Genetically Based Kidney Transport Disorders


The coming of age of clinical chemistry in the latter half of the 20th century, bringing with it the routine measurement of electrolytes and minerals in patient samples, produced descriptions of distinct inherited syndromes of abnormal tubular transport in the kidney. Clinical investigation led to speculation, often ingenious and sometimes controversial, regarding the underlying causes of these syndromes. More recently, the tools of molecular biology made possible the cloning of mutated genes found in patients with monogenic disorders of kidney tubular transport. These diseases represent experiments of nature, and for anyone interested in pathophysiology, and specifically for kidney physiologists, the insights they have revealed are exciting. Some provide gratifying confirmation of our existing knowledge of transport mechanisms along the nephron. Examples include mutations in diuretic-sensitive transporters in the Bartter and Gitelman syndromes. In other cases, positional cloning led to the discovery of previously unknown proteins, often surprising ones that appear to play important roles in epithelial transport. For example, the chloride transporter CLC-5 (gene name CLCN5 ), the tight junction claudin 16 (paracellin 1), and the phosphaturic hormone fibroblast growth factor 23 (FGF23) were discovered through positional cloning in the study of Dent disease, inherited hypomagnesemic hypercalciuria, and autosomal dominant hypophosphatemic rickets (ADHR), respectively.

Table 37.1 summarizes genetic diseases of kidney tubular transport for which the molecular basis is known. The diseases listed are explained by abnormalities in the corresponding gene product. They are all inherited in Mendelian fashion, either autosomal or X-linked, with the single exception of a syndrome of hypomagnesemia with maternal inheritance that results from mutation in a mitochondrial tRNA rather than in the nuclear genome.

TABLE 37.1
Molecular Bases of Genetic Disorders of Renal Transport
Inherited Disorder Defective Gene Product
Proximal Tubule
Glucose-galactose malabsorption syndrome Sodium-glucose transporter 1
Dibasic aminoaciduria Basolateral dibasic amino acid transporter (lysinuric protein intolerance)
XLHR PHEX
ADHR FGF23 (excess)SGKL
Autosomal recessive hypophosphatemic rickets DMP1ENPP1FAM20C
HHRH Sodium-phosphate cotransporter NPT2c
Familial hyperostosis-hyperphosphatemia FGF23 (deficiency)GalNac transferase 3Klotho (FGF23 co-receptor)
Fanconi syndromeAutosomal dominantAutosomal recessiveHereditary fructose intolerance Glycine amidinotransferasePeroxisomal enoyl-CoA Hydratase-L-3-hydroxyacyl-CoA dehydrogenase (L-PBE)Nuclear hormone receptor HNF4ASodium-phosphate cotransporter NPT2aNADH:ubiquinone oxidoreductase complex assembly factor 6Aldolase B
Fanconi-Bickel syndrome Facilitated GLUT2
Oculocerebrorenal syndrome of Lowe Inositol polyphosphate-5-phosphatase (OCRL1)
Dent disease (X-linked nephrolithiasis) Chloride transporter (ClC-5)Inositol polyphosphate-5-phosphatase (OCRL1)
Cystinuria Apical cystine-dibasic amino acid transporter rBATLight subunit of rBAT
Autosomal recessive proximal RTA Basolateral sodium-bicarbonate cotransporter NBC1
TAL of Loop of Henle
Bartter syndrome
Type I Bumetanide-sensitive Na-K-2Cl cotransporter NKCC2
Type II Apical potassium channel ROMK
Type III Basolateral chloride channel ClC-Kb
Type IV, with sensorineural deafness Barttin (ClC-Kb–associated protein)
Familial hypocalcemia with Bartter featuresType V, transient antenatal with polyhydramnios CaSR (activation)Melanoma antigen, Family D2 (MAGED2)
Familial hypomagnesemia with hypercalciuria and nephrocalcinosis
Without ocular abnormalities Claudin-16 (paracellin-1)
With ocular abnormalities Claudin-19
Familial hypocalciuric hypercalcemia a CaSR (inactivation)
Neonatal severe hyperparathyroidism b CaSR (inactivation)
Autosomal dominant hypercalciuric hypocalcemia CaSR (activation) (type 1)Gα 11 G-protein (type 2)
Familial juvenile hyperuricemic nephropathy Uromodulin (Tamm-Horsfall protein)
Distal Convoluted Tubule
Gitelman syndrome Thiazide-sensitive NaCl cotransporter NCC
Pseudohypoparathyroidism type Ia c Guanine nucleotide-binding protein (Gs)
Familial hypomagnesemia with secondary hypocalcemia TRPM6 cation channel d
Isolated recessive renal hypomagnesemia EGF
Autosomal dominant hypomagnesemia with hypocalciuria γ subunit of Na/K-ATPaseHNF1B transcription factorPCBD1 dimerization cofactor
SeSAME/EAST syndromes Kir4.1 potassium channel
Dominant hypomagnesemia with ataxia Kv1.1 potassium channel
Familial tubulopathy with hypomagnesemia Mitochondrial tRNAisoleucine
Dominant or recessive hypomagnesemia De novo severe infantile hypomagnesemia with seizures and intellectual disability CNNM2 (cyclin M2)α1 subunit of Na/K-ATPase
Collecting Duct
Liddle syndrome α, β, γ subunits of epithelial Na channel ENaC
Pseudohypoaldosteronism
Type 1
Autosomal recessive α, β, γ subunits of ENaC
Autosomal dominant (Geller syndrome) Mineralocorticoid (type I) receptor
Type 2 (Gordon syndrome) WNK1, WNK4 kinasesCullin-3 scaffold proteinKelch3 adaptor protein
Glucocorticoid-remediable aldosteronism 11β-hydroxylase and aldosterone synthase (chimeric gene) e
Syndrome of AME 11β-hydroxysteroid dehydrogenase type II
Distal RTA
Autosomal dominant Basolateral anion exchanger (AE1) (band 3 protein)
Autosomal recessive, with hemolytic anemia Basolateral anion exchanger (AE1) (band 3 protein)
Autosomal recessive (with hearing deficit) β1 subunit of proton ATPaseForkhead transcription factor FOXI1
Autosomal recessive (hearing deficit variable) α4 isoform of α subunit of proton ATPase
Carbonic anhydrase II deficiency f Carbonic anhydrase type II
Nephrogenic diabetes insipidus
X-linked AVP 2 (V2) receptor
Autosomal AQP-2 water channel

a Results from heterozygous mutation.

b Results from homozygous mutation.

c Gene also expressed in proximal tubule where functional abnormalities are clinically apparent.

d Gene also expressed in intestine.

e Gene expressed in adrenal gland.

f Clinical phenotype can be of proximal RTA, distal RTA, or combined. ADHR, Autosomal dominant hypophosphatemic rickets; AME, apparent mineralocorticoid excess; AQP-2, aquaporin 2; AVP, arginine vasopressin; CaSR, calcium-sensing receptor; DMP1, dentin matrix protein 1; EAST, epilepsy, ataxia, sensorineural deafness, and tubulopathy; EGF, epidermal growth factor; ENPP1, ectonucleotide pyrophosphatase/phosphodiesterase 1; FGF23, fibroblast growth factor 23; GLUT2, glucose transporter; GRA, glucocorticoid-remediable aldosteronism; HHRH, hereditary hypophosphatemic rickets with hypercalciuria; NCCT, sodium chloride cotransporter; NKCC2, Na + -K + -2Cl cotransporter; PHEX, phosphate-regulating gene with homologies to endopeptidases on the X chromosome; RTA, renal tubular acidosis; SeSAME, seizures, sensorineural deafness, ataxia, mental retardation, and electrolyte imbalance; TAL, thick ascending limb; XLHR, X-linked hypophosphatemic rickets.

Disorders of Proximal Tubular Transport Function

Selective Proximal Transport Defects

Sodium resorption in the proximal tubule occurs through secondary active transport processes in which the entry of sodium is coupled either to the entry of substrates (e.g., glucose, amino acids, or phosphate) or to the exit of protons. Autosomal recessive conditions of impaired transepithelial transport of glucose and dibasic amino acids have been shown to be caused by mutations in sodium-dependent transporters that are expressed in both kidney and intestine, resulting in urinary losses and intestinal malabsorption of these solutes. Other disorders with kidney-selective transport defects result from mutations in transporters expressed specifically in kidney.

Impaired Proximal Phosphate Reabsorption

Hypophosphatemic rickets can be inherited in X-linked, autosomal dominant, and autosomal recessive patterns. All three modes are associated with kidney phosphate wasting, with a reduced maximal transport capacity for phosphate. In all three, serum levels of the phosphate-regulating hormone (phosphatonin) FGF23 are elevated, and serum levels of 1,25-dihydroxyvitamin D are not elevated despite the stimulus of hypophosphatemia.

X-linked (dominant) hypophosphatemic rickets (XLHR) is the most common form of hereditary rickets. Mutations in XLHR involve a phosphate-regulating gene with homologies to a neutral endopeptidase on the X chromosome (PHEX) that is expressed in bone and is indirectly involved in the degradation and processing of FGF23. Elevations in FGF23 are sufficient to explain the kidney phosphate wasting, but other factors independent of FGF23 appear to contribute as well to the bone demineralization and rickets. Recent studies demonstrate the effectiveness of a monoclonal anti-FGF23 antibody, burosumab, to correct hypophosphatemia and improve bone mineralization in children with XLHR, further validating a central role for FGF23 in this disease.

The rare ADHR is associated with mutations in the gene encoding FGF23 that protects the phosphatonin from proteolytic cleavage. FGF23 disrupts kidney phosphate reabsorption by inhibiting expression of two genes encoding sodium-dependent phosphate transporters in proximal tubule, SLC34A1 (encoding the Na-dependent phosphate transporter Npt2a) and SLC34A3 (encoding Npt2c). FGF23 inhibits the 1-hydroxylation of 25-hydroxyvitamin D, likely explaining why hypophosphatemia in these three conditions is not associated with either elevated levels of 1,25-dihydroxyvitamin D or hypercalciuria. A single kindred has been reported with autosomal dominant hypophosphatemia and rickets with mutation in SGK3 , encoding a proximal tubular protein kinase SGKL.

Autosomal recessive inheritance of hypophosphatemic rickets has been reported in association with mutations in one of three genes. In ARHR type 1, the mutated gene is DMP1 , encoding the dentin matrix protein 1 (DMP-1), a bone matrix protein that appears to play a role with PHEX in regulating bone mineralization and FGF23 production. ARHR2 is associated with mutations in ENPP1 , encoding ectonucleotide pyrophosphatase/phosphodiesterase 1. Patients with either subtype resemble those with autosomal dominant (i.e., FGF23 mutations) and X-linked (PHEX mutations) hypophosphatemic rickets. ARHR3 is associated with mutations in FAM20C , encoding a protein kinase that phosphorylates FGF23; this subtype may also manifest features of osteosclerosis.

Hereditary hypophosphatemic rickets with hypercalciuria (HHRH), an autosomal recessive disorder, is different from XLHR, ADRH, and ARHR, all of which are associated with reduced urinary calcium excretion. In contrast, hypophosphatemia in HHRH is associated with appropriate elevations of 1,25-dihydroxyvitamin D levels, and FGF23 levels are normal or reduced. This profile in HHRH is consistent with a primary defect in phosphate transport and, indeed, the disease is associated with mutations in SLC34A3 , encoding the proximal tubule phosphate transporter Npt2c. Expression of this transporter, as well as that of SLC34A1 encoding the more abundant transporter Npt2a, responds to physiologic stimuli such as parathyroid hormone (PTH) and dietary phosphate. Knockout of the mouse homologue of SLC34A1 reproduces the features of human HHRH except for rickets, mouse knockout of SLC34A3 manifests hypercalcemia and hypercalciuria, but not hypophosphatemia, nephrocalcinosis, or rickets, and a double knockout of both genes produces mice with the full phenotype of hypophosphatemia, hypercalciuria, nephrocalcinosis, and rickets. In one human kindred, apparent autosomal dominant inheritance of HHRH was associated with compound heterozygosity for SLC34A3 and SLC34A1 .

Excessive Proximal Phosphate Reabsorption

Inherited hyperphosphatemia in the familial hyperostosis-hyperphosphatemia syndrome represents a mirror image of ADHR and XLHR, with excessive kidney phosphate reabsorption, persistent hyperphosphatemia, inappropriately normal levels of 1,25-dihydroxyvitamin D, and low levels of FGF23. This can result from mutations in one of three genes identified to date. These genes encode FGF23 itself, a Golgi-associated biosynthetic enzyme, N -acetylglucosaminyl (GalNac) transferase 3 that is involved in glycosylation of FGF23 and is necessary for its secretion, and Klotho, a transmembrane protein that complexes with the FGF23 receptor and regulates its affinity for FGF23. Together, these discoveries augment our understanding of the role of bone in the complex regulation of mineral metabolism.

Impaired Proximal Bicarbonate Transport

Selective proximal renal tubular acidosis (RTA) is inherited in an autosomal recessive manner and is associated with mutations that inactivate the basolateral sodium bicarbonate cotransporter NBC1, encoded by the gene SLC4A4 . These patients typically exhibit short stature and often suffer blindness from ocular abnormalities, including band keratopathy, cataracts, and glaucoma; these ocular manifestations probably are a consequence of impaired bicarbonate transport in the eye. Mutations in the SLC4A4 gene result in impaired transporter function or aberrant trafficking of the protein to the basolateral surface. This gene belongs to the same group as the gene encoding the anion exchanger AE1 (now designated SLC4A1 ), which is mutated in distal RTA. Proximal RTA can also be inherited in an autosomal dominant fashion, though the responsible gene(s) have not yet been identified.

Inherited Fanconi Syndrome: Hereditary Fructose Intolerance, Lowe Syndrome, and Dent Disease

The Fanconi syndrome represents a generalized impairment in reabsorptive function of the proximal tubule and comprises proximal RTA with aminoaciduria, glycosuria, hypouricemia, and hypophosphatemia. Some or all of these abnormalities are present in individual patients with Fanconi syndrome. Generalized Fanconi syndrome is genetically heterogeneous. It has been reported with autosomal dominant inheritance in association with mutations in genes encoding glycine amidinotransferase, the enzyme L-PBE, and the nuclear hormone receptor HNF4A (associated with MODY), and with recessive inheritance in the cases of genes encoding the sodium-phosphate transporter NaPi-IIa and the NADH:ubiquinone oxidoreductase complex assembly factor 6. Inherited causes of partial or complete Fanconi syndrome also include hereditary fructose intolerance, Lowe syndrome, and Dent disease.

Hereditary fructose intolerance is caused by mutations that result in deficiency of the aldolase B isoenzyme, which cleaves fructose-1-phosphate. Symptoms are precipitated by intake of sweets. Massive accumulation of fructose-1-phosphate occurs, leading to sequestration of inorganic phosphate and deficiency of adenosine triphosphate (ATP). Acute consequences can include hypoglycemic shock, severe abdominal symptoms, and impaired function of the Krebs cycle that produces metabolic acidosis; this is exacerbated by impaired kidney bicarbonate reabsorption. ATP deficiency leads to impaired proximal tubular function in general, including the full expression of the Fanconi syndrome with consequent rickets and stunted growth. ATP breakdown can be so dramatic as to produce hyperuricemia, as well as hypermagnesemia from the dissolution of the magnesium-ATP complex. Avoiding dietary sources of fructose can minimize acute symptoms and chronic consequences such as liver disease.

Characteristic features of the oculocerebrorenal syndrome of Lowe include congenital cataracts, mental retardation, muscular hypotonia, and the Fanconi syndrome. In contrast, Dent disease is confined to the kidney. In both syndromes, low-molecular-weight (LMW) proteinuria is a prominent feature along with other evidence of proximal tubulopathy such as glycosuria, aminoaciduria, and phosphaturia. One important and puzzling difference is that proximal RTA with growth retardation can be severe in patients with Lowe syndrome, but it is not a part of Dent disease. Some patients with Lowe syndrome or Dent disease may have rickets, which is thought to be a consequence of hypophosphatemia and, in Lowe syndrome, of acidosis as well. Hypercalciuria is a characteristic feature of Dent disease and is associated with nephrocalcinosis in most and kidney stones in many patients with Dent disease; nephrocalcinosis and nephrolithiasis are less common in Lowe syndrome. Kidney failure is common in both of these conditions, typically occurring in young adulthood in Dent disease and even earlier in patients with Lowe syndrome.

Dent disease is caused in most cases by mutations that inactivate the chloride transporter CLC-5. This transport protein is expressed in the proximal tubule, the medullary thick ascending limb (MTAL) of the loop of Henle, and the α-intercalated cells of the collecting tubule. In the cells of the proximal tubule, CLC-5 co-localizes with the H + -ATPase in subapical endosomes. These endosomes are important in the processing of proteins that are filtered at the glomerulus and taken up by the proximal tubule through adsorptive endocytosis. The activity of the H + -ATPase acidifies the endosomal space, releasing the proteins from membrane-binding sites and making them available for proteolytic degradation. CLC-5 mediates electrogenic exchange of chloride for protons in these endosomes, facilitating endosomal acidification. Mutations that inactivate CLC-5 in patients with Dent disease interfere with the mechanism for reabsorption of LMW proteins and explain the consistent finding of LMW proteinuria. Glycosuria, aminoaciduria, and phosphaturia occur commonly but less consistently and may reflect altered membrane protein recycling, as CLC-5 interacts directly with several trafficking proteins. Hypercalciuria appears largely to reflect a dysregulation of kidney 1-hydroxylation of 25(OH)-vitamin D.

Lowe syndrome is associated with mutations in OCRL1, which encodes a phosphatidylinositol-4,5-bisphosphate-5-phosphatase. In tubular epithelial cells, this phosphatase is localized to the trans -Golgi network, which plays an important role in directing proteins to the appropriate membrane. The CLC-5 protein and the OCRL1 phosphatase interact with the actin cytoskeleton and are involved in assembly of the endosomal apparatus. Similarities in the kidney features of these two syndromes may be the result of defective membrane trafficking. A subset of patients with mutations in OCRL1 have no cataracts or cerebral dysfunction and no RTA (“Dent 2” disease). Such mutations occur predominantly in the 5′ end of the gene and are critical to expression of the gene in the kidney, but apparently not to expression of transcripts in eye or brain.

Disorders of Transport in the Medullary Thick Ascending Limb of Henle

Bartter Syndrome

Solute transport in the MTAL involves the coordinated functions of a set of transport proteins depicted in Fig. 37.1 . These proteins are the loop diuretic-sensitive Na + -K + -2Cl cotransporter (NKCC2) and the renal outer medullary potassium channel (ROMK) on the apical surface of cells of the MTAL, and the chloride channel ClC-Kb on the basolateral surface. Optimal function of the ClC-Kb chloride channel requires interaction with a subunit called barttin . Mutations in any of the genes encoding these four proteins lead to the phenotype of Bartter syndrome. In addition, activation of the epithelial CaSR inhibits activity of the ROMK potassium channel. Mutations producing constitutive activation of the CaSR cause familial hypocalcemic hypercalciuria. Some patients with hypocalcemic hypercalciuria have the phenotype of Bartter syndrome, and mutations in the CaSR may be considered a fifth molecular cause of this syndrome. However, defects in these five genes still do not account for all patients with Bartter syndrome.

Fig. 37.1, Transport mechanisms in the thick ascending limb of the loop of Henle. Reabsorption of sodium chloride occurs through the electroneutral activity of the loop diuretic-sensitive Na + -K + -2Cl − cotransporter (NKCC2). Activity of the basolateral sodium-potassium adenosine triphosphatase (Na + ,K + -ATPase) provides the driving force for this transport and also generates a high intracellular concentration of potassium, which exits through the ATP-regulated apical potassium channel, ROMK. This ensures an adequate supply of potassium for the activity of the NKCC2 and also produces a lumen-positive electrical potential, which itself is the driving force for paracellular reabsorption of calcium, magnesium, and sodium ions through the tight junctions, involving the protein paracellin 1. Chloride transported into the cell by NKCC2 exits the basolateral side of the cell through the voltage-gated chloride channel, ClC-Kb. Activation of the extracellular calcium-sensing receptor, CaSR, inhibits solute transport in the TAL by inhibiting activity of the ROMK and possibly by other mechanisms. Mutations that inactivate the CaSR are associated with enhanced calcium transport and hypocalciuria in familial benign hypercalcemia, and mutations that activate the CaSR occur in patients with familial hypercalciuria with hypocalcemia. ROMK, Renal outer medullary potassium channel; TAL, thick ascending limb.

The ClC-Kb basolateral chloride channel provides the route for chloride exit to the interstitium. Flow of potassium through the ROMK channel ensures that potassium concentrations in the tubular lumen do not limit the activity of the NKCC2 while maintaining a positive electrical potential in the lumen of this nephron segment. This positive charge is the driving force for paracellular reabsorption of calcium and magnesium.

Bartter syndrome manifests in infancy or childhood with polyuria and failure to thrive, often occurring after a pregnancy with polyhydramnios. It is characterized by hypokalemic metabolic alkalosis, typically with hypercalciuria, and these patients resemble patients chronically taking loop diuretics that inhibit activity of NKCC2 pharmacologically. Defective function of NKCC2, ROMK, ClC-Kb, or barttin leads to impaired salt reabsorption in the MTAL, resulting in volume contraction and activation of the renin-angiotensin-aldosterone axis, which subsequently stimulates distal tubular secretion of potassium and protons, resulting in hypokalemic metabolic alkalosis. Despite impaired reabsorption of magnesium, serum magnesium levels are usually normal or only mildly reduced in patients with Bartter syndrome. Severity, age at onset of symptoms, and particular clinical features vary with the gene abnormality. For example, nephrocalcinosis as a consequence of hypercalciuria is most common in individuals with mutations in genes encoding NKCC2 and ROMK. Barttin is expressed in the inner ear, and patients with mutations in its gene have sensorineural deafness. As Barttin is also expressed beyond the TAL in the DCT, patients with type IV Bartter syndrome often have more severe salt wasting and typically lack hypercalciuria; they often develop progressive GFR loss. Cases of severe but transient antenatal Bartter syndrome have been reported with mutations in MAGED2 on the X chromosome, which regulates expression of NKCC2 in the MTAL, and of the thiazide-sensitive sodium-chloride cotransporter NCC in DCT.

Bartter syndrome is discussed further in Chapter 10.

Inherited Hypomagnesemic Hypercalciuria

Reabsorption of calcium and magnesium in the MTAL occurs through the paracellular route, driven by the positive electrical potential in the tubular lumen. The tight junctions between the epithelial cells determine the selective movement of cations (i.e., calcium, magnesium, and sodium). Disturbance of this selective paracellular barrier would be expected to produce parallel disorders in the reabsorption of calcium and magnesium.

Familial hypomagnesemia with substantial kidney magnesium losses, hypercalciuria, and nephrocalcinosis (FHHNC) is inherited in an autosomal recessive fashion. These patients develop kidney failure and kidney stones. Investigation of families first led to identification by positional cloning of the gene encoding a tight junction protein designated claudin 16 (also called paracellin 1 ). This was the first instance of a disease shown to result from mutations that alter a tight junction protein. Another member of this family, claudin 19, is mutated in other pedigrees in whom FHHNC is associated with ocular abnormalities (e.g., macular colobomas, myopia, horizontal nystagmus) with severe visual impairment. Both claudin 16 and claudin 19 are expressed in the thick ascending limb (TAL; Fig. 37.2 ), but claudin 19 also is expressed in the retina. These two proteins interact in the tight junction to regulate cation permeability. It is unclear why a defect in tight junctions is associated with hyperuricemia, a consistent finding in this disease.

Fig. 37.2, Transport mechanisms in the distal convoluted tubule. The basolateral sodium-potassium adenosine triphosphatase (Na + /K + -ATPase), composed of α, β, and γ subunits, establishes the low-intracellular-sodium concentration that provides the driving force for coupled sodium and chloride entry across the apical NCC transporter. It also maintains the high intracellular potassium concentration that drives potassium exit across the apical Kv1.1 potassium channel, which establishes a positive lumen-to-cytosol electrical gradient that drives magnesium entry across the apical TRPM6 cation channel. The basolateral Kir4.1 potassium channel allows potassium exit that may serve to assure an adequate potassium supply for the Na + /K + -ATPase. The EGF receptor stimulates trafficking of TRPM6 to the apical membrane and stimulates activity of that transporter. Genes encoding these proteins are responsible for inherited electrolyte disturbances discussed in the text. DCT, Distal convoluted tubule; EGF, epidermal growth factor; EGFR, epidermal growth factor receptor; NCC, sodium chloride cotransporter.

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here