Skip to main content
Erschienen in: medizinische genetik 1/2019

Open Access 01.02.2019 | Übersichten

Disorders of renal NaCl transport and implications for blood pressure regulation

verfasst von: J. Christopher Hennings, Prof. Dr. Christian A. Hübner

Erschienen in: medizinische genetik | Ausgabe 1/2019

Abstract

Hypertension is one of the major risk factors for cardiovascular disease in industrialized societies. Substantial progress has been made in understanding its epidemiology, its pathophysiology, and its associated risks such as coronary artery disease, stroke, and heart failure. Because there is consensus that the abnormal retention of sodium by the kidney is a major important pathophysiological event in hypertension, this review focuses on mechanisms of renal NaCl transport and associated genetic disorders.

Blood pressure and arterial hypertension

The requirement of a circulatory system is a consequence of the increasing size of multicellular organisms during evolution. Arterial blood pressure is a result of blood flow through the vasculature, which ensures the transportation of cells, nutrients, hormones, metabolites, O2, and CO2 throughout the body. It is determined by the total mean flow in the circulation (cardiac output) and the peripheral resistance of the vasculature. Starting from 115 mm Hg, each difference of 20 mm Hg in systolic blood pressure is associated with a twofold increase in the risk of dying from stroke and ischemic heart disease [30]. Because the relationship between blood pressure and cardiovascular and renal events is continuous, the distinction between normal blood pressure and hypertension is somewhat arbitrary [5]. High blood pressure is the leading cause of death and disability-adjusted life years worldwide [33, 61] Therefore, understanding of the regulation of blood pressure and identification of the genetic factors that contribute to blood pressure are of key relevance.

Role of NaCl in blood pressure and arterial hypertension

Because NaCl is the major osmotic constituent of the extracellular fluid (ECF), it largely determines ECF volume and cardiac output. Usually dietary Na+ uptake matches Na+ excretion. If this balance is disturbed, Na+ loss leads to decreased blood pressure, whereas Na+ retention leads to increased blood pressure. How this can lead to chronic hypertension is still controversial. Although it is widely assumed that all pressor ‘sensitivity’ to dietary NaCl depends on its Na+, selective dietary loading of Na+ without Cl has repeatedly failed to induce a pressor effect [62]. Because substitution of dietary NaCl with equimolar sodium bicarbonate leads to a reduction in blood pressure [34], there is evidence that renal Cl transport is an independent determinant of vascular volume and blood pressure regulation. The identification of genetic defects (Table 1) that are linked to altered renal Na+ and Cl handling characterized by either decreased or increased blood pressure in rare monogenic disorders highlights the critical role of NaCl homeostasis in blood pressure regulation [32, 46].
Table 1
Genes linked with blood pressure regulation/pathological conditions
Gene
Full gene name
Syndrome/animal model
Defective/altered function
Ref.
Proximal tubule
SLC9A3
Solute carrier 9, member 3 (NHE3)
Mouse: blood pressure reduced, hyperaldosteronism
Apical transporter using sodium gradient for urinary acidification
[45]
SLC4A4
Solute carrier 4, member 4 (NBC1)
Mouse: blood pressure reduced, hyperaldosteronism
Apical transporter using sodium gradient for bicarbonate recovery
[16]
Thick ascending limb
SLC12A1
Solute carrier 12, member 1 (NKCC2)
Bartter syndrome type 1
Apical ion transport
[50]
KCNJ1
Potassium inwardly-rectifying channel, subfamily J, member 1 (ROMK)
Bartter syndrome type 2
Apical potassium recycling
[51]
CLCNKB
Chloride channel, voltage-sensitive Kb (ClC-K2)
Bartter syndrome type 3
Basolateral chloride exit in TAL
[49]
BSND
Barttin CLCNK-type accessory beta subunit
Bartter syndrome type 4
Correct membrane targeting of CLCNK channels
[2]
CASR
Calcium-sensing receptor
Hypocalcemia with Bartter syndrome
CASR activation inhibits ROMK in rats, hyperprostaglandinism
[60]
MAGED2
Melanoma antigen, family D, 2
Transient Bartter syndrome 5
Regulates stability of NKCC2 and ROMK in the unborn and during early life
[28]
UMOD
Uromodulin
Variant associated with blood pressure
GPI-anchored glycoprotein secreted by cleavage into urine
[40]
Distal convoluting tubule
SLC12A3
Solute carrier 12, member 3 (NCC)
Gitelman syndrome
Apical ion transport
[52]
WNK1
WNK lysine-deficient protein kinase 1
PHAIIC
Phosphorylation NCC
[63]
WNK4
WNK lysine deficient protein kinase 4
PHAIIB
Regulation of WNK1 activity by phosphorylation
[63]
KLHL3
Kelch-like 3
PHAIID
E3 Ubiquitin ligase
[4]
CUL3
Cullin 3
PHAIIE
E3 Ubiquitin ligase
[4]
KCNJ10
Potassium inwardly rectifying channel, subfamily J, member 10
EAST syndrome (SeSAME syndrome)
Basolateral potassium recycling with impact on NCC regulation
[3]
Cortical collecting duct
SLC26A4
Solute carrier family 26, member 4 (PDS)
Mouse: blood pressure changed if challenged
Apical ion transport
[13]
SLC4A8
Solute carrier family 4, member 8 (NDCBE)
Mouse: blood pressure changed if challenged
Apical ion transport
[29]
SCNN1A
Sodium channel, nonvoltage-gated 1 alpha (ENaC alpha)
Liddle syndrome, PHAI
Apical localization in principal cells
[8]
SCNN1B
Sodium channel, nonvoltage-gated 1 beta (ENaC beta)
Liddle syndrome, PHAI
Apical localization in principal cells
[48]
SCNN1G
Sodium channel, nonvoltage-gated 1 gamma (ENaC gamma)
Liddle syndrome, PHAI
Apical localization in principal cells
[19]
NEDD4L
Neural precursor cell expressed, developmentally down-regulated gene 4‑like
Variant associated with blood pressure
Ubiquinates ENaC subunits
[36]
Aldosterone axis
CYP11B2
Cytochrome P450, family 11, subfamily b, polypeptide 2 (aldosterone synthase)
Familial hyper-/hypoaldosteronism
Aldosterone synthesis
[31]
CYP11B1
Cytochrome P450, family 11, subfamily b, polypeptide 1 (11-beta-hydroxylase)
Aldosteronism, glucocorticoid-remediable
Aldosterone synthesis
[31]
NR3C2
Nuclear receptor family 3, group C, member 2 (mineralocorticoid receptor)
Hypertension, PHAI
Aldosterone receptor
[17]
KCNJ5
Potassium inwardly rectifying channel, subfamily J, member 5
Familial hyperaldosteronism type III
Regulation of aldosterone production
[9]
CACNA1D
Calcium channel, voltage-dependent, L‑type, alpha, 1D subunit
PASNA
Regulation of aldosterone production
[1]
CACNA1H
Calcium channel, voltage-dependent, L‑type, alpha, 1H subunit
Familial hyperaldosteronism type IV
Regulation of aldosterone production
[44]
Genes are sorted according to the localization of the respective proteins along the nephron or listed as genes encoding extrarenal proteins. For some genes, the function has been resolved by animal models, but no human disorder has yet been reported. For many genes, the expression is not limited to the kidney. Owing to space limitations, extrarenal functions are not discussed in this review
PHA pseudohyperaldosteronism, PASNA Primary aldosteronism, seizures, and neurological abnormalities (PHA with seizures and neuronal abnormalities), TAL thick ascending limb

Renal NaCl uptake and associated monogenic disorders

The basic functional unit of the kidney is the nephron, which consists of the glomerulus, where Na+ and Cl are freely filtered, and the tubular system, which conveys the urine into the renal pelvis (Fig. 1). Along the passage of the tubular epithelium many constituents of the primary filtrate including Na+ and Cl are reabsorbed by an orchestra of different transport systems. Most but not all of the filtered Na+ is reabsorbed together with Cl. The segmental Cl handling differs from Na+ and involves both paracellular and transcellular routes.
Roughly 67% of the filtered NaCl and water reabsorption takes place in near isotonic fashion in the proximal tubule (PT). The driving force is mainly provided by the basolateral Na+/K+-ATPase, which sets the electrochemical gradient to drive a number of apical transporters that mediate Na+ and/or Cl+ entry such as Na+-dependent glucose transporters (SGLTs), the Na+/H+ exchanger NHE3/SLC9A3 [57], which exploits the downhill Na+ gradient across the apical cell membrane generated by the Na+/K+-ATPase, and the electrogenic Na+/HCO3 co-transporter NBC1/SLC4A4, which is mutated in proximal renal tubular acidosis [21]. In addition, the blood pressure of Slc4a4 [16] and Slc9a3 [45] knockout mice was reduced. This example illustrates the enormous capacity of the downstream nephron to compensate for impaired proximal NaCl uptake. In fact, patients with SLC9A3 mutations mainly suffer from congenital secretory sodium diarrhea [24]. Further, a considerable part of Cl reabsorption in the PT occurs in a paracellular manner, which is driven by the lumen-negative transepithelial potential difference. An additional 25% of the Na+ and Cl is reabsorbed by the Na+/K+/2Cl co-transporter NKCC2/SLC12A1 [14] in the thick ascending limb of Henle’s loop, which is inhibited by loop diuretics such as furosemide and bumetanide. These drugs are widely used to treat hypertension and edema associated with congestive heart failure. To maintain NKCC2-mediated NaCl uptake, K+ entering the cell via NKCC2 has to be recycled by the apical K+ channel ROMK/KCNJ1. At the basolateral side, Na+ leaves the cell via the Na+/K+-ATPase, whereas Cl efflux is largely mediated by the Cl channel CLCNKB/ClC-K2 [20], which requires its β‑subunit Barttin/BSND [11]. Notably, mutations in SLC12A1 [50], KCNJ1 [51], CLCNKB [49] or BSND [2] can cause Bartter syndrome, which mimics the effects of loop diuretics with marked secondary hyperaldosteronism originating from renal salt-wasting with normal or low blood pressure, hypokalemia, and metabolic alkalosis. Gain-of-function mutations of the calcium sensing receptor CaSR, which is involved in parathyroid hormone secretion, renal calcium, and to some extent renal salt reabsorption, are another cause of Bartter syndrome [60]. Recently, a transient form of antenatal Bartter syndrome was identified, which is caused by MAGED2 mutations [28]. It is assumed that MAGED2 participates in the maturation of membrane proteins within the endoplasmic reticulum and thereby affects the expression of NKCC2, ROMK, and possibly other membrane proteins.
In the distal convoluted tubule (DCT) another 5% of the filtered Na+ is taken up by the apical NaCl co-transporter NCC/SLC12A3 [15], which is highly sensitive to thiazide diuretics. NCC loss-of-function mutations are found in Gitelman syndrome patients [52], who generally have a low or normal blood pressure with low levels of chloride, potassium, and magnesium, and show decreased urinary calcium excretion. NCC regulation is critical, as familial hyperkalemic hypertension can be caused by mutations in WNK kinases WNK1 and WNK4 [63], both controlling NCC activity in the DCT [64]. Moreover, mutations in KLHL3 and CUL3 have been reported in patients with pseudohypoaldosteronism II featuring hypertension, hyperkalemia, and metabolic acidosis [4]. CUL3 and BTB-domain-containing kelch proteins, such as KLHL3, are components of cullin-RING E3 ligase complexes that ubiquitinate substrates such as WNK kinases and thereby control NCC activity [10, 47].
A salt-wasting renal tubulopathy similar to Gitelman syndrome is accompanied by epilepsy, ataxia, and sensorineural deafness in EAST syndrome, which is caused by mutations in the K+ channel KCNJ10 [3]. KCNJ10 is expressed basolaterally in the DCT and connecting tubule of the kidney [42].
Finally, the remaining 3% enter the collecting system, which consists of the connecting tubule and the collecting duct (CD) and is under control of the renin–angiotensin–aldosterone system, antidiuretic hormone/vasopressin, and natriuretic peptides (NPs), which are released from the heart in response to pressure and volume overload and inhibit renal sodium reabsorption. Therefore, the distal tubule plays an essential role in the fine tuning of salt, electrolyte, water, and acid-base balance. Its epithelium comprises two main cell types, principal cells and intercalated cells [37].
In the connecting tubule and CD, Na+ reabsorption is largely achieved through the apical epithelial sodium channel ENaC [6], in tandem with the basolateral Na+/K+-ATPase, both expressed in connecting tubule cells and principal cells of the CD. Aldosterone increases both Na+ entry through ENaC and basolateral Na+ extrusion via the Na+/K+ ATPase. Notably, mutations in the β‑ [48] or γ‑subunit [19] of ENaC, which result in an increased number of these channels in the apical cell membrane, cause Liddle syndrome with severe early onset hypertension, whereas loss of function mutations in the α‑, β‑ or γ‑subunit cause pseudohypoaldosteronism type I [8, 54]. Pseudohypoaldosteronism type I is characterized by salt wasting and severe hypotension. Thereby, the mutation–phenotype correlation for ENaC illustrates the concept of activating and inactivating mutations resulting in opposite phenotypes (hyper- versus hypotension).
Because approximately 50% of Na+ absorption in the cortical collecting duct of rats is insensitive to amiloride but sensitive to thiazides [56], a second mechanism independent of principal cells and ENaC was suspected. First evidence that intercalated cells are also involved in NaCl homeostasis came from the targeted inactivation of the apical HCO3/Cl exchanger pendrin/Slc26a4 of type B intercalated cells, which protected mice against mineralocorticoid-induced hypertension and favored hypotension upon NaCl depletion [58, 59]. Moreover, overexpression of pendrin induced arterial hypertension in mice fed on a high salt diet [23]. Mutations in SLC26A4 result in Pendred syndrome, the most common syndromic form of deafness, associated with developmental abnormalities of the cochlea, sensorineural hearing loss, and diffuse thyroid enlargement [13]. Although hypertension was not reported in a small retrospective analysis of patients with Pendred syndrome [35], it is still unclear whether Pendred syndrome protects against the development of hypertension. Furthermore, studies on isolated cortical collecting tubules suggested a second mechanism of Na+ reabsorption different than ENaC, which was blocked by thiazide diuretics, although NCC is absent from CDs. Because isolated CDs of mice, which lack the Na+-dependent Cl/HCO3 exchanger Ndcbe/Slc4a8, were devoid of ENaC-independent NaCl reabsorption [29], thiazide-sensitive NaCl uptake in the CD likely results from the functional coupling of pendrin and Slc4a8. The basolateral NaCl exit is independent of the Na+/K+-ATPase but critically relies on the presence of the basolateral V‑ATPase and Ae4/Slc4a9 [7].
Excessive aldosterone secretion causes hypokalemia and hypertension, whereas too little aldosterone results in hyperkalemia and hypotension. Mutations that affect aldosterone synthesis result in severe hypotension [41]. Glucocorticoid-remediable aldosteronism is caused by a gene duplication arising by unequal crossing over between two closely related genes involved in adrenal steroid biosynthesis, i. e., aldosterone synthase (encoded by CYP11B2) and 11-β-hydroxylase (encoded by CYP11B1) [31]. As a consequence, regulatory sequences of 11-β-hydroxylase are fused with the coding sequences for aldosterone synthase thus leading to early onset of hypertension with normal or elevated aldosterone levels despite suppressed plasma renin activity. Normally, 11-β-hydroxylase protects the mineralocorticoid receptor (encoded by NR3C2) from cortisol by metabolizing it to cortisone. Because cortisol is also able to effectively activate the mineralocorticoid receptor, patients lacking the 11-β-hydroxylase develop apparent hyperaldosteronism with early onset hypertension and hypokalemia in the virtual absence of aldosterone. Chronic consumption of licorice can result in a similar condition, because a metabolite of licorice inhibits 11-β-hydroxylase [53]. Mutations in the mineralocorticoid receptor gene itself cause autosomal-dominant pseudohypoaldosteronism type I with severe hypotension [17].
Adrenocortical adenomas can lead to the autonomous secretion of aldosterone responsible for primary aldosteronism and secondary arterial hypertension. Notably, somatic mutations in genes regulating intracellular ionic homeostasis and membrane potential are a recurrent finding in aldosterone-producing adenomas [1, 9, 44, 65]. As β‑catenin mutations are frequent in both aldosterone- and cortisol-producing adenomas [55], the Wnt/β-catenin pathway appears to play an important role in adrenal tumorigenesis.

Common variants and renal NaCl uptake

The numerous monogenic disorders caused by mutations in genes involved in NaCl homeostasis outlined above and the potency of antihypertensive agents such as thiazides, ACE inhibitors and AT1 and mineralocorticoid receptor blockers in the treatment of hypertension confirm the concept that hypertension is primarily a renal disorder characterized by abnormal handling of sodium [18] (and chloride). As another proof of concept, it has been shown that rare heterozygous mutations in the genes encoding NCC, NKCC2, and ROMK reduce blood pressure in participants of the Framingham Heart Study [25]. Whether more common variants in genes involved in NaCl homeostasis play a role in blood pressure control in the larger population is not yet finally solved. Several genome-wide association studies (GWAS) have been carried out to detect such common genetic variations with modest effect sizes. The largest cardiovascular genetic association study to date, with over 1 million participants, demonstrated more than 900 different genetic loci [12]. Because of the small effects, causal relationships are often difficult to establish. Notably, one of these variants lies in the promoter region of UMOD [40], which is specifically expressed in the thick ascending limb. Mouse studies suggest a functional link between UMOD and NKCC2, because NKCC2-mediated transport was reduced in Umod knockout mice [38]. Another SNP maps close to SLC4A7 encoding the electroneutral sodium-bicarbonate co-transporter 1 [22], which is also expressed in the thick ascending limb. Functional data, however, may point to a vascular effect, because this SNP was associated with altered SLC4A7 expression and defective pH regulation in vascular smooth muscle cells [39]. NEDD4L, which encodes an ubiquitin ligase regulating the cell surface expression of ENaC by targeting it for degradation, may also contribute to blood pressure [36]. Finally, variants in NPR3 encoding the natriuretic peptide receptor C, which controls the clearance of natriuretic peptides [43], have been linked with variation in blood pressure regulation [22, 26].

Future

Mendelian genetics helped to identify molecules that are crucial for renal NaCl homeostasis. Although their loss can cause extreme perturbations of blood pressure, the respective disorders are very rare and there is currently limited evidence that variants in the same genes play a common role in the determination of BP in the general population. Although the heritability of blood pressure from family studies is assumed to account for up to 50% [27], the collective effect of all loci identified by GWAS is thought to explain only roughly 2% of blood pressure heritability [22]. Therefore, the molecular mechanisms leading to hypertension are still poorly understood in the majority of patients. In addition to the few loci that can be linked to NaCl handling, the vasculature appears to be an important target organ in the pathogenesis of hypertension. Whether such genetic risk scores based on GWAS data prove to be clinically useful, is as of yet unclear. From the current data, recommendations for lifestyle management should be for the whole population, rather than targeted using genetic information.

Compliance with ethical guidelines

Conflict of interest

J. C. Hennings and C.A. Hübner declare that they have no competing interests.
All procedures performed in studies involving human participants were in accordance with the ethical standards of the institutional and/or national research committee and with the 1975 Declaration of Helsinki and its later amendments or comparable ethical standards. Informed consent was obtained from all individual participants included in the study. Additional informed consent was obtained from all patients for whom identifying information is included in this article. All institutional and national guidelines for the care and use of laboratory animals were followed. This article does not contain any studies with human or animal subjects.
Open Access. This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

e.Med Pädiatrie

Kombi-Abonnement

Mit e.Med Pädiatrie erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Pädiatrie, den Premium-Inhalten der pädiatrischen Fachzeitschriften, inklusive einer gedruckten Pädiatrie-Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Azizan EA, Poulsen H, Tuluc P et al (2013) Somatic mutations in ATP1A1 and CACNA1D underlie a common subtype of adrenal hypertension. Nat Genet 45:1055–1060PubMedCrossRef Azizan EA, Poulsen H, Tuluc P et al (2013) Somatic mutations in ATP1A1 and CACNA1D underlie a common subtype of adrenal hypertension. Nat Genet 45:1055–1060PubMedCrossRef
2.
Zurück zum Zitat Birkenhager R, Otto E, Schurmann MJ et al (2001) Mutation of BSND causes Bartter syndrome with sensorineural deafness and kidney failure. Nat Genet 29:310–314PubMedCrossRef Birkenhager R, Otto E, Schurmann MJ et al (2001) Mutation of BSND causes Bartter syndrome with sensorineural deafness and kidney failure. Nat Genet 29:310–314PubMedCrossRef
3.
Zurück zum Zitat Bockenhauer D, Feather S, Stanescu HC et al (2009) Epilepsy, ataxia, sensorineural deafness, tubulopathy, and KCNJ10 mutations. N Engl J Med 360:1960–1970PubMedPubMedCentralCrossRef Bockenhauer D, Feather S, Stanescu HC et al (2009) Epilepsy, ataxia, sensorineural deafness, tubulopathy, and KCNJ10 mutations. N Engl J Med 360:1960–1970PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat Boyden LM, Choi M, Choate KA et al (2012) Mutations in kelch-like 3 and cullin 3 cause hypertension and electrolyte abnormalities. Nature 482:98–102PubMedPubMedCentralCrossRef Boyden LM, Choi M, Choate KA et al (2012) Mutations in kelch-like 3 and cullin 3 cause hypertension and electrolyte abnormalities. Nature 482:98–102PubMedPubMedCentralCrossRef
5.
Zurück zum Zitat Burt VL, Whelton P, Roccella EJ et al (1995) Prevalence of hypertension in the US adult population. Results from the Third National Health and Nutrition Examination Survey, 1988–1991. Hypertension 25:305–313CrossRefPubMed Burt VL, Whelton P, Roccella EJ et al (1995) Prevalence of hypertension in the US adult population. Results from the Third National Health and Nutrition Examination Survey, 1988–1991. Hypertension 25:305–313CrossRefPubMed
6.
Zurück zum Zitat Canessa CM, Schild L, Buell G et al (1994) Amiloride-sensitive epithelial Na+ channel is made of three homologous subunits. Nature 367:463–467PubMedCrossRef Canessa CM, Schild L, Buell G et al (1994) Amiloride-sensitive epithelial Na+ channel is made of three homologous subunits. Nature 367:463–467PubMedCrossRef
7.
Zurück zum Zitat Chambrey R, Kurth I, Peti-Peterdi J et al (2013) Renal intercalated cells are rather energized by a proton than a sodium pump. Proc Natl Acad Sci USA 110:7928–7933PubMedCrossRefPubMedCentral Chambrey R, Kurth I, Peti-Peterdi J et al (2013) Renal intercalated cells are rather energized by a proton than a sodium pump. Proc Natl Acad Sci USA 110:7928–7933PubMedCrossRefPubMedCentral
8.
Zurück zum Zitat Chang SS, Grunder S, Hanukoglu A et al (1996) Mutations in subunits of the epithelial sodium channel cause salt wasting with hyperkalaemic acidosis, pseudohypoaldosteronism type 1. Nat Genet 12:248–253PubMedCrossRef Chang SS, Grunder S, Hanukoglu A et al (1996) Mutations in subunits of the epithelial sodium channel cause salt wasting with hyperkalaemic acidosis, pseudohypoaldosteronism type 1. Nat Genet 12:248–253PubMedCrossRef
9.
Zurück zum Zitat Choi M, Scholl UI, Yue P et al (2011) K+ channel mutations in adrenal aldosterone-producing adenomas and hereditary hypertension. Science 331:768–772PubMedPubMedCentralCrossRef Choi M, Scholl UI, Yue P et al (2011) K+ channel mutations in adrenal aldosterone-producing adenomas and hereditary hypertension. Science 331:768–772PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat Cornelius RJ, Si J, Cuevas CA et al (2018) Renal COP9 signalosome deficiency alters CUL3-KLHL3-WNK signaling pathway. J Am Soc Nephrol 29:2627–2640PubMedPubMedCentral Cornelius RJ, Si J, Cuevas CA et al (2018) Renal COP9 signalosome deficiency alters CUL3-KLHL3-WNK signaling pathway. J Am Soc Nephrol 29:2627–2640PubMedPubMedCentral
11.
Zurück zum Zitat Estevez R, Boettger T, Stein V et al (2001) Barttin is a Cl-channel beta-subunit crucial for renal Cl-reabsorption and inner ear K+ secretion. Nature 414:558–561CrossRefPubMed Estevez R, Boettger T, Stein V et al (2001) Barttin is a Cl-channel beta-subunit crucial for renal Cl-reabsorption and inner ear K+ secretion. Nature 414:558–561CrossRefPubMed
12.
Zurück zum Zitat Evangelou E, Warren HR, Mosen-Ansorena D et al (2018) Genetic analysis of over 1 million people identifies 535 new loci associated with blood pressure traits. Nat Genet 50:1412–1425PubMedPubMedCentralCrossRef Evangelou E, Warren HR, Mosen-Ansorena D et al (2018) Genetic analysis of over 1 million people identifies 535 new loci associated with blood pressure traits. Nat Genet 50:1412–1425PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Everett LA, Glaser B, Beck JC et al (1997) Pendred syndrome is caused by mutations in a putative sulphate transporter gene (PDS). Nat Genet 17:411–422PubMedCrossRef Everett LA, Glaser B, Beck JC et al (1997) Pendred syndrome is caused by mutations in a putative sulphate transporter gene (PDS). Nat Genet 17:411–422PubMedCrossRef
14.
Zurück zum Zitat Gamba G, Miyanoshita A, Lombardi M et al (1994) Molecular cloning, primary structure, and characterization of two members of the mammalian electroneutral sodium-(potassium)-chloride cotransporter family expressed in kidney. J Biol Chem 269:17713–17722PubMed Gamba G, Miyanoshita A, Lombardi M et al (1994) Molecular cloning, primary structure, and characterization of two members of the mammalian electroneutral sodium-(potassium)-chloride cotransporter family expressed in kidney. J Biol Chem 269:17713–17722PubMed
15.
Zurück zum Zitat Gamba G, Saltzberg SN, Lombardi M et al (1993) Primary structure and functional expression of a cDNA encoding the thiazide-sensitive, electroneutral sodium-chloride cotransporter. Proc Natl Acad Sci USA 90:2749–2753PubMedCrossRefPubMedCentral Gamba G, Saltzberg SN, Lombardi M et al (1993) Primary structure and functional expression of a cDNA encoding the thiazide-sensitive, electroneutral sodium-chloride cotransporter. Proc Natl Acad Sci USA 90:2749–2753PubMedCrossRefPubMedCentral
16.
Zurück zum Zitat Gawenis LR, Bradford EM, Prasad V et al (2007) Colonic anion secretory defects and metabolic acidosis in mice lacking the NBC1 Na+/HCO3 – cotransporter. J Biol Chem 282:9042–9052PubMedCrossRef Gawenis LR, Bradford EM, Prasad V et al (2007) Colonic anion secretory defects and metabolic acidosis in mice lacking the NBC1 Na+/HCO3 cotransporter. J Biol Chem 282:9042–9052PubMedCrossRef
17.
Zurück zum Zitat Geller DS, Rodriguez-Soriano J, Vallo Boado A et al (1998) Mutations in the mineralocorticoid receptor gene cause autosomal dominant pseudohypoaldosteronism type I. Nat Genet 19:279–281PubMedCrossRef Geller DS, Rodriguez-Soriano J, Vallo Boado A et al (1998) Mutations in the mineralocorticoid receptor gene cause autosomal dominant pseudohypoaldosteronism type I. Nat Genet 19:279–281PubMedCrossRef
18.
Zurück zum Zitat Guyton AC (1991) Blood pressure control—special role of the kidneys and body fluids. Science 252:1813–1816PubMedCrossRef Guyton AC (1991) Blood pressure control—special role of the kidneys and body fluids. Science 252:1813–1816PubMedCrossRef
19.
Zurück zum Zitat Hansson JH, Nelson-Williams C, Suzuki H et al (1995) Hypertension caused by a truncated epithelial sodium channel gamma subunit: genetic heterogeneity of Liddle syndrome. Nat Genet 11:76–82PubMedCrossRef Hansson JH, Nelson-Williams C, Suzuki H et al (1995) Hypertension caused by a truncated epithelial sodium channel gamma subunit: genetic heterogeneity of Liddle syndrome. Nat Genet 11:76–82PubMedCrossRef
20.
Zurück zum Zitat Hennings JC, Andrini O, Picard N et al (2017) The ClC-K2 chloride channel is critical for salt handling in the distal nephron. J Am Soc Nephrol 28:209–217CrossRefPubMed Hennings JC, Andrini O, Picard N et al (2017) The ClC-K2 chloride channel is critical for salt handling in the distal nephron. J Am Soc Nephrol 28:209–217CrossRefPubMed
21.
Zurück zum Zitat Igarashi T, Inatomi J, Sekine T et al (1999) Mutations in SLC4A4 cause permanent isolated proximal renal tubular acidosis with ocular abnormalities. Nat Genet 23:264–266PubMedCrossRef Igarashi T, Inatomi J, Sekine T et al (1999) Mutations in SLC4A4 cause permanent isolated proximal renal tubular acidosis with ocular abnormalities. Nat Genet 23:264–266PubMedCrossRef
22.
Zurück zum Zitat International Consortium for Blood Pressure Genome-Wide Association S, Ehret GB, Munroe PB et al (2011) Genetic variants in novel pathways influence blood pressure and cardiovascular disease risk. Nature 478:103–109CrossRef International Consortium for Blood Pressure Genome-Wide Association S, Ehret GB, Munroe PB et al (2011) Genetic variants in novel pathways influence blood pressure and cardiovascular disease risk. Nature 478:103–109CrossRef
23.
Zurück zum Zitat Jacques T, Picard N, Miller RL et al (2013) Overexpression of pendrin in intercalated cells produces chloride-sensitive hypertension. J Am Soc Nephrol 24:1104–1113PubMedPubMedCentralCrossRef Jacques T, Picard N, Miller RL et al (2013) Overexpression of pendrin in intercalated cells produces chloride-sensitive hypertension. J Am Soc Nephrol 24:1104–1113PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Janecke AR, Heinz-Erian P, Yin J et al (2015) Reduced sodium/proton exchanger NHE3 activity causes congenital sodium diarrhea. Hum Mol Genet 24:6614–6623PubMedPubMedCentralCrossRef Janecke AR, Heinz-Erian P, Yin J et al (2015) Reduced sodium/proton exchanger NHE3 activity causes congenital sodium diarrhea. Hum Mol Genet 24:6614–6623PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Ji W, Foo JN, O’roak BJ et al (2008) Rare independent mutations in renal salt handling genes contribute to blood pressure variation. Nat Genet 40:592–599PubMedPubMedCentralCrossRef Ji W, Foo JN, O’roak BJ et al (2008) Rare independent mutations in renal salt handling genes contribute to blood pressure variation. Nat Genet 40:592–599PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Kato N, Takeuchi F, Tabara Y et al (2011) Meta-analysis of genome-wide association studies identifies common variants associated with blood pressure variation in east Asians. Nat Genet 43:531–538PubMedPubMedCentralCrossRef Kato N, Takeuchi F, Tabara Y et al (2011) Meta-analysis of genome-wide association studies identifies common variants associated with blood pressure variation in east Asians. Nat Genet 43:531–538PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Kupper N, Willemsen G, Riese H et al (2005) Heritability of daytime ambulatory blood pressure in an extended twin design. Hypertension 45:80–85PubMedCrossRef Kupper N, Willemsen G, Riese H et al (2005) Heritability of daytime ambulatory blood pressure in an extended twin design. Hypertension 45:80–85PubMedCrossRef
28.
Zurück zum Zitat Laghmani K, Beck BB, Yang SS et al (2016) Polyhydramnios, transient antenatal Bartter’s syndrome, and MAGED2 mutations. N Engl J Med 374:1853–1863PubMedCrossRef Laghmani K, Beck BB, Yang SS et al (2016) Polyhydramnios, transient antenatal Bartter’s syndrome, and MAGED2 mutations. N Engl J Med 374:1853–1863PubMedCrossRef
29.
Zurück zum Zitat Leviel F, Hubner CA, Houillier P et al (2010) The Na+-dependent chloride-bicarbonate exchanger SLC4A8 mediates an electroneutral Na+ reabsorption process in the renal cortical collecting ducts of mice. J Clin Invest 120:1627–1635PubMedPubMedCentralCrossRef Leviel F, Hubner CA, Houillier P et al (2010) The Na+-dependent chloride-bicarbonate exchanger SLC4A8 mediates an electroneutral Na+ reabsorption process in the renal cortical collecting ducts of mice. J Clin Invest 120:1627–1635PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Lewington S, Clarke R, Qizilbash N et al (2002) Age-specific relevance of usual blood pressure to vascular mortality: a meta-analysis of individual data for one million adults in 61 prospective studies. Lancet 360:1903–1913PubMedCrossRef Lewington S, Clarke R, Qizilbash N et al (2002) Age-specific relevance of usual blood pressure to vascular mortality: a meta-analysis of individual data for one million adults in 61 prospective studies. Lancet 360:1903–1913PubMedCrossRef
31.
Zurück zum Zitat Lifton RP, Dluhy RG, Powers M et al (1992) A chimaeric 11 beta-hydroxylase/aldosterone synthase gene causes glucocorticoid-remediable aldosteronism and human hypertension. Nature 355:262–265PubMedCrossRef Lifton RP, Dluhy RG, Powers M et al (1992) A chimaeric 11 beta-hydroxylase/aldosterone synthase gene causes glucocorticoid-remediable aldosteronism and human hypertension. Nature 355:262–265PubMedCrossRef
32.
Zurück zum Zitat Lifton RP, Gharavi AG, Geller DS (2001) Molecular mechanisms of human hypertension. Cell 104:545–556PubMedCrossRef Lifton RP, Gharavi AG, Geller DS (2001) Molecular mechanisms of human hypertension. Cell 104:545–556PubMedCrossRef
33.
Zurück zum Zitat Lim SS, Vos T, Flaxman AD et al (2012) A comparative risk assessment of burden of disease and injury attributable to 67 risk factors and risk factor clusters in 21 regions, 1990–2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet 380:2224–2260PubMedPubMedCentralCrossRef Lim SS, Vos T, Flaxman AD et al (2012) A comparative risk assessment of burden of disease and injury attributable to 67 risk factors and risk factor clusters in 21 regions, 1990–2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet 380:2224–2260PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Luft FC, Zemel MB, Sowers JA et al (1990) Sodium bicarbonate and sodium chloride: effects on blood pressure and electrolyte homeostasis in normal and hypertensive man. J Hypertens 8:663–670PubMedCrossRef Luft FC, Zemel MB, Sowers JA et al (1990) Sodium bicarbonate and sodium chloride: effects on blood pressure and electrolyte homeostasis in normal and hypertensive man. J Hypertens 8:663–670PubMedCrossRef
35.
Zurück zum Zitat Madeo AC, Manichaikul A, Pryor SP et al (2009) Do mutations of the Pendred syndrome gene, SLC26A4, confer resistance to asthma and hypertension? J Med Genet 46:405–406PubMedCrossRef Madeo AC, Manichaikul A, Pryor SP et al (2009) Do mutations of the Pendred syndrome gene, SLC26A4, confer resistance to asthma and hypertension? J Med Genet 46:405–406PubMedCrossRef
36.
Zurück zum Zitat McDonough CW, Burbage SE, Duarte JD et al (2013) Association of variants in NEDD4L with blood pressure response and adverse cardiovascular outcomes in hypertensive patients treated with thiazide diuretics. J Hypertens 31:698–704PubMedPubMedCentralCrossRef McDonough CW, Burbage SE, Duarte JD et al (2013) Association of variants in NEDD4L with blood pressure response and adverse cardiovascular outcomes in hypertensive patients treated with thiazide diuretics. J Hypertens 31:698–704PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Meneton P, Loffing J, Warnock DG (2004) Sodium and potassium handling by the aldosterone-sensitive distal nephron: the pivotal role of the distal and connecting tubule. Am J Physiol Renal Physiol 287:F593–F601PubMedCrossRef Meneton P, Loffing J, Warnock DG (2004) Sodium and potassium handling by the aldosterone-sensitive distal nephron: the pivotal role of the distal and connecting tubule. Am J Physiol Renal Physiol 287:F593–F601PubMedCrossRef
38.
Zurück zum Zitat Mutig K, Kahl T, Saritas T et al (2011) Activation of the bumetanide-sensitive Na+,K+,2Cl-cotransporter (NKCC2) is facilitated by Tamm-Horsfall protein in a chloride-sensitive manner. J Biol Chem 286:30200–30210PubMedPubMedCentralCrossRef Mutig K, Kahl T, Saritas T et al (2011) Activation of the bumetanide-sensitive Na+,K+,2Cl-cotransporter (NKCC2) is facilitated by Tamm-Horsfall protein in a chloride-sensitive manner. J Biol Chem 286:30200–30210PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Ng FL, Boedtkjer E, Witkowska K et al (2017) Increased NBCn1 expression, Na+/HCO3 - co-transport and intracellular pH in human vascular smooth muscle cells with a risk allele for hypertension. Hum Mol Genet 26:989–1002PubMedPubMedCentral Ng FL, Boedtkjer E, Witkowska K et al (2017) Increased NBCn1 expression, Na+/HCO3 - co-transport and intracellular pH in human vascular smooth muscle cells with a risk allele for hypertension. Hum Mol Genet 26:989–1002PubMedPubMedCentral
40.
Zurück zum Zitat Padmanabhan S, Melander O, Johnson T et al (2010) Genome-wide association study of blood pressure extremes identifies variant near UMOD associated with hypertension. PLoS Genet 6:e1001177PubMedPubMedCentralCrossRef Padmanabhan S, Melander O, Johnson T et al (2010) Genome-wide association study of blood pressure extremes identifies variant near UMOD associated with hypertension. PLoS Genet 6:e1001177PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Pascoe L, Curnow KM, Slutsker L et al (1992) Mutations in the human CYP11B2 (aldosterone synthase) gene causing corticosterone methyloxidase II deficiency. Proc Natl Acad Sci USA 89:4996–5000PubMedCrossRefPubMedCentral Pascoe L, Curnow KM, Slutsker L et al (1992) Mutations in the human CYP11B2 (aldosterone synthase) gene causing corticosterone methyloxidase II deficiency. Proc Natl Acad Sci USA 89:4996–5000PubMedCrossRefPubMedCentral
42.
Zurück zum Zitat Reichold M, Zdebik AA, Lieberer E et al (2010) KCNJ10 gene mutations causing EAST syndrome (epilepsy, ataxia, sensorineural deafness, and tubulopathy) disrupt channel function. Proc Natl Acad Sci USA 107:14490–14495PubMedCrossRefPubMedCentral Reichold M, Zdebik AA, Lieberer E et al (2010) KCNJ10 gene mutations causing EAST syndrome (epilepsy, ataxia, sensorineural deafness, and tubulopathy) disrupt channel function. Proc Natl Acad Sci USA 107:14490–14495PubMedCrossRefPubMedCentral
43.
Zurück zum Zitat Ren M, Ng FL, Warren HR et al (2018) The biological impact of blood pressure-associated genetic variants in the natriuretic peptide receptor C gene on human vascular smooth muscle. Hum Mol Genet 27:199–210PubMedCrossRef Ren M, Ng FL, Warren HR et al (2018) The biological impact of blood pressure-associated genetic variants in the natriuretic peptide receptor C gene on human vascular smooth muscle. Hum Mol Genet 27:199–210PubMedCrossRef
44.
Zurück zum Zitat Scholl UI, Stolting G, Nelson-Williams C et al (2015) Recurrent gain of function mutation in calcium channel CACNA1H causes early-onset hypertension with primary aldosteronism. Elife 4:e6315PubMedPubMedCentralCrossRef Scholl UI, Stolting G, Nelson-Williams C et al (2015) Recurrent gain of function mutation in calcium channel CACNA1H causes early-onset hypertension with primary aldosteronism. Elife 4:e6315PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Schultheis PJ, Clarke LL, Meneton P et al (1998) Renal and intestinal absorptive defects in mice lacking the NHE3 Na+/H+ exchanger. Nat Genet 19:282–285PubMedCrossRef Schultheis PJ, Clarke LL, Meneton P et al (1998) Renal and intestinal absorptive defects in mice lacking the NHE3 Na+/H+ exchanger. Nat Genet 19:282–285PubMedCrossRef
46.
Zurück zum Zitat Seidel E, Scholl UI (2017) Genetic mechanisms of human hypertension and their implications for blood pressure physiology. Physiol Genomics 49:630–652PubMedCrossRef Seidel E, Scholl UI (2017) Genetic mechanisms of human hypertension and their implications for blood pressure physiology. Physiol Genomics 49:630–652PubMedCrossRef
47.
Zurück zum Zitat Shibata S, Arroyo JP, Castaneda-Bueno M et al (2014) Angiotensin II signaling via protein kinase C phosphorylates Kelch-like 3, preventing WNK4 degradation. Proc Natl Acad Sci USA 111:15556–15561PubMedCrossRefPubMedCentral Shibata S, Arroyo JP, Castaneda-Bueno M et al (2014) Angiotensin II signaling via protein kinase C phosphorylates Kelch-like 3, preventing WNK4 degradation. Proc Natl Acad Sci USA 111:15556–15561PubMedCrossRefPubMedCentral
48.
Zurück zum Zitat Shimkets RA, Warnock DG, Bositis CM et al (1994) Liddle’s syndrome: heritable human hypertension caused by mutations in the beta subunit of the epithelial sodium channel. Cell 79:407–414PubMedCrossRef Shimkets RA, Warnock DG, Bositis CM et al (1994) Liddle’s syndrome: heritable human hypertension caused by mutations in the beta subunit of the epithelial sodium channel. Cell 79:407–414PubMedCrossRef
49.
Zurück zum Zitat Simon DB, Bindra RS, Mansfield TA et al (1997) Mutations in the chloride channel gene, CLCNKB, cause Bartter’s syndrome type III. Nat Genet 17:171–178PubMedCrossRef Simon DB, Bindra RS, Mansfield TA et al (1997) Mutations in the chloride channel gene, CLCNKB, cause Bartter’s syndrome type III. Nat Genet 17:171–178PubMedCrossRef
50.
Zurück zum Zitat Simon DB, Karet FE, Hamdan JM et al (1996) Bartter’s syndrome, hypokalaemic alkalosis with hypercalciuria, is caused by mutations in the Na-K-2Cl cotransporter NKCC2. Nat Genet 13:183–188PubMedCrossRef Simon DB, Karet FE, Hamdan JM et al (1996) Bartter’s syndrome, hypokalaemic alkalosis with hypercalciuria, is caused by mutations in the Na-K-2Cl cotransporter NKCC2. Nat Genet 13:183–188PubMedCrossRef
51.
Zurück zum Zitat Simon DB, Karet FE, Rodriguez-Soriano J et al (1996) Genetic heterogeneity of Bartter’s syndrome revealed by mutations in the K+ channel, ROMK. Nat Genet 14:152–156PubMedCrossRef Simon DB, Karet FE, Rodriguez-Soriano J et al (1996) Genetic heterogeneity of Bartter’s syndrome revealed by mutations in the K+ channel, ROMK. Nat Genet 14:152–156PubMedCrossRef
52.
Zurück zum Zitat Simon DB, Nelson-Williams C, Bia MJ et al (1996) Gitelman’s variant of Bartter’s syndrome, inherited hypokalaemic alkalosis, is caused by mutations in the thiazide-sensitive Na-Cl cotransporter. Nat Genet 12:24–30PubMedCrossRef Simon DB, Nelson-Williams C, Bia MJ et al (1996) Gitelman’s variant of Bartter’s syndrome, inherited hypokalaemic alkalosis, is caused by mutations in the thiazide-sensitive Na-Cl cotransporter. Nat Genet 12:24–30PubMedCrossRef
53.
Zurück zum Zitat Stewart PM, Wallace AM, Valentino R et al (1987) Mineralocorticoid activity of liquorice: 11-beta-hydroxysteroid dehydrogenase deficiency comes of age. Lancet 2:821–824CrossRefPubMed Stewart PM, Wallace AM, Valentino R et al (1987) Mineralocorticoid activity of liquorice: 11-beta-hydroxysteroid dehydrogenase deficiency comes of age. Lancet 2:821–824CrossRefPubMed
54.
Zurück zum Zitat Strautnieks SS, Thompson RJ, Gardiner RM et al (1996) A novel splice-site mutation in the gamma subunit of the epithelial sodium channel gene in three pseudohypoaldosteronism type 1 families. Nat Genet 13:248–250PubMedCrossRef Strautnieks SS, Thompson RJ, Gardiner RM et al (1996) A novel splice-site mutation in the gamma subunit of the epithelial sodium channel gene in three pseudohypoaldosteronism type 1 families. Nat Genet 13:248–250PubMedCrossRef
55.
Zurück zum Zitat Tadjine M, Lampron A, Ouadi L et al (2008) Frequent mutations of beta-catenin gene in sporadic secreting adrenocortical adenomas. Clin Endocrinol (Oxf) 68:264–270 Tadjine M, Lampron A, Ouadi L et al (2008) Frequent mutations of beta-catenin gene in sporadic secreting adrenocortical adenomas. Clin Endocrinol (Oxf) 68:264–270
56.
Zurück zum Zitat Tomita K, Pisano JJ, Burg MB et al (1986) Effects of vasopressin and bradykinin on anion transport by the rat cortical collecting duct. Evidence for an electroneutral sodium chloride transport pathway. J Clin Invest 77:136–141PubMedPubMedCentralCrossRef Tomita K, Pisano JJ, Burg MB et al (1986) Effects of vasopressin and bradykinin on anion transport by the rat cortical collecting duct. Evidence for an electroneutral sodium chloride transport pathway. J Clin Invest 77:136–141PubMedPubMedCentralCrossRef
57.
Zurück zum Zitat Tse CM, Brant SR, Walker MS et al (1992) Cloning and sequencing of a rabbit cDNA encoding an intestinal and kidney-specific Na+/H+ exchanger isoform (NHE-3). J Biol Chem 267:9340–9346PubMed Tse CM, Brant SR, Walker MS et al (1992) Cloning and sequencing of a rabbit cDNA encoding an intestinal and kidney-specific Na+/H+ exchanger isoform (NHE-3). J Biol Chem 267:9340–9346PubMed
58.
Zurück zum Zitat Verlander JW, Hassell KA, Royaux IE et al (2003) Deoxycorticosterone upregulates PDS (Slc26a4) in mouse kidney: role of pendrin in mineralocorticoid-induced hypertension. Hypertension 42:356–362PubMedCrossRef Verlander JW, Hassell KA, Royaux IE et al (2003) Deoxycorticosterone upregulates PDS (Slc26a4) in mouse kidney: role of pendrin in mineralocorticoid-induced hypertension. Hypertension 42:356–362PubMedCrossRef
59.
Zurück zum Zitat Wall SM, Kim YH, Stanley L et al (2004) NaCl restriction upregulates renal Slc26a4 through subcellular redistribution: role in Cl-conservation. Hypertension 44:982–987PubMedCrossRef Wall SM, Kim YH, Stanley L et al (2004) NaCl restriction upregulates renal Slc26a4 through subcellular redistribution: role in Cl-conservation. Hypertension 44:982–987PubMedCrossRef
60.
Zurück zum Zitat Watanabe S, Fukumoto S, Chang H et al (2002) Association between activating mutations of calcium-sensing receptor and Bartter’s syndrome. Lancet 360:692–694PubMedCrossRef Watanabe S, Fukumoto S, Chang H et al (2002) Association between activating mutations of calcium-sensing receptor and Bartter’s syndrome. Lancet 360:692–694PubMedCrossRef
61.
Zurück zum Zitat Whelton PK, Carey RM, Aronow WS et al (2018) 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. J Am Coll Cardiol 71:e127–e248PubMedCrossRef Whelton PK, Carey RM, Aronow WS et al (2018) 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. J Am Coll Cardiol 71:e127–e248PubMedCrossRef
62.
Zurück zum Zitat Whitescarver SA, Ott CE, Jackson BA et al (1984) Salt-sensitive hypertension: contribution of chloride. Science 223:1430–1432PubMedCrossRef Whitescarver SA, Ott CE, Jackson BA et al (1984) Salt-sensitive hypertension: contribution of chloride. Science 223:1430–1432PubMedCrossRef
63.
Zurück zum Zitat Wilson FH, Disse-Nicodeme S, Choate KA et al (2001) Human hypertension caused by mutations in WNK kinases. Science 293:1107–1112PubMedCrossRef Wilson FH, Disse-Nicodeme S, Choate KA et al (2001) Human hypertension caused by mutations in WNK kinases. Science 293:1107–1112PubMedCrossRef
64.
65.
Zurück zum Zitat Zennaro MC, Boulkroun S, Fernandes-Rosa F (2017) Genetic causes of functional adrenocortical adenomas. Endocr Rev 38:516–537PubMedCrossRef Zennaro MC, Boulkroun S, Fernandes-Rosa F (2017) Genetic causes of functional adrenocortical adenomas. Endocr Rev 38:516–537PubMedCrossRef
Metadaten
Titel
Disorders of renal NaCl transport and implications for blood pressure regulation
verfasst von
J. Christopher Hennings
Prof. Dr. Christian A. Hübner
Publikationsdatum
01.02.2019
Verlag
Springer Medizin
Erschienen in
medizinische genetik / Ausgabe 1/2019
Print ISSN: 0936-5931
Elektronische ISSN: 1863-5490
DOI
https://doi.org/10.1007/s11825-019-0232-8

Weitere Artikel der Ausgabe 1/2019

medizinische genetik 1/2019 Zur Ausgabe

Abstracts

Abstracts

Mitteilungen des BVDH

Mitteilungen des BVDH

30. Jahrestagung der Deutschen Gesellschaft für Humangenetik

30. Jahrestagung der Deutschen Gesellschaft für Humangenetik

Mitteilungen der GfH

Mitteilungen der GfH