blocking therapy were divided into tertiles according to salt intake (estimated 24-h urinary sodium excretion at baseline) and compared in terms of percent reduction in urinary albumin-to-creatinine ratio (UACR) at 52 weeks relative to baseline. The eplerenone-treated patients in the highest sodium excretion tertile exhibited significantly greater reduction in UACR than the placebo subjects in the same tertile (-22.5% vs. +21.8%, p = 0.02). This disparity was not observed in the lowest (-10.2% vs. -0.84%, p = 0.65) or middle (-19.5% vs. +9.5%, p = 0.22) tertiles. Similar systolic blood pressure changes were observed. In the whole cohort, reduction in UACR correlated positively with reduction in systolic blood pressure (r2 = 0.04, p = 0.02). These results support the hypothesis that excessive salt intake can enhance resistance to RAS blockade by activating MR. They also suggest that eplerenone plus RAS blockade may be effective for CKD in hypertensive patients, especially those with excessive salt intake.


Subject(s)
Albuminuria/drug therapy , Blood Pressure/drug effects , Eplerenone/therapeutic use , Hypertension/complications , Mineralocorticoid Receptor Antagonists/therapeutic use , Adult , Aged , Albuminuria/complications , Albuminuria/physiopathology , Blood Pressure/physiology , Eplerenone/pharmacology , Female , Humans , Hypertension/physiopathology , Male , Middle Aged , Mineralocorticoid Receptor Antagonists/pharmacology , Renin-Angiotensin System/drug effects , Sodium Chloride, Dietary , Young Adult
14.
Clin Sci (Lond) ; 133(1): 75-82, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30622159

ABSTRACT

Distal nephron of the kidney plays key roles in fluid volume and electrolyte homeostasis by tightly regulating reabsorption and excretion of Na+, K+, and Cl- Studies to date demonstrate the detailed electrolyte transport mechanisms in principal cells of the cortical collecting duct, and their regulation by renin-angiotensin-aldosterone system (RAAS). In recent years, however, accumulating data indicate that intercalated cells, another cell type that is present in the cortical collecting duct, also play active roles in the regulation of blood pressure. Notably, pendrin in ß-intercalated cells not only controls acid/base homeostasis, but is also one of the key components controlling salt and K+ transport in distal nephron. We have recently shown that pendrin is regulated by the co-ordinated action of angiotensin II (AngII) and aldosterone, and at the downstream of AngII, mammalian target of rapamycin (mTOR) signaling regulates pendrin through inhibiting the kinase unc51-like-kinase 1 and promoting dephosphorylation of mineralocorticoid receptor (MR). In this review, we summarize recent advances in the current knowledge on the salt transport mechanisms in the cortical collecting duct, and their regulation by the RAAS.


Subject(s)
Electrolytes/metabolism , Kidney Tubules, Collecting/metabolism , Membrane Transport Proteins/metabolism , Renal Elimination , Renal Reabsorption , Renin-Angiotensin System , Water-Electrolyte Balance , Animals , Blood Pressure , Humans , Sulfate Transporters/metabolism
15.
JCI Insight ; 3(21)2018 11 02.
Article in English | MEDLINE | ID: mdl-30385711

ABSTRACT

Maternal malnutrition, which causes prenatal exposure to excessive glucocorticoid, induces adverse metabolic programming, leading to hypertension in offspring. In offspring of pregnant rats receiving a low-protein diet or dexamethasone, a synthetic glucocorticoid, mRNA expression of angiotensin receptor type 1a (Agtr1a) in the paraventricular nucleus (PVN) of the hypothalamus was upregulated, concurrent with reduced expression of DNA methyltransferase 3a (Dnmt3a), reduced binding of DNMT3a to the Agtr1a gene, and DNA demethylation. Salt loading increased BP in both types of offspring, suggesting that elevated hypothalamic Agtr1a expression is epigenetically modulated by excessive glucocorticoid and leads to adult-onset salt-sensitive hypertension. Consistent with this, dexamethasone treatment of PVN cells upregulated Agtr1a, while downregulating Dnmt3a, and decreased DNMT3a binding and DNA demethylation at the Agtr1a locus. In addition, Dnmt3a knockdown upregulated Agtr1a independently of dexamethasone. Hypothalamic neuron-specific Dnmt3a-deficient mice exhibited upregulation of Agtr1a in the PVN and salt-induced BP elevation without dexamethasone treatment. By contrast, dexamethasone-treated Agtr1a-deficient mice failed to show salt-induced BP elevation, despite reduced expression of Dnmt3a. Thus, epigenetic modulation of hypothalamic angiotensin signaling contributes to salt-sensitive hypertension induced by prenatal glucocorticoid excess in offspring of mothers that are malnourished during pregnancy.


Subject(s)
DNA Methylation/genetics , Hypertension/genetics , Paraventricular Hypothalamic Nucleus/metabolism , Receptor, Angiotensin, Type 1/metabolism , Animals , Animals, Newborn , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A , Dexamethasone/supply & distribution , Epigenomics , Female , Glucocorticoids/supply & distribution , Hypertension/metabolism , Male , Mice , Pregnancy , Prenatal Exposure Delayed Effects , Protein-Energy Malnutrition/complications , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Up-Regulation/physiology
16.
Sci Rep ; 8(1): 16338, 2018 11 05.
Article in English | MEDLINE | ID: mdl-30397232

ABSTRACT

Epigenetic modulation may underlie the progression of diabetic nephropathy (DN). Involvement of TGFB1 in mesangial fibrosis of DN led us to hypothesize that Tgfb1 DNA demethylation contributes to progression of DN. In primary mesangial cells from diabetic (db/db) mouse kidneys, demethylation of Tgfb1 DNA and upregulation of Tgfb1 mRNA progressed simultaneously. USF1 binding site in Tgfb1 promoter region were demethylated, and binding of USF1 increased, with decreased binding of DNMT1 in db/db compared with control. Given downregulation of Tgfb1 expression by folic acid, antioxidant Tempol reversed DNA demethylation, with increased and decreased recruitment of DNMT1 and USF1 to the promoter, resulting in decreased Tgfb1 expression in db/db mice. Addition of H2O2 to mesangial cells induced DNA demethylation and upregulated Tgfb1 expression. Finally, Tempol attenuated mesangial fibrosis in db/db mice. We conclude that aberrant DNA methylation of Tgfb1 due to ROS overproduction play a key to mesangial fibrosis during DN progression.


Subject(s)
DNA Methylation , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Mesangial Cells/metabolism , Transforming Growth Factor beta1/genetics , Animals , Diabetes Mellitus, Experimental/metabolism , Disease Progression , Fibrosis , Male , Mesangial Cells/pathology , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Up-Regulation/genetics
17.
Am J Physiol Renal Physiol ; 314(4): F551-F560, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29212764

ABSTRACT

Epigenetic abnormalities have been suggested to mediate metabolic memory observed in diabetic complications. We have shown that epigenetic alterations may induce persistent phenotypic changes in the proximal tubules of the diabetic kidneys. In this study, we show that pregnane X receptor (PXR), a xenobiotic nuclear receptor, is epigenetically altered and upregulated and may have a possible function in the diabetic kidney. PXR has been shown to play a critical role in metabolic changes in obesity and diabetes; however, its distribution and function in the kidney are unknown. In the normal kidney, Pxr was selectively expressed in the proximal tubular cells with demethylation in the promoter DNA. In db/db mice, significant increases in Pxr mRNA, further demethylation of DNA, and stimulatory histone marks in the promoter were observed. Epigenetic changes are likely to play a causative role in PXR induction, since a DNA methyltransferase inhibitor increased PXR mRNA in cultured human proximal tubular cells. Administration of a PXR agonist increased mRNA levels of solute carrier organic anion transporter family member 2B1 ( Slco2b1), a xenobiotic transporter; response gene to complement 32 ( Rgc32), a molecule known to exert fibrotic effects in the kidney; and phosphoenolpyruvate carboxykinase 1 ( Pck1), a gluconeogenic enzyme in the kidney. The expressions of these genes were inhibited by PXR small interfering RNA in cultured proximal tubular cells. Increased mRNA levels of Slco2b1, Rgc32, and Pck1 were also observed in the kidney of db/db mice. These data indicate that PXR is upregulated in the diabetic kidney with aberrant epigenetic modifications and may modulate the course of diabetic kidney disease through the activation of these genes.


Subject(s)
DNA Methylation , Diabetic Nephropathies/genetics , Energy Metabolism/genetics , Epigenesis, Genetic , Kidney Tubules, Proximal/metabolism , Pregnane X Receptor/genetics , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , Diabetic Nephropathies/metabolism , Disease Models, Animal , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice, Inbred C57BL , Muscle Proteins/genetics , Muscle Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Organic Anion Transporters/genetics , Organic Anion Transporters/metabolism , Phenotype , Phosphoenolpyruvate Carboxykinase (GTP)/genetics , Phosphoenolpyruvate Carboxykinase (GTP)/metabolism , Pregnane X Receptor/metabolism , Promoter Regions, Genetic
18.
J Am Soc Nephrol ; 29(1): 57-68, 2018 01.
Article in English | MEDLINE | ID: mdl-29021385

ABSTRACT

The renin-angiotensin-aldosterone system has an important role in the control of fluid homeostasis and BP during volume depletion. Dietary salt restriction elevates circulating angiotensin II (AngII) and aldosterone levels, increasing levels of the Cl-/HCO3- exchanger pendrin in ß-intercalated cells and the Na+-Cl- cotransporter (NCC) in distal convoluted tubules. However, the independent roles of AngII and aldosterone in regulating these levels remain unclear. In C57BL/6J mice receiving a low-salt diet or AngII infusion, we evaluated the membrane protein abundance of pendrin and NCC; assessed the phosphorylation of the mineralocorticoid receptor, which selectively inhibits aldosterone binding in intercalated cells; and measured BP by radiotelemetry in pendrin-knockout and wild-type mice. A low-salt diet or AngII infusion upregulated NCC and pendrin levels, decreased the phosphorylation of mineralocorticoid receptor in ß-intercalated cells, and increased plasma aldosterone levels. Notably, a low-salt diet did not alter BP in wild-type mice, but significantly decreased BP in pendrin-knockout mice. To dissect the roles of AngII and aldosterone, we performed adrenalectomies in mice to remove aldosterone from the circulation. In adrenalectomized mice, AngII infusion again upregulated NCC expression, but did not affect pendrin expression despite the decreased phosphorylation of mineralocorticoid receptor. By contrast, AngII and aldosterone coadministration markedly elevated pendrin levels in adrenalectomized mice. Our results indicate that aldosterone is necessary for AngII-induced pendrin upregulation, and suggest that pendrin contributes to the maintenance of normal BP in cooperation with NCC during activation of the renin-angiotensin-aldosterone system by dietary salt restriction.


Subject(s)
Aldosterone/blood , Angiotensin II/pharmacology , Sodium Chloride Symporters/metabolism , Sulfate Transporters/metabolism , Vasoconstrictor Agents/pharmacology , Adrenalectomy , Aldosterone/pharmacology , Animals , Blood Pressure/genetics , Kidney Tubules, Distal/cytology , Male , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Receptors, Mineralocorticoid/metabolism , Sodium Chloride, Dietary/administration & dosage , Sulfate Transporters/genetics , Up-Regulation/drug effects
19.
Hypertension ; 70(1): 111-118, 2017 07.
Article in English | MEDLINE | ID: mdl-28559392

ABSTRACT

Genome-wide analysis of renal sodium-transporting system has identified specific variations of Mendelian hypertensive disorders, including HSD11B2 gene variants in apparent mineralocorticoid excess. However, these genetic variations in extrarenal tissue can be involved in developing hypertension, as demonstrated in former studies using global and brain-specific Hsd11b2 knockout rodents. To re-examine the importance of renal dysfunction on developing hypertension, we generated kidney-specific Hsd11b2 knockout mice. The knockout mice exhibited systemic hypertension, which was abolished by reducing salt intake, suggesting its salt-dependency. In addition, we detected an increase in renal membrane expressions of cleaved epithelial sodium channel-α and T53-phosphorylated Na+-Cl- cotransporter in the knockout mice. Acute intraperitoneal administration of amiloride-induced natriuresis and increased urinary sodium/potassium ratio more in the knockout mice compared with those in the wild-type control mice. Chronic administration of amiloride and high-KCl diet significantly decreased mean blood pressure in the knockout mice, which was accompanied with the correction of hypokalemia and the resultant decrease in Na+-Cl- cotransporter phosphorylation. Accordingly, a Na+-Cl- cotransporter blocker hydrochlorothiazide significantly decreased mean blood pressure in the knockout mice. Chronic administration of mineralocorticoid receptor antagonist spironolactone significantly decreased mean blood pressure of the knockout mice along with downregulation of cleaved epithelial sodium channel-α and phosphorylated Na+-Cl- cotransporter expression in the knockout kidney. Our data suggest that kidney-specific deficiency of 11ß-HSD2 leads to salt-dependent hypertension, which is attributed to mineralocorticoid receptor-epithelial sodium channel-Na+-Cl- cotransporter activation in the kidney, and provides evidence that renal dysfunction is essential for developing the phenotype of apparent mineralocorticoid excess.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 2/genetics , Blood Pressure/genetics , Hypertension , Receptors, Mineralocorticoid/metabolism , Renal Insufficiency , Sodium Chloride, Dietary , Animals , Disease Models, Animal , Epithelial Sodium Channels/drug effects , Epithelial Sodium Channels/metabolism , Gene Deletion , Hypertension/genetics , Hypertension/metabolism , Hypertension/physiopathology , Hypertension/therapy , Ion Transport/drug effects , Mice , Mice, Knockout , Mineralocorticoid Receptor Antagonists/pharmacology , Renal Insufficiency/metabolism , Renal Insufficiency/physiopathology , Sodium Chloride, Dietary/adverse effects , Sodium Chloride, Dietary/metabolism
20.
Hypertension ; 69(5): 855-862, 2017 05.
Article in English | MEDLINE | ID: mdl-28289181

ABSTRACT

Aldosterone plays an important role in regulating Na-Cl reabsorption and blood pressure. Epithelial Na+ channel, Na+-Cl- cotransporter, and Cl-/HCO3- exchanger pendrin are the major mediators of Na-Cl transport in the aldosterone-sensitive distal nephron. Existing evidence also suggests that plasma K+ concentration affects renal Na-Cl handling. In this study, we posited that hypokalemia modulates the effects of aldosterone on pendrin in hyperaldosteronism. Chronic aldosterone infusion in mice increased pendrin levels at the plasma membrane, and correcting hypokalemia in this model almost completely blocked pendrin upregulation. However, hypokalemia induced by a low-K+ diet resulted in pendrin downregulation along with reduced plasma aldosterone levels, indicating that both hypokalemia and aldosterone excess are necessary for pendrin induction. In contrast, decreased plasma K+ levels were sufficient to increase Na+-Cl- cotransporter levels. We found that phosphorylation of mineralocorticoid receptor that prevents aldosterone binding in intercalated cells was suppressed by hypokalemia, which resulted in enhanced pendrin response to aldosterone, explaining the coordinated action of aldosterone and hypokalemia in pendrin regulation. Finally, to address the physiological significance of our observations, we administered aldosterone to mice lacking pendrin. Notably, plasma K+ levels were significantly lower in pendrin knockout mice (2.7±0.1 mmol/L) than in wild-type mice (3.0±0.1 mmol/L) after aldosterone infusion, demonstrating that pendrin alleviates hypokalemia in a state of aldosterone excess. These data indicate that the decreased plasma K+ levels promote pendrin induction by aldosterone, which, in concert with Na+-Cl- cotransporter, counteracts the progression of hypokalemia but promotes hypertension in primary aldosterone excess.


Subject(s)
Aldosterone/pharmacology , Anion Transport Proteins/metabolism , Blood Pressure/physiology , Hypokalemia/metabolism , Kidney/metabolism , Aldosterone/blood , Animals , Anion Transport Proteins/genetics , Blood Pressure/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Diet , Down-Regulation/drug effects , Hypertension/metabolism , Hypokalemia/genetics , Kidney/drug effects , Mice , Mice, Knockout , Phosphorylation/drug effects , Receptors, Mineralocorticoid/metabolism , Sodium Chloride Symporters/genetics , Sodium Chloride Symporters/metabolism , Sulfate Transporters , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...