Urolithiasis is one of the most common urologic diseases in industrialized societies. More than 80% of renal stones are composed of calcium oxalate, and small changes in urinary oxalate concentrations affect the risk of stone formation. Elucidation of the source of oxalate and its mechanism of transport is crucial for understanding the etiology of urolithiasis. Sources of oxalate can be both endogenous and exogenous. With regard to oxalate transport, tests were carried out to prove the function of solute-linked carrier 4 (SLC4) and SLC26. The molecular mechanism of urolithiasis caused by SLC4 and SLC26 is still unclear. The growing number of studies on the molecular physiology of SLC4 and SLC26, together with knockout genetic mouse model experiments, suggest that SLC4 and SLC26 may be a contributing element to urolithiasis. This review summarizes recent research on the sources of oxalate and characterization of the oxalate transport ionic exchangers SLC4 and SLC26, with an emphasis on different physiological defects in knockout mouse models including kidney stone formation. Furthermore, SLC4 and SLC26 exchangers provide new insight into urolithiasis and may be a novel therapeutic target for modification of urinary oxalate excretion.

Urolithiasis is a common urologic condition with an increasing incidence [1-3]. In Asia, about 1–19.1% of the population suffers from urolithiasis [4]. The lifetime recurrence risk is 50%, with an estimated time to recurrence of up to 1 year in 10% of cases, up to 5 years in 35–50% and up to 10 years in at least 50% of cases [5]. With lifestyle changes and economic development, the prevalence of urolithiasis has consistently increased in the last few decades. Calcium oxalate (CaOx) is the most common type of stone removed from patients [6, 7]. More than 70% of renal stone patients suffer from urolithiasis caused by CaOx stones; of these, CaOx dihydrate is the most common crystal in healthy human urine and CaOx monohydrate is the most common crystal in renal stones [7, 8]. Supersaturation of urine with CaOx is a major contributor to CaOx stone formation [2]. Thus, hyperoxaluria has been well recognized as one of the independent risk factors for stone formation. Oxalate is derived from dietary sources (e.g., plants and plant-derived foods), it can be synthesized by the body, and it is excreted in the urine [9, 10]. The etiology of hyperoxaluria has different aspects in different people. Frequent consumption of foods containing oxalate and oxalate precursors is thought to contribute to hyperoxaluria in some individuals. A low or suboptimal colonization of oxalate-degrading bacteria in the gastrointestinal tract [11], short bowel or malabsorptive bowel disease [12], and bariatric surgery for obesity [13] are also thought to be contributing factors.

Recently, research has focused on oxalate transport, which is a transcellular process that depends on anion exchangers. Thus, the interest in research on SLC4 and SLC26 has gradually increased. SLC, which belongs to a multifunctional anion exchanger system, is strongly involved in the homeostasis of oxalate [14]. The human erythrocyte band 3 protein (AE1 or SLC4A1) involved in oxalate transport in red blood cells has not been studied in detail, but some papers describe that treatment of red blood cell with different concentrations of H2DIDS (4,4’-diisothiocyanatostilbene-2,2’-disulfonate) inhibits Cl-Cl and oxalate-oxalate exchange to the same extent, suggesting that SLC4A1 is the major transport pathway for oxalate [15]. Mutations and knockout of SLC4A1 cause distal renal tubular acidosis (dRTA) and spherocytic hemolytic anemia [16]. SCL26A6 is an anion exchanger expressed on the apical membrane in many tissues, including the kidneys and intestines [17-20]. Among its transport activities, SLC26A6 serves as the major pathway for oxalate secretion [21]. Mice lacking SLC26A6 have a defect in intestinal oxalate secretion resulting in an enhanced net absorption of oxalate [22]. SLC26A1, A2, A3, and A7 may also be involved in oxalate exchange [23]. Besides, members of the SLC4 and SLC26 gene families encode Na+-independent Cl/HCO3 exchanges which are expressed in many tissues including the kidneys and the gastrointestinal tract. Secretion and reabsorption of acid-base equivalents and of Cl the Cl/HCO3 exchangers regulate the intracellular pH and Cl levels as well as the cell volume [17]. Next, the current knowledge of the mechanisms of SLC4 and SLC26 oxalate exchangers will be discussed in relation to the different etiological types of urolithiasis.

Sources of Oxalate

Urinary oxalate is derived from both exogenous and endogenous sources that, depending on the dietary intake, may equally contribute to urinary oxalate excretion [2]. There is new evidence that dietary oxalate may contribute to up to 50–80% of the oxalate excreted in urine [2, 24]. Exogenous sources of oxalate are vegetables, fruits, and tea. Foods with a high oxalate content are spinach, rhubarb, tea, spinach, and soya beans. Endogenous sources of oxalate are mainly synthesized by erythrocytes and the liver.The endogenoussources of oxalate include ascorbic acid, protein (through metabolism of the amino acids tyrosine, tryptophan, phenylalanine, and hydroxyproline) and the immediate precursors of oxalate, such as L-glycerate glycollate and glyoxylate [25]. The process of oxalate synthesis in erythrocytes is unknown and the contribution of ascorbate catabolism to urinary oxalate is controversial [26-31]. The liver is the primary source of endogenous oxalate, and glyoxylate is the primary immediate precursor of oxalate [32, 33]. Glyoxylate concentration is mainly regulated by many pathways. Glyoxylate can be metabolized to oxalate by glycolate oxidase or lactate dehydrogenase, and glyoxylate is an important intermediary product in several reactions. Excess glyoxylate will enhance the metabolism of glyoxylate to oxalate, leading to hyperoxaluria. Glyoxylate and alanine can be converted to glycine and pyruvate, respectively, via transamination by alanine/glyoxylate aminotransferase (AGT). AGT is a liver-specific enzyme localized in the peroxisomes that is dependent on the cofactor pyridoxine (vitamin B6) for full activity [34]. Glycine is metabolized to seine, and serine is converted to OH-pyruvate which is metabolized glycoaldehyde by glyoxylate reductase/OH-pyruvate reductase (GRHPR). Ultimately, glycoaldehyde can be metabolized to glyoxylate via several steps. Another glyoxylate source of pathways is through the breakdown of hydroxyproline [35] via 4-hydroxy-2-oxoglutarate aldolase (HOGA1).

The Etiology of Primary Hyperoxaluria

Hyperoxaluria is usually divided into the following 2 categories: primary hyperoxaluria (PH) and secondary hyperoxaluria. Depending on the dietary intake, the daily urinary oxalate excretion in healthy individuals ranges between 10 and 40 mg per 24 h (0.1–0.45 mmol per 24 h) [36]. Concentrations over 40–45 mg per 24 h (0.45–0.5 mmol per 24 h) are considered as indicative of clinical hyperoxaluria [34]. PH is a rare inborn error of glyoxylate metabolism that results in a high oxalate production mainly by the liver [37, 38], and PH is divided into the following 3 classes: PH I, PH II, PHII. PH I, accounting for the majority of all cases (70–80%) [38], is a peroxisomal disease and the most severe type. It is caused by mutations in AGT [39], a peroxisomal enzyme located in the liver; it is a genetic defect disease and it is inherited with an autosomal recessive pattern. Mutation or the absence of AGT resulting in a severe reduction of AGT enzymatic activity in the peroxisome produces more glyoxylate aggregation which is metabolized to oxalate via glycolate oxidase or lactate dehydrogenase, leading to hyperoxaluria. PH II, accounting for around 10% of PH cases, is caused by a deficiency of the cytosolic liver enzyme GRHPR [40, 41]. The deficiency or mutation of GRHPR activities in PH2 patients results in an increase in oxalate and L-glycerate concentrations in urine. The symptoms of hyperoxaluria caused by PH II are slighter than those caused by PH I. Severe hyperoxaluria is the clinical hallmark of these 2 types of PH, with reported values ranging between 88 and 352 mg per 24 h (1–4 mmol per 24 h) for PH I and 88 and 176 mg per 24 h (1–2 mmol per 24 h) for PH II [42]. PH III, accounting for 10% of PH cases, is caused by deficient HOGA1, which is a mitochondrial enzyme that cleaves 4-hydroxy-2-oxoglutarate into pyruvate and glyoxylate and the last step of the hydroxyproline degradation pathway [43]. The loss of HOGA1 enzyme activity leading a wide range of oxalate elevations in urine is the characterization of PH III which is becoming more frequent than PH2 as a cause of PH. All types of PH causing oxalate metabolism disorders in early childhood to adolescence have a major effect on CaOx supersaturation, resulting in the formation of nephrocalcinosis/nephrolithiasis. The age of PH at presentation varies from birth to the sixth decade of life, with a median of 5.5 years [44]. Besides, PH can severely damage renal function and make difficult the diagnosis of PH based on the lack of significant symptoms and effective inspection methods. In some end-stage renal patients, CaOx deposited in multiple organs with a renal function decline and increased blood oxalate levels causes systemic oxalosis including metabolic bone disease, anemia, and skin ulcerations.

The Etiology of Secondary Hyperoxaluria

Secondary hyperoxaluira involves many aspects including: (1) a high-oxalate diet (2) enteric hyperoxaluria, (3) oxalate-degrading microorganisms (4), and SLC4 and SLC26 ionic exchangers.

Secondary hyperoxaluria is the chief risk factor for urolithiasis which can be caused by either an increase in absorption of oxalate in the intestine or intake of a high-oxalate diet [45]. For many years it was thought that the dietary source of oxalate contributed only 10–15% of the daily oxalate excretion. However, it has been reported that dietary oxalate may contribute up to 50–80% of the oxalate excreted in urine [2, 24]. Daily dietary oxalate intakes are in the range of 0.05–0.2 g but may surpass 1 g if oxalate-rich foods are consumed [46-48]. Some foods, including strawberries, leafy spinach, rhubarb, beets, nuts, wheat bran, chocolate, tea, sorrel, sesame, carambola, almonds, and coffee, have considerably high oxalate concentrations [47, 49]. Studies have shown that an ample amount of calcium [50], especially when distributed within the food intake, protects against the risk of CaOx stone formation in patients with stone formation [51, 52], apparently owing to the calcium-oxalate interaction and excretion in the gastrointestinal tract. The intake of magnesium is suggested to reduce oxalate absorption and urinary excretion nearly as effectively as calcium by binding oxalate in the gut [53]. Furthermore, hypomagnesiuria is a known risk factor for formation of calcium stones in that magnesium binds to oxalate and reduces the urinary saturation of CaOx [54]. Protein plays an important role in the process of stone formation [55]. Some amino acids such as L-hydroxyproline are precursors of oxalate which increases the oxalate urine concentration and the relative supersaturation for CaOx and apparently increases oxalate renal excretion [56].

Oxalate is an endogenous byproduct of amino acid metabolism and it is also absorbed by the stomach, the small bowel, and the colon [57]. Enteric hyperoxaluria is a form of secondary hyperoxaluria caused by disease or resection of the gastrointestinal tract leading to malabsorption syndromes. Small bowel resection or inflammatory bowel disease could interrupt the enterohepatic circulation of bile salts, resulting in bile acid loss and fat malabsorption, which lead to steatorrhea effects on oxalate movement. Enteric hyperoxaluria may have the following 2 mainmechanisms: (1) alteration of mucosal oxalate permeability (both dihydroxy bile acid salts and fatty acids increase the permeability of the intestinal mucosa to oxalate; perfusion of rat colonic mucosa with 5 mM sodium deoxycholate markedly increased the colonic permeability to oxalate) [58] and (2) increased luminal ionic oxalate concentrations (in the malabsorption setting with steatorrhea, dietary calcium is bound by the free fatty acids in the intestinal lumen, so less calcium is bound to oxalate; complexation of fatty acids with luminal calcium increases the amount of soluble oxalate that is available for absorption as insoluble CaOx complexes are no longer formed) [13]. Soluble oxalate is consequently present in relatively high concentrations in the colon and can diffuse passively into blood, where it is filtered and excreted by the kidneys [61]. A number of patients with small bowel resection or inflammatory bowel disease have a high incidence of CaOx stone formation. In bariatric surgery patients it has long been linked to a higher incidence of new-onset nephrolithiasis, with a mean interval from surgery to diagnosis of nephrolithiasis ranging from 1.5 to 3.6 years [59-61]. The risk of nephrolithiasis varies by procedure, being the highest for malabsorptive procedures (22–28.7%) [59, 62] which could lead to enteric hyperoxaluria. The association between small bowel resection and increased urinary excretion of oxalate was first recognized in 1970 [63]; subsequently numerous cases have been reported, and many patients undergoing Rouxen-Y gastric bypass for morbid obesity are at risk for hyperoxaluria, nephrolithiasis, and oxalate nephropathy [64]. Enteric hyperoxaluria can cause CaOx stone disease and, as in PH, the urinary oxalate excretion can be high enough to cause renal damage and eventual end-stage renal disease [65].

It has been recognized for a few decades that intestinal microorganisms can degrade oxalate and reduce the concentration of oxalate in blood and urine. There are many bacteria including Enterococcus faecalis [66], Bifidobacterium infantis [67], and Oxalobacter formigenes [68]. The best well known oxalate-degrading microorganism is O. formigenes, a gram-negative anaerobic bacterium isolated from human feces and other animals that is capable of reducing intestinal oxalate absorption. O. formigenes potentially metabolizes the oxalate to formate and CO2 through the formyl-CoA transferase enzyme and the oxalyl-CoA decarboxylase enzyme. Many cases and evidence have confirmed that O. formigenes play an important role in degradation of oxalate in the intestinal tract. In a study, the absence of O. formigenses in stone formers is associated with an increase in urinary oxalate [69], and stoneformers generally have lower levels of colonization with O. formigenses than non-stone formers [70-72]. Besides, bariatric surgery, bowel diseases, and patients with cystic fibrosis could inhibit recolonization of O. formigenses in the intestinal lumen, leading to increased absorption of free oxalate and the concentration of oxalate in blood and urine.

SLC4 Transporters

The SLC4 gene family comprises 3 major clades [73], including Na+-independent electroneutral Cl/HCO3 exchangers (SLC4A1/AE1, SLC4A2/AE2, and SLC4A3/AE3), Na+-independent HCO3 transporters (electrogenic SLC4A4/NBCel and SLC4A5/NBCe2), electroneutral Na+/HCO3 cotransports (SLC4A7/NBCn1 and SLC4A10/NBCn2), and Na+-2HCO3 exchanger (SLC4A8/NDCBE) and SLC4A11/BTR1. All SLC4 polypeptides share a common structural pattern of 3 domains. An N-terminal cytoplasmic domain of 400–700 amino acids is followed by a C-terminal polytopic transmembrane domain of ∼500 amino acids and a C-terminal cytoplasmic domain of ∼30–100 amino acids [17]. Besides, SLC4 anion exchangers are involved in anion transport, e.g., plasmalemmal Cl/HCO3 exchangers which regulate intracellular pH and Cl and cell volume and contribute to transepithelial secretion and reabsorption of acid-base equivalents and of Cl. In addition, SLC4 exchangers involved in oxalate transport have been reported. SLC4A1, a member of the SLC4 exchangers, has been studied with respect to oxalate transport. We will focus on the structure and function of SLC4A1.

The human SLC4A1/AE1 gene encoding the anion exchanger 1 (AE1/band3) is located on chromosome 17q21-q22, encompassing approximately 20 kb and consisting of 20 exons separated by 19 introns [74]. The human SLC4A1 encodes both erythroid (eAE1/band3) and kidney (kAE1) isoforms of the AE1 protein by using different promoters and alternative splicing. Band 3 mRNA is transcribed from all exons by its upstream promoter, while kAE1 mRNA is transcribed by the promoter in intron 3. So, kAE1 mRNA lacks the sequences of exons 1–3 of the eAE1 and sequences shorter than eAE1 which in humans initiates at Met66. Human band 3 protein or eAE1, consisting of a 911-amino acid glycoprotein, comprises 2 distinct structural and functional domains, i.e., a 40-kDa N-terminal cytoplasmic domain and a 50-kDa C-terminal integral membrane domain. The N-terminal cytosolic domain, from residues 1–360, interacts with hemoglobin [75], glycolytic enzymes [76], and cytoskeletal proteins including ankyrin [77], protein 4.1 [78], and protein 4.2 [79], while the N-terminal cytoplasmic domain of kAE1 (which is 65 amino acids shorter than that of eAE1) does not bind to ankyrin or glycolytic enzymes and its interacting proteins in the a-intercalated cell remain to be identified. The eAE1 tetramers bind to ankyrin and cytoskeleton proteins. which is critical for maintenance of the biconcave disc shape of red blood cells. The 50-kDa C-terminal in the tergal membrane domain with 12–13 transmembrane spans from residue 361 to residue 882 and mediates Cl/HCO3 exchange [80], while the region of mediated oxalate is still unknown. Both eAE1 and kAE1 have a short acidic cytoplasmic C-terminal tail (residues 883–911), containing binding sites for carbonic anhydrase (CA) II [81]. The identical C-terminal tails of AE1 and kAE1 can bind to CAII via an acidic motif (-LDADD-). Bicarbonate (HCO3) from the breakdown of carbonic acid (H2CO3) generated from hydration of carbon dioxide (CO2) by CA II is transported across the cell membrane in exchange for chloride (Cl) by AE1 [74]. Red cell eAE1 expressed in the erythrocyte plasma membrane mediates the 1:1 electroneutral exchange of many anions, while Cl and HCO3 are its major physiological substrates [82]. eAE1 has also been confirmed to mediate oxalate exchange for Cl. Cousin and Motais [93] showed that various reversibly acting inhibitors have parallel effects on Cl-Cl and oxalate-oxalate exchange, suggesting that red cell oxalate transport is mediate by band 3, but the characteristics of the transport of oxalate across erythrocyte membrane are still unknown. Jennings and Adame [16] used a low concentration of H2 DIDS (4,4’-diisothiocyanatostilbene-2,2’-disulfonate) with red cells to explore the kinetics of oxalate exchange and Cl. The results showed that red cell eAE1 mediates H+/oxalate cotransport in electroneutral exchange for Cl and the apparent Cl affinity is 2–3 times higher than that of oxalate. Besides, oxalate, at a low extracellular pH, is transported by band 3 nearly as rapidly as Cl is at neutral pH. To examine the functional difference of erythrocyte band 3 protein in stone formation patients and normal patients, Oehlschläger et al. [84] found that the concentration of erythrocyte band 3 transport protein in stone formation patients is about 2 times higher than that of normal patients, but the plasma oxalate, cellular oxalate, and erythrocyte oxalate fluxes are no different in male stone formation patients versus normal patients. What is more, under normal conditions (pH 7.55), the oxalate efflux in stone formation patients is about 2 times higher than that in normal patients in vivo. SLC4A1 may influence cellular oxalate levels and urine oxalate clearance through the exchange of Cl/HCO3 and pH levels.

The SLC4A1 polypeptide is widely expressed in erythrocytes and in type A acid-secreting intercalated cells of the renal collecting duct. In erythrocytes, eAE1 stabilizes the lipid bilayer and its linkage to ankyrin and cytoskeleton proteins to maintain the biconcave disc shape of red blood cells. In the kidney, type A intercalated cells reabsorb HCO3 through apical H+ excretion into the urine. Mutations or knockout of the human SLC4A1 gene is divided into erythroid and renal phenotypes and it is linked to a number of human diseases. One group mutation of eAE1 encodes almost entirely asymptomatic polymorphisms encoding blood group antigens recognized by patient antisera in intact erythrocytes [85]. The largest group of mutations or knockout of eAE1 can result in hereditary spherocytosis, a common autosomal dominant hereditary spherocytic anemia with a reduction of osmotic fragility and surface area. Hereditary spherocytosis, involved in many polypeptides such as E40K, G130R, and L707P, can occur from gene nonsense, frameshift, and missense mutations of SLC4A1. Many mutant alleles gene generate unstable mRNA, and the resultant erythrocytes express reduced levels of wild-type eAE1 polypeptide often in the setting of dosage compensation by the wild-type allele. Usually, these patients have a normal renal acidification phenotype, but homozygous SLC4A1 V488M mutation can cause severe hereditary spherocytosis and dRTA [86]. There is a wide variation in clinical phenotypes, ranging from asymptomatic individuals to those with severe hemolytic anemia requiring a blood transfusion [87]. In addition, Southeast Asian ovalocytosis is a morphological red cell abnormality caused by a mutational deletion of 27 bp in exon 11 of SLC4A1 leading to an in-frame 9-amino acid deletion involving Ala400-Ala408 of eAE1 at the junction between the N-terminal domain and the first transmembrane span [88]. The deletion of Ala400-Ala408 of eAE1 impairs insertion of the mutant protein into the membrane [89]. The mutant protein is inactive for anion transport function and decreases red cell formability by forming oligomers. The homozygous mutation is fully lethal in utero in the absence of extraordinary medical intervention [90], while the heterozygous one exhibits inactivity for anion transport and membrane rigidity. The heterozygous SLC4A1 Southeast Asian ovalocytosis mutation does not cause dRTA [91]. However, mutations or knockout of kAE1 can cause dRTA. dRTA is a disease of defective urinary acidification, which is caused by insufficient net acid excretion into the urine by type A-intercalated cells of the kidney. Type A-intercalated cells mediate bicarbonate reabsorption into the blood in exchange for chloride and protons are transported into the urine by apical V-type H+. Patients with dRTA are defective in acid secretion and show an increased urine pH even after an acid challenge. These patients with dRTA are more susceptible to urolithiasis than normal subjects. The kidney disease dRTA presents in dominant and recessive forms [92]. Dominant mutants of kAE1 like R589Q can form heterodimers with wild-type AE1 and retain the wide-type protein in endoplasmic reticulum [93]. Homozygous kAE1 mutants, such as s773p [94], causing recessive renal tubular acidosis, impair protein trafficking to the cell surface. Compound heterozygote recessive mutants such as G701D and A858D of kAE1 can form heterodimers that are predominantly retained in the endoplasmic reticulum [95], while heterodimers with the wild-type protein can traffic to the plasma membrane in transfected human embryonic kidneys. Recently, the developed high-resolution melting method is efficient and reproducible for detection of SLC4A1 mutations and polymorphisms [96], making the diagnosis of dRTA caused by SLC4A1 easily.

SLC26 Transporters

SLC26 isoforms constitute a conserved family of anion transporters with 10 distinct members [97], including SLC26A1, SLC26A2, SLC26A3, SLC26A4, SLC26A5, SLC26A6, SLC26A7, SLC26A8, SLC26A9, and SLC26A11. SLC26A10 is a pseudogene and therefore not involved. SLC26 polypeptides are characterized by N-terminal cytoplasmic domains, 10–14 hydrophobic transmembrane spans, and C-terminal cytoplasmic STAS domains and appear to be homo-oligomeric [24].The SLC26 gene family, except for SLC26A5/prestin, encodes multifunctional anion exchangers and channels transporting amounts of substrates, including Cl, HCO3, oxalate, sulfate, and formate. Several SLC26 family members can specifically function as Cl/HCO3, including SLC26A3, SLC26A4, SLC26A6, SLC26A7, SLC26A9, and SLC26A9. SLC26 family members, including SLC26A1, SLC26A2, SLC26A3, SLC26A6, and SLC26A7, may be involved in oxalate transport.

SLC26A1/Sat1, the first member of the SLC26 family, was cloned from a rat liver [98] and is localized to the basolateral plasma membrane of hepatocytes and proximal tubular cells of the kidney (segments S1, S2, and S3), respectively [99, 100]. The human SLC26A1/Sat1 gene is mapped to chromosome 4p 16.3 [101]. SLC26A1/Sat1 is an anion exchanger for sulfate and bicarbonate that is capable of mediating oxalate transport and the affinity for oxalate is lower than for counter anions. Some studies in rats and mice have reported that gender differences in SLC26A1/Sat1 expression, with a male-dominant expression, may explain the morbidity of oxalate urolithiasis in males and females. A study of SLC26A1/Sat1 in rats indicated that male rats, in comparison to female rats, exhibited higher plasma oxalate levels, a higher renal expression of SLC26A1/Sat1, and a higher excretion of oxalate in urine [102]. In rats treated with ethylene glycol, female rats showed awake oxalemia and an absence of oxaluria compared to male rats which showed hyperoxalemia, hyperoxaluria, and oxalate urolithiasis by elevating protein expression of the hepatic and renal oxalate transporter SCL26A1/Sat1 [103]. In humans, the incidence of urolithiasis is 2- to 3-fold higher in middle aged male patients (age 30–59 years) but similar in both genders in young and old patients [104]. These studies indicated that sex hormones may be a contributor to gender-dependent development of oxalate nephrolithiasis in humans and rats. Research in SLC26A1/ Sat1-null mice has demonstrated that Sat1 plays an important role in homeostasis of both sulfate and oxalate [105]. Slc26A1/Sat1-null mice have a hyperoxalemia:plasma oxalate level that is higher in Sat1-null mice than in wild-type Sat1 mice, and Sat1-null mice exhibits hyperoxalemia, hyperoxaluria, and calcium urolithiasis. The loss of SLC26A1/Sat1 leads not only to urinary stone formation and drug-induced hepatotoxicity but also to hyposulfatemia, hypersulfaturia, and enhanced hepatotoxicity in SLC26A1/Sat1-null mice [106]. Recent research has indicated that mutations SLC26A1 could cause a recessive mendelian form of nephrolithiasis [107].

SLC26A2/diastrophic dysplasia sulfate transporter (DTDST) was first identified by positional cloning as the gene underlying autosomal recessive diastrophic dysplasia [108]. In humans, SLC26A2/DTDST comprises 3 exons and encodes a 739-amino acid protein. SLC26A2/DTDST is a DTDST and it is located at the apical membrane of the rat small intestine, the rat proximal tubule, the human colon, and prominently in chondrocytes. SLC26A2/DSDST can mediate SO42–, Cl, HCO3, and oxalate transport [109]. Exchange SO42– of SLC26A2/DSDST is critical for cartilage development and function [110], and the function of SLC26A2/DSDST in chondrocytes cannot be compensated for by any of the other SLC26 SO42– transporters, such as the ubiquitous SCL26A1 and SLC26A6 or SLC26A3 [111, 118]. Mutations in SLC26A2/DSDST are associated with diastrophic dysplasia [112]. The features of the disease are generally attributed to a lack of SO42– transport which is needed for sulfation of extracellular matrix proteins, and knockdown and overexpression of SLC26A2/DSDST have revealed an essential role of SLC26A2/DSDST in chondrocyte differentiation and maturation [113]. Some results have indicated that the properties of oxalate transport closely resemble those of SO42– transport, but the oxalate transport rate is ∼50% slower than the SO42– transport rate [114]. Besides, SLC26A2 is not likely to play a major role in oxalate metabolism in the colon or the kidney [114]; neither hyperoxaluria nor lithiasis has been noted among mice or patients with SLC26A2 chondrodysplasias.

SLC26A3/DRA (downregulated in adenoma) was first identified as a candidate tumor suppressor gene [115]. SLC26A3/DRA is expressed on the apical membrane of enterocytes of humans and it mediates Cl, HCO3, OH, and oxalate transport. Recent research has demonstrated that the deletion of SLC26A3/DRA in knockout mice results in a significant reduction of urinary oxalate excretion and serum oxalate concentrations [116], suggesting that SLC26A3/DRA has a critical role in oxalate transport in enterocytes by reducing the mucosa-to-serosa oxalate transport in the ileum and colon. Human diseases are associated with mutations of SLC26A3/DRA, such as congenital chloride-losing diarrhea, which is characterized by watery stool with an excess of chloride and metabolic alkalosis [117]. Mice lacking SLC26A3/DRA show a reduced Cl/HCO3 transport ability in the colon epithelium that leads to acidic diarrhea with chloride content and volume depletion and growth retardation [118].

SLC26A6/PAT1 was first cloned by homology from humans [19], encompassing 21 coding exons and about 10 kb of genomic DNA. The expression of the human SLC26A6/PAT1 gene on human chromosome 3p21 [18] is widespread, with considerable transcripts in several epithelial and nonepithelial tissues [119], including the intestine, the kidney proximal tubule, and the heart. SLC26A6/PAT1 is a multifunction mode of anion exchange involving Cl, HCO3, sulfate, hydroxyl, and oxalate. The mouse SLC26A6-mediated Cl/formate exchange is electroneutral [120], while Cl/oxalate is electronic [121]. SLC26A6/PAT1 is expressed on the apical membrane in many gastrointestinal tissues, including the pancreas and the small intestine [19]. SLC26A6/PAT1 is expressed at high levels in the small intestine (at equivalent levels in the duodenum and jejunum and at lower levels in the ileum), with far lower levels of expression in the large intestine [21]. Recently, it was observed that SLC26A6-knockout mice have a reduced (50–75%) enteric secretion of oxalate and an increased net amount of oxalate absorbed in the gastrointestinal tract rather than a net proximal tubular secretion of oxalate, elevating plasma oxalate and causing hyperoxaluria. Moreover, SLC26A6 knockout mice present urinary oxalate calcium stones while wild-type mice present no birefringent deposits in the kidneys. This research confirms the fact that SLC26A6/PAT1 is a major oxalate-secreting transporter and may be a new target for curing CaOx stones in the gastrointestinal tract. Human SLC26A6/PAT1 has a lower affinity for extracellular Cl, and Cl-oxalate exchange appears to be electroneutral, suggesting that human intestinal oxalate secretion is less efficient relative to SLC26A6-null mice [121].

SLC26A7 was cloned on the basis of homology to other SLC26 isoforms [122]. In humans, SLC26A7 maps to chromosome 8 and encodes a 656-amino acid protein. SLC26A7 has been reported to exhibit Cl, HCO3, and oxalate exchange activity. SLC26A7 is localized to the basolateral membrane of the stomach’s parietal cells and the basolateral membrane of renal collecting duct type A intercalated cells [123]. The mechanism of oxalate transport of SLC26A7 is unknown, but the transport may be involved in the deposition of CaOx crystals in the lumens of the medullary collecting ducts in hyperoxaluric rats [124]. In addition, one study showed that oxalate transport at the papillary epithelial membrane can be involved in deposition of CaOx crystals onto Randall’s plaques [125]. SLC26A7-null mouse was remarkable for dRTA and gastric hypochlorhydria [126] because of a reduced basolateral Cl-HCO3 anion exchange activity.

Oxalate transport in the human body, including the intestine, red blood cells, and kidneys) is complicated. There are many factors that could change the normal manner of oxalate transport, contributing to elevate oxalate concentration of blood and urine and increasing the incidence rate of CaOx stone formation. However, it is important to be aware that SLC26 and SLC4 anion exchange transporters play an indispensable role in maintaining oxalate homeostasis. In the future, SLC26 and SLC4 anion exchange transporters may become new targets to cure CaOx stones. Our next research will elaborate on the relationship of CaOx stones and anion exchange transporters.

Thanks go to Prof. YuChen Liu for language editing.

The authors declare no conflict of interests.

None.

Yi Huang, Ze Pai Chi, and Ru Huang are first authors. Yong Hai Zhang is the corresponding author. Yi Huang drafted this paper. Ze Pai Chi and Ru Huang contributed to revision of this paper for important content. Yong Hai Zhang revised and approved the final version of this paper. All of the authors read and approved the final version of this paper.

1.
Li
YH
,
Zhang
J
,
Liu
HY
.
Curcumin ameliorates glyoxylate-induced calcium oxalate deposition and renal injuries in mice
.
Phytomedicine, Phymed
;
2019
. p.
152861
.
2.
Holmes
RP
,
Goodman
HO
,
Assimos
DG
.
Contribution of dietary oxalate to urinary oxalate excretion
.
Kidney Int
.
2001
Jan
;
59
(
1
):
270
6
.
[PubMed]
0085-2538
3.
Ortiz
YV
,
Arestin
CC
,
Medina
PG
,
Torres
PP
.
Urolithiasis: population analysis and composition
.
Clin Chim Acta
.
2019
;
493
:
S460
92
. 0009-8981
4.
Liu
Y
,
Chen
YT
.
Epidemiology of urolithiasis in Asia.
Asian Journal of Urology.
2018
; S2214-3882(18)30072-9
5.
Penniston
KL
,
McLaren
ID
,
Greenlee
RT
,
Nakada
SY
.
Urolithiasis in a rural Wisconsin population from 1992 to 2008: narrowing of the male-to-female ratio
.
J Urol
.
2011
May
;
185
(
5
):
1731
6
.
[PubMed]
0022-5347
6.
Tsujihata
M
.
Mechanism of calcium oxalate renal stone formation and renal tubular cell injury
.
Int J Urol
.
2008
Feb
;
15
(
2
):
115
20
.
[PubMed]
0919-8172
7.
Ivanovski
O
,
Drüeke
TB
.
A new era in the treatment of calcium oxalate stones?
Kidney Int
.
2013
Jun
;
83
(
6
):
998
1000
.
[PubMed]
0085-2538
8.
Schubert
G
.
Stone analysis
.
Urol Res
.
2006
Apr
;
34
(
2
):
146
50
.
[PubMed]
0300-5623
9.
Holmes
RP
,
Assimos
DG
.
The impact of dietary oxalate on kidney stone formation
.
Urol Res
.
2004
Oct
;
32
(
5
):
311
6
.
[PubMed]
0300-5623
10.
Holmes
RP
,
Ambrosius
WT
,
Assimos
DG
.
Dietary oxalate loads and renal oxalate handling
.
J Urol
.
2005
Sep
;
174
(
3
):
943
7
.
[PubMed]
0022-5347
11.
Hatch
M
,
Freel
RW
.
The roles and mechanisms of intestinal oxalate transport in oxalate homeostasis
.
Semin Nephrol
.
2008
Mar
;
28
(
2
):
143
51
.
[PubMed]
0270-9295
12.
Worcester
EM
.
Stones from bowel disease
.
Endocrinol Metab Clin North Am
.
2002
Dec
;
31
(
4
):
979
99
.
[PubMed]
0889-8529
13.
Lieske
JC
,
Kumar
R
,
Collazo-Clavell
ML
.
Nephrolithiasis after bariatric surgery for obesity
.
Semin Nephrol
.
2008
Mar
;
28
(
2
):
163
73
.
[PubMed]
0270-9295
14.
Kleta
R
.
A key stone cop regulates oxalate homeostasis
.
Nat Genet
.
2006
Apr
;
38
(
4
):
403
4
.
[PubMed]
1061-4036
15.
Jennings
ML
,
Adame
MF
.
Characterization of oxalate transport by the human erythrocyte band 3 protein
.
J Gen Physiol
.
1996
Jan
;
107
(
1
):
145
59
.
[PubMed]
0022-1295
16.
Alper
SL
.
Molecular physiology of SLC4 anion exchangers
.
Exp Physiol
.
2006
Jan
;
91
(
1
):
153
61
.
[PubMed]
0958-0670
17.
Waldegger
S
,
Moschen
I
,
Ramirez
A
,
Smith
RJ
,
Ayadi
H
,
Lang
F
, et al
Cloning and characterization of SLC26A6, a novel member of the solute carrier 26 gene family
.
Genomics
.
2001
Feb
;
72
(
1
):
43
50
.
[PubMed]
0888-7543
18.
Lohi
H
,
Kujala
M
,
Kerkelä
E
,
Saarialho-Kere
U
,
Kestilä
M
,
Kere
J
.
Mapping of five new putative anion transporter genes in human and characterization of SLC26A6, a candidate gene for pancreatic anion exchanger
.
Genomics
.
2000
Nov
;
70
(
1
):
102
12
.
[PubMed]
0888-7543
19.
Knauf
F
,
Yang
CL
,
Thomson
RB
,
Mentone
SA
,
Giebisch
G
,
Aronson
PS
.
Identification of a chloride-formate exchanger expressed on the brush border membrane of renal proximal tubule cells
.
Proc Natl Acad Sci USA
.
2001
Jul
;
98
(
16
):
9425
30
.
[PubMed]
0027-8424
20.
Wang
Z
,
Petrovic
S
,
Mann
E
,
Soleimani
M
.
Identification of an apical Cl(-)/HCO3(-) exchanger in the small intestine
.
Am J Physiol Gastrointest Liver Physiol
.
2002
Mar
;
282
(
3
):
G573
9
.
[PubMed]
0193-1857
21.
Mukaibo
T
,
Munemasa
T
,
George
AT
,
Tran
DT
,
Gao
X
,
Herche
JL
, et al
The apical anion exchanger Slc26a6 promotes oxalate secretion by murine submandibular gland acinar cells
.
J Biol Chem
.
2018
Apr
;
293
(
17
):
6259
68
.
[PubMed]
0021-9258
22.
Jiang
Z
,
Asplin
JR
,
Evan
AP
,
Rajendran
VM
,
Velazquez
H
,
Nottoli
TP
, et al
Calcium oxalate urolithiasis in mice lacking anion transporter Slc26a6
.
Nat Genet
.
2006
Apr
;
38
(
4
):
474
8
.
[PubMed]
1061-4036
23.
Alper
SL
,
Sharma
AK
.
The SLC26 gene family of anion transporters and channels
.
Mol Aspects Med
.
2013
Apr-Jun
;
34
(
2-3
):
494
515
.
[PubMed]
0098-2997
24.
Assimos
DG
,
Holmes
RP
.
Role of diet in the therapy of urolithiasis
.
Urol Clin North Am
.
2000
May
;
27
(
2
):
255
68
.
[PubMed]
0094-0143
25.
Fargue
S
,
Knight
J
,
Holmes
RP
,
Rumsby
G
,
Danpure
CJ
.
Effects of alanine:glyoxylate aminotransferase variants and pyridoxine sensitivity on oxalate metabolism in a cell-based cytotoxicity assay
.
Biochim Biophys Acta
.
2016
Jun
;
1862
(
6
):
1055
62
.
[PubMed]
0006-3002
26.
Chai
W
,
Liebman
M
,
Kynast-Gales
S
,
Massey
L
.
Oxalate absorption and endogenous oxalate synthesis from ascorbate in calcium oxalate stone formers and non-stone formers
.
Am J Kidney Dis
.
2004
Dec
;
44
(
6
):
1060
9
.
[PubMed]
0272-6386
27.
Massey
LK
,
Liebman
M
,
Kynast-Gales
SA
.
Ascorbate increases human oxaluria and kidney stone risk
.
J Nutr
.
2005
Jul
;
135
(
7
):
1673
7
.
[PubMed]
0022-3166
28.
Taylor
EN
,
Stampfer
MJ
,
Curhan
GC
.
Dietary factors and the risk of incident kidney stones in men: new insights after 14 years of follow-up
.
J Am Soc Nephrol
.
2004
Dec
;
15
(
12
):
3225
32
.
[PubMed]
1046-6673
29.
Auer
BL
,
Auer
D
,
Rodgers
AL
.
The effect of ascorbic acid ingestion on the biochemical and physicochemical risk factors associated with calcium oxalate kidney stone formation
.
Clin Chem Lab Med
.
1998
Mar
;
36
(
3
):
143
7
.
[PubMed]
1434-6621
30.
Hatch
M
,
Mulgrew
S
,
Bourke
E
,
Keogh
B
,
Costello
J
.
Effect of megadoses of ascorbic acid on serum and urinary oxalate
.
Eur Urol
.
1980
;
6
(
3
):
166
9
.
[PubMed]
0302-2838
31.
Auer
BL
,
Auer
D
,
Rodgers
AL
.
Relative hyperoxaluria, crystalluria and haematuria after megadose ingestion of vitamin C
.
Eur J Clin Invest
.
1998
Sep
;
28
(
9
):
695
700
.
[PubMed]
0014-2972
32.
Baker
PR
,
Cramer
SD
,
Kennedy
M
,
Assimos
DG
,
Holmes
RP
.
Glycolate and glyoxylate metabolism in HepG2 cells
.
Am J Physiol Cell Physiol
.
2004
Nov
;
287
(
5
):
C1359
65
.
[PubMed]
0363-6143
33.
Behnam
JT
,
Williams
EL
,
Brink
S
,
Rumsby
G
,
Danpure
CJ
.
Reconstruction of human hepatocyte glyoxylate metabolic pathways in stably transformed Chinese-hamster ovary cells
.
Biochem J
.
2006
Mar
;
394
(
Pt 2
):
409
16
.
[PubMed]
0264-6021
34.
Marengo
SR
,
Romani
AM
.
Oxalate in renal stone disease: the terminal metabolite that just won’t go away
.
Nat Clin Pract Nephrol
.
2008
Jul
;
4
(
7
):
368
77
.
[PubMed]
1745-8323
35.
Knight
J
,
Jiang
J
,
Assimos
DG
,
Holmes
RP
.
Hydroxyproline ingestion and urinary oxalate and glycolate excretion
.
Kidney Int
.
2006
Dec
;
70
(
11
):
1929
34
.
[PubMed]
0085-2538
36.
Robijn
S
,
Hoppe
B
,
Vervaet
BA
,
D’Haese
PC
,
Verhulst
A
.
Hyperoxaluria: a gut-kidney axis?
Kidney Int
.
2011
Dec
;
80
(
11
):
1146
58
.
[PubMed]
0085-2538
37.
Hoppe
B
,
Beck
BB
,
Milliner
DS
.
The primary hyperoxalurias
.
Kidney Int
.
2009
Jun
;
75
(
12
):
1264
71
.
[PubMed]
0085-2538
38.
Danpure
CJ
.
Molecular etiology of primary hyperoxaluria type 1: new directions for treatment
.
Am J Nephrol
.
2005
May-Jun
;
25
(
3
):
303
10
.
[PubMed]
0250-8095
39.
Wood
DK
,
Holmes
RP
,
Erbe
D
.
Reduction in urinary oxalate excretion in mouse models of primary hyperoxaluria by RNA interference inhibition of liver lactate dehydrogenase activity.
Biochimica et Biophysica Acta- Molecular Basis of Disease,
2019
; S0925-4439(19)30145-0
40.
Bobrowski
AE
,
Langman
CB
.
The primary hyperoxalurias
.
Semin Nephrol
.
2008
Mar
;
28
(
2
):
152
62
.
[PubMed]
0270-9295
41.
Leumann
E
,
Hoppe
B
.
The primary hyperoxalurias
.
J Am Soc Nephrol
.
2001
Sep
;
12
(
9
):
1986
93
.
[PubMed]
1046-6673
42.
De Broe
ME
,
Porter
GA
. Chapter 32: Oxalate. In:
De Broe
ME
,
Porter
GA
, editors
.
Clinical Nephrotoxins: Renal Injury from Drugs and Chemicals
. 3rd ed.
New York, NY, USA
:
Springer
;
2008
. pp.
749
56
.
43.
Phang
JM
,
Hu
CA
,
Valle
D
. Disorders in proline and hydroxyproline metabolism, in: C. Scriver, A. Beaudet W. Valle D, Childs B (Eds.). The Metabolic and Molecular Bases of Inherited Disease, 8th ed, McGraw-Hill, New York.
2001
;1821–1838.
44.
Cochat
P
,
Rumsby
G
.
Primary hyperoxaluria
.
N Engl J Med
.
2013
Aug
;
369
(
7
):
649
58
.
[PubMed]
0028-4793
45.
Siener
R
,
Bade
DJ
,
Hesse
A
,
Hoppe
B
. Dietary hyperoxaluria is not reduced by treatment with Lactic acid bacteria, J. Transl. Med.
2013
;11:306e313.
46.
Siener
R
,
Schade
N
,
Nicolay
C
. The efficacy of dietary intervention on urinary risk factors for stone formation in recurrent calcium oxalate stone patients, J. Urology.
2005
;173:1601e1605.
47.
Siener
R
, H€onow R, Voss S, Seidler A, Hesse A. Oxalate content of cereals and cereal products, J. Agric. Food Chem.
2006
;54:3008e3011.
48.
Taylor
EN
,
Curhan
GC
. Oxalate intake and the risk for nephrolithiasis, J. Am. Soc. Nephrol.
2007
; 18,:2198e2204.
49.
Duncan
SH
,
Richardson
AJ
,
Kaul
P
,
Holmes
RP.
,
Allisonand
MJ
,
Stewart
CS.O.
Formigenes and its potential role in human health, Appl. Environ. Microbiol.
2002
;68:3841e3847.
50.
Borghi
L
,
Schianchi
T
,
Meschi
T
,
Guerra
A
,
Allegri
F
,
Maggiore
U
, et al
Comparison of two diets for the prevention of recurrent stones in idiopathic hypercalciuria
.
N Engl J Med
.
2002
Jan
;
346
(
2
):
77
84
.
[PubMed]
0028-4793
51.
Domrongkitchaiporn
S
,
Sopassathit
W
,
Stitchantrakul
W
,
Prapaipanich
S
,
Ingsathit
A
,
Rajatanavin
R
.
Schedule of taking calcium supplement and the risk of nephrolithiasis
.
Kidney Int
.
2004
May
;
65
(
5
):
1835
41
.
[PubMed]
0085-2538
52.
de O G Mendonça
C
,
Martini
LA
,
Baxmann
AC
,
Nishiura
JL
,
Cuppari
L
,
Sigulem
DM
, et al
Effects of an oxalate load on urinary oxalate excretion in calcium stone formers
.
J Ren Nutr
.
2003
Jan
;
13
(
1
):
39
46
.
[PubMed]
1051-2276
53.
Liebman
M
,
Costa
G
.
Effects of calcium and magnesium on urinary oxalate excretion after oxalate loads
.
J Urol
.
2000
May
;
163
(
5
):
1565
9
.
[PubMed]
0022-5347
54.
Hornberger
B
,
Bollner
MR
.
Kidney Stones
.
Physician Assist Clin
.
2018
;
3
(
1
):
37
54
.
55.
Canales
BK
,
Anderson
L
,
Higgins
L
,
Slaton
J
,
Roberts
KP
,
Liu
N
, et al
Second prize: comprehensive proteomic analysis of human calcium oxalate monohydrate kidney stone matrix
.
J Endourol
.
2008
Jun
;
22
(
6
):
1161
7
.
[PubMed]
0892-7790
56.
Mendonca
FS
,
Pedreira
RS
.
Hydroxyproline and starch consumption and urinary supersaturation with calcium oxalate in cats.
Animal Feed Science and Technology.
2018
; S0377-8401(18)30150-0
57.
Nazzal
L
,
Puri
S
,
Goldfarb
DS
. Enteric hyperoxaluria: an important cause of end-stage kidney disease, Nephrol. Dial.Transpl.
2016
; 31:375e382,
58.
Dobbins
JW
,
Binder
HJ
.
Effect of bile salts and fatty acids on the colonic absorption of oxalate
.
Gastroenterology
.
1976
Jun
;
70
(
6
):
1096
100
.
[PubMed]
0016-5085
59.
Sinha
MK.
,
Collazo-Clavell
ML
,
Rule
A
,
Milliner
DS.
Hyperoxaluric nephrolithiasis is a complication of Roux-en-Y gastric bypass surgery, Kidney Int.
2007
;72:100e107,
60.
Asplin
JR.
,
Coe
FL.
Hyperoxaluria in kidney stone formers treated with modern bariatric surgery. J. Urol.
2007
; 177:565e569
61.
Durrani
O
,
Morrisroe
S
,
Jackman
S
,
Averch
T
. Analysis of stone disease in morbidly obese patients undergoing gastric bypass surgery. J Endourol.
2006
;20:749e752,
62.
Requarth
JA
,
Burchard
KW
,
Colacchio
TA
,
Stukel
TA
,
Mott
LA
,
Greenberg
ER
, et al
Long-term morbidity following jejunoileal bypass. The continuing potential need for surgical reversal
.
Arch Surg
.
1995
Mar
;
130
(
3
):
318
25
.
[PubMed]
0004-0010
63.
Kaye
MC
,
Streem
SB
,
Hall
PM
.
Enteric hyperoxaluria associated with external biliary drainage
.
J Urol
.
1994
Feb
;
151
(
2
):
396
7
.
[PubMed]
0022-5347
64.
Duffey
BG
,
Alanee
S
. Hyperoxaluria Is a Long-Term Consequence of Roux-en-Y Gastric Bypass: A 2-Year Prospective Longitudinal Study. Elsevier Inc;
2010
;211,1
65.
Mole
DR
,
Tomson
CR
,
Mortensen
N
,
Winearls
CG
.
Renal complications of jejuno-ileal bypass for obesity
.
QJM
.
2001
Feb
;
94
(
2
):
69
77
.
[PubMed]
1460-2725
66.
Hokama
S
,
Honma
Y
,
Toma
C
,
Ogawa
Y
.
Oxalate-degrading Enterococcus faecalis
.
Microbiol Immunol
.
2000
;
44
(
4
):
235
40
.
[PubMed]
0385-5600
67.
Campieri
C
,
Campieri
M
,
Bertuzzi
V
,
Swennen
E
,
Matteuzzi
D
,
Stefoni
S
, et al
Reduction of oxaluria after an oral course of lactic acid bacteria at high concentration
.
Kidney Int
.
2001
Sep
;
60
(
3
):
1097
105
.
[PubMed]
0085-2538
68.
Turkmen
K
,
Erdur
FM
.
The relationship between colonization of Oxalobacter formigenes serum oxalic acid and endothelial dysfunction in hemodialysis patients: from impaired colon to impaired endothelium
.
Med Hypotheses
.
2015
Mar
;
84
(
3
):
273
5
.
[PubMed]
0306-9877
69.
Kwak
C
,
Kim
HK
,
Kim
EC
,
Choi
MS
,
Kim
HH
.
Urinary oxalate levels and the enteric bacterium Oxalobacter formigenes in patients with calcium oxalate urolithiasis
.
Eur Urol
.
2003
Oct
;
44
(
4
):
475
81
.
[PubMed]
0302-2838
70.
Kumar
R
,
Mukherjee
M
,
Bhandari
M
,
Kumar
A
,
Sidhu
H
,
Mittal
RD
.
Role of Oxalobacter formigenes in calcium oxalate stone disease: a study from North India
.
Eur Urol
.
2002
Mar
;
41
(
3
):
318
22
.
[PubMed]
0302-2838
71.
Troxel
SA
,
Sidhu
H
,
Kaul
P
,
Low
RK
.
Intestinal Oxalobacter formigenes colonization in calcium oxalate stone formers and its relation to urinary oxalate
.
J Endourol
.
2003
Apr
;
17
(
3
):
173
6
.
[PubMed]
0892-7790
72.
Mikami
K
,
Akakura
K
,
Takei
K
,
Ueda
T
,
Mizoguchi
K
,
Noda
M
, et al
Association of absence of intestinal oxalate degrading bacteria with urinary calcium oxalate stone formation
.
Int J Urol
.
2003
Jun
;
10
(
6
):
293
6
.
[PubMed]
0919-8172
73.
Alper
SL
.
Genetic diseases of acid-base transporters
.
Annu Rev Physiol
.
2002
;
64
(
1
):
899
923
.
[PubMed]
0066-4278
74.
Yenchitsomanus
PT
,
Kittanakom
S
,
Rungroj
N
,
Cordat
E
,
Reithmeier
RA
.
Molecular mechanisms of autosomal dominant and recessive distal renal tubular acidosis caused by SLC4A1 (AE1) mutations
.
J Mol Genet Med
.
2005
Nov
;
1
(
2
):
49
62
.
[PubMed]
1747-0862
75.
Basu
A
,
Chakrabarti
A
.
Hemoglobin interacting proteins and implications of spectrin hemoglobin interaction
.
J Proteomics
.
2015
Oct
;
128
:
469
75
.
[PubMed]
1874-3919
76.
Chu
H
,
Low
PS
.
Mapping of glycolytic enzyme-binding sites on human erythrocyte band 3
.
Biochem J
.
2006
Nov
;
400
(
1
):
143
51
.
[PubMed]
0264-6021
77.
Chang
SH
,
Low
PS
.
Identification of a critical ankyrin-binding loop on the cytoplasmic domain of erythrocyte membrane band 3 by crystal structure analysis and site-directed mutagenesis
.
J Biol Chem
.
2003
Feb
;
278
(
9
):
6879
84
.
[PubMed]
0021-9258
78.
Lombardo
CR
,
Willardson
BM
,
Low
PS
.
Localization of the protein 4.1-binding site on the cytoplasmic domain of erythrocyte membrane band 3
.
J Biol Chem
.
1992
May
;
267
(
14
):
9540
6
.
[PubMed]
0021-9258
79.
Low
PS
.
Structure and function of the cytoplasmic domain of band 3: center of erythrocyte membrane-peripheral protein interactions
.
Biochim Biophys Acta
.
1986
Sep
;
864
(
2
):
145
67
.
[PubMed]
0006-3002
80.
Groves
JD
,
Tanner
MJ
.
Topology studies with biosynthetic fragments identify interacting transmembrane regions of the human red-cell anion exchanger (band 3; AE1)
.
Biochem J
.
1999
Dec
;
344
(
Pt 3
):
687
97
.
[PubMed]
0264-6021
81.
Vince
JW
,
Reithmeier
RA
.
Carbonic anhydrase II binds to the carboxyl terminus of human band 3, the erythrocyte C1-/HCO3- exchanger
.
J Biol Chem
.
1998
Oct
;
273
(
43
):
28430
7
.
[PubMed]
0021-9258
82.
Alper
SL
. Molecular physiology and genetics of Na+-independent SLC4 anion exchangers. the Journal of Experimental Biology
2009
;212: 1672-1683
83.
Cousin
JL
,
Motais
R
.
Inhibition of anion permeability by amphiphilic compounds in human red cell: evidence for an interaction of niflumic acid with the band 3 protein
.
J Membr Biol
.
1979
Apr
;
46
(
2
):
125
53
.
[PubMed]
0022-2631
84.
Oehlschläger
S
,
Fuessel
S
,
Meye
A
,
Herrmann
J
,
Lotzkat
U
,
Froehner
M
, et al
Importance of erythrocyte band III anion transporter (SLC4A1) on oxalate clearance of calcium oxalate monohydrate stone-formering patients vs. normal controls
.
Urology
.
2011
Jan
;
77
(
1
):
250.e1
5
.
[PubMed]
0090-4295
85.
Alper
SL
.
Molecular physiology of SLC4 anion exchangers.
Exp Physiol
2006
; 91.1 153–161
86.
Ribeiro
ML
,
Alloisio
N
,
Almeida
H
,
Gomes
C
,
Texier
P
,
Lemos
C
, et al
Severe hereditary spherocytosis and distal renal tubular acidosis associated with the total absence of band 3
.
Blood
.
2000
Aug
;
96
(
4
):
1602
4
.
[PubMed]
0006-4971
87.
Trompeter
S
,
King
MJ
.
Hereditary spherocytosis
.
Paediatr Child Health
.
2019
;
•••
:
368
79
.1205-7088
88.
Jarolim
P
,
Palek
J
,
Amato
D
,
Hassan
K
,
Sapak
P
,
Nurse
GT
, et al
Deletion in erythrocyte band 3 gene in malaria-resistant Southeast Asian ovalocytosis
.
Proc Natl Acad Sci USA
.
1991
Dec
;
88
(
24
):
11022
6
.
[PubMed]
0027-8424
89.
Cheung
JC
,
Li
J
,
Reithmeier
RA
.
Topology of transmembrane segments 1-4 in the human chloride/bicarbonate anion exchanger 1 (AE1) by scanning N-glycosylation mutagenesis
.
Biochem J
.
2005
Aug
;
390
(
Pt 1
):
137
44
.
[PubMed]
0264-6021
90.
Picard
V
,
Proust
A
,
Eveillard
M
,
Flatt
JF
,
Couec
ML
,
Caillaux
G
, et al
Homozygous Southeast Asian ovalocytosis is a severe dyserythropoietic anemia associated with distal renal tubular acidosis
.
Blood
.
2014
Mar
;
123
(
12
):
1963
5
.
[PubMed]
0006-4971
91.
Bruce
LJ
,
Wrong
O
,
Toye
AM
,
Young
MT
,
Ogle
G
,
Ismail
Z
, et al
Band 3 mutations, renal tubular acidosis and South-East Asian ovalocytosis in Malaysia and Papua New Guinea: loss of up to 95% band 3 transport in red cells
.
Biochem J
.
2000
Aug
;
350
(
Pt 1
):
41
51
.
[PubMed]
0264-6021
92.
Cordat
E
,
Reithmeier
RA
.
Expression and interaction of two compound heterozygous distal renal tubular acidosis mutants of kidney anion exchanger 1 in epithelial cells
.
Am J Physiol Renal Physiol
.
2006
Dec
;
291
(
6
):
F1354
61
.
[PubMed]
1931-857X
93.
Quilty
JA
,
Li
J
,
Reithmeier
RA
.
Impaired trafficking of distal renal tubular acidosis mutants of the human kidney anion exchanger kAE1
.
Am J Physiol Renal Physiol
.
2002
May
;
282
(
5
):
F810
20
.
[PubMed]
1931-857X
94.
Kittanakom
S
,
Cordat
E
,
Reithmeier
RA
.
Dominant-negative effect of Southeast Asian ovalocytosis anion exchanger 1 in compound heterozygous distal renal tubular acidosis
.
Biochem J
.
2008
Mar
;
410
(
2
):
271
81
.
[PubMed]
0264-6021
95.
Ungsupravate
D
,
Sawasdee
N
,
Khositseth
S
,
Udomchaiprasertkul
W
,
Khoprasert
S
,
Li
J
, et al
Impaired trafficking and intracellular retention of mutant kidney anion exchanger 1 proteins (G701D and A858D) associated with distal renal tubular acidosis
.
Mol Membr Biol
.
2010
Apr
;
27
(
2-3
):
92
103
.
[PubMed]
0968-7688
96.
Nettuwakul
C
,
Sawasdee
N
,
Yenchitsomanus
PT
.
Rapid detection of solute carrier family 4, member 1 (SLC4A1) mutations and polymorphisms by high-resolution melting analysis
.
Clin Biochem
.
2010
Mar
;
43
(
4-5
):
497
504
.
[PubMed]
0009-9120
97.
Soleimani
M.
SLC26 Cl-/HCO3 - exchangers in the kidney: roles in health and disease. Kidney International advance online publication.
2013
; 2013(4).138
98.
Bissig
M
,
Hagenbuch
B
,
Stieger
B
,
Koller
T
,
Meier
PJ
.
Functional expression cloning of the canalicular sulfate transport system of rat hepatocytes
.
J Biol Chem
.
1994
Jan
;
269
(
4
):
3017
21
.
[PubMed]
0021-9258
99.
Karniski
LP
,
Lötscher
M
,
Fucentese
M
,
Hilfiker
H
,
Biber
J
,
Murer
H
.
Immunolocalization of sat-1 sulfate/oxalate/bicarbonate anion exchanger in the rat kidney
.
Am J Physiol
.
1998
Jul
;
275
(
1
):
F79
87
.
[PubMed]
0002-9513
100.
Quondamatteo
F
,
Krick
W
,
Hagos
Y
,
Krüger
MH
,
Neubauer-Saile
K
,
Herken
R
, et al
Localization of the sulfate/anion exchanger in the rat liver
.
Am J Physiol Gastrointest Liver Physiol
.
2006
May
;
290
(
5
):
G1075
81
.
[PubMed]
0193-1857
101.
Regeer
RR
,
Lee
A
,
Markovich
D
.
Characterization of the human sulfate anion transporter (hsat-1) protein and gene (SAT1; SLC26A1)
.
DNA Cell Biol
.
2003
Feb
;
22
(
2
):
107
17
.
[PubMed]
1044-5498
102.
Brzica
H
,
Breljak
D
,
Krick
W
,
Lovrić
M
,
Burckhardt
G
,
Burckhardt
BC
, et al
The liver and kidney expression of sulfate anion transporter sat-1 in rats exhibits male-dominant gender differences
.
Pflugers Arch
.
2009
Apr
;
457
(
6
):
1381
92
.
[PubMed]
0031-6768
103.
Breljak
D
,
Brzica
H
,
Vrhovac
I
,
Micek
V
,
Karaica
D
,
Ljubojević
M
, et al
In female rats, ethylene glycol treatment elevates protein expression of hepatic and renal oxalate transporter sat-1 (Slc26a1) without inducing hyperoxaluria
.
Croat Med J
.
2015
Oct
;
56
(
5
):
447
59
.
[PubMed]
0353-9504
104.
Costa-Bauzá
A
,
Ramis
M
,
Montesinos
V
,
Grases
F
,
Conte
A
,
Pizá
P
, et al
Type of renal calculi: variation with age and sex
.
World J Urol
.
2007
Aug
;
25
(
4
):
415
21
.
[PubMed]
0724-4983
105.
Markovich
D.
Slc13a1 and Slc26a1 KO models reveal physiological roles of anion transporters. union physiol. Sci./Am. Physiol. Soc.
2012
;1548-9213/12
106.
Dawson
PA
,
Russell
CS
,
Lee
S
,
McLeay
SC
,
van Dongen
JM
,
Cowley
DM
, et al
Urolithiasis and hepatotoxicity are linked to the anion transporter Sat1 in mice
.
J Clin Invest
.
2010
Mar
;
120
(
3
):
706
12
.
[PubMed]
0021-9738
107.
Gee
HY
,
Jun
I
,
Braun
DA
,
Lawson
JA
,
Halbritter
J
,
Shril
S
, et al
Mutations in SLC26A1 cause nephrolithiasis
.
Am J Hum Genet
.
2016
Jun
;
98
(
6
):
1228
34
.
[PubMed]
0002-9297
108.
Hästbacka
J
,
de la Chapelle
A
,
Mahtani
MM
,
Clines
G
,
Reeve-Daly
MP
,
Daly
M
, et al
The diastrophic dysplasia gene encodes a novel sulfate transporter: positional cloning by fine-structure linkage disequilibrium mapping
.
Cell
.
1994
Sep
;
78
(
6
):
1073
87
.
[PubMed]
0092-8674
109.
Dorwart
MR
,
Shcheynikov
N
,
Yang
D
,
Muallem
S
.
The solute carrier 26 family of proteins in epithelial ion transport
.
Physiology (Bethesda)
.
2008
Apr
;
23
(
2
):
104
14
.
[PubMed]
1548-9213
110.
Hästbacka
J
,
Superti-Furga
A
,
Wilcox
WR
,
Rimoin
DL
,
Cohn
DH
,
Lander
ES
.
Sulfate transport in chondrodysplasia
.
Ann N Y Acad Sci
.
1996
Jun
;
785
(
785
):
131
6
.
[PubMed]
0077-8923
111.
Markovich
D
,
Aronson
PS
.
Specificity and regulation of renal sulfate transporters
.
Annu Rev Physiol
.
2007
;
69
(
1
):
361
75
.
[PubMed]
0066-4278
112.
Dwyer
E
,
Hyland
J
,
Modaff
P
,
Pauli
RM
.
Genotype-phenotype correlation in DTDST dysplasias: atelosteogenesis type II and diastrophic dysplasia variant in one family
.
Am J Med Genet A
.
2010
Dec
;
152A
(
12
):
3043
50
.
[PubMed]
1552-4825
113.
Park
M
,
Ohana
E
,
Choi
SY
,
Lee
MS
,
Park
JH
,
Muallem
S
.
Multiple roles of the SO4(2-)/Cl-/OH- exchanger protein Slc26a2 in chondrocyte functions
.
J Biol Chem
.
2014
Jan
;
289
(
4
):
1993
2001
.
[PubMed]
0021-9258
114.
Ohana
E
,
Shcheynikov
N
,
Park
M
,
Muallem
S
.
Solute carrier family 26 member a2 (Slc26a2) protein functions as an electroneutral SOFormula/OH-/Cl- exchanger regulated by extracellular Cl-
.
J Biol Chem
.
2012
Feb
;
287
(
7
):
5122
32
.
[PubMed]
0021-9258
115.
Schweinfest
CW
,
Henderson
KW
,
Suster
S
,
Kondoh
N
,
Papas
TS
.
Identification of a colon mucosa gene that is down-regulated in colon adenomas and adenocarcinomas
.
Proc Natl Acad Sci USA
.
1993
May
;
90
(
9
):
4166
70
.
[PubMed]
0027-8424
116.
Freel
RW
,
Whittamore
JM
,
Hatch
M
.
Transcellular oxalate and Cl- absorption in mouse intestine is mediated by the DRA anion exchanger Slc26a3, and DRA deletion decreases urinary oxalate
.
Am J Physiol Gastrointest Liver Physiol
.
2013
Oct
;
305
(
7
):
G520
7
.
[PubMed]
0193-1857
117.
Stenson
PD
,
Mort
M
,
Ball
EV
,
Shaw
K
,
Phillips
A
,
Cooper
DN
.
The Human Gene Mutation Database: building a comprehensive mutation repository for clinical and molecular genetics, diagnostic testing and personalized genomic medicine
.
Hum Genet
.
2014
Jan
;
133
(
1
):
1
9
.
[PubMed]
0340-6717
118.
Schweinfest
CW
,
Spyropoulos
DD
,
Henderson
KW
,
Kim
JH
,
Chapman
JM
,
Barone
S
, et al
slc26a3 (dra)-deficient mice display chloride-losing diarrhea, enhanced colonic proliferation, and distinct up-regulation of ion transporters in the colon
.
J Biol Chem
.
2006
Dec
;
281
(
49
):
37962
71
.
[PubMed]
0021-9258
119.
Lohi
H
,
Lamprecht
G
,
Markovich
D
,
Heil
A
,
Kujala
M
,
Seidler
U
, et al
Isoforms of SLC26A6 mediate anion transport and have functional PDZ interaction domains
.
Am J Physiol Cell Physiol
.
2003
Mar
;
284
(
3
):
C769
79
.
[PubMed]
0363-6143
120.
Ohana
E
,
Shcheynikov
N
,
Yang
D
,
So
I
,
Muallem
S
.
Determinants of coupled transport and uncoupled current by the electrogenic SLC26 transporters
.
J Gen Physiol
.
2011
Feb
;
137
(
2
):
239
51
.
[PubMed]
0022-1295
121.
Chernova
MN
,
Jiang
L
,
Friedman
DJ
,
Darman
RB
,
Lohi
H
,
Kere
J
, et al
Functional comparison of mouse slc26a6 anion exchanger with human SLC26A6 polypeptide variants: differences in anion selectivity, regulation, and electrogenicity
.
J Biol Chem
.
2005
Mar
;
280
(
9
):
8564
80
.
[PubMed]
0021-9258
122.
Lohi
H
,
Kujala
M
,
Makela
S
,
Lehtonen
E
,
Kestila
M
,
Saarialho-Kere
U
, et al
Functional characterization of three novel tissue-specific anion exchangers SLC26A7, -A8, and -A9
.
J Biol Chem
.
2002
Apr
;
277
(
16
):
14246
54
.
[PubMed]
0021-9258
123.
Petrovic
S
,
Barone
S
,
Xu
J
,
Conforti
L
,
Ma
L
,
Kujala
M
, et al
SLC26A7: a basolateral Cl/ HCO3 cells of the outer medullary collecting duct
.
Am J Physiol Renal Physiol
.
2004
;
286
(
1
):
F161
9
.
[PubMed]
1931-857X
124.
Marengo
SR
,
Chen
DH
,
Evan
AP
,
Sommer
AJ
,
Stowe
NT
,
Ferguson
DG
, et al
Continuous infusion of oxalate by minipumps induces calcium oxalate nephrocalcinosis
.
Urol Res
.
2006
Jun
;
34
(
3
):
200
10
.
[PubMed]
0300-5623
125.
Evan
AP
,
Coe
FL
,
Lingeman
JE
,
Shao
Y
,
Sommer
AJ
,
Bledsoe
SB
, et al
Mechanism of formation of human calcium oxalate renal stones on Randall’s plaque
.
Anat Rec (Hoboken)
.
2007
Oct
;
290
(
10
):
1315
23
.
[PubMed]
1932-8486
126.
Xu
J
,
Song
P
,
Nakamura
S
,
Miller
M
,
Barone
S
,
Alper
SL
. Deletion of the chloride transporter slc26a7 causes distal renal tubular acidosis and impairs gastric acid secretion. J. Biol. Chem.
2009
.284 (43): 29470–29479.
Copyright / Drug Dosage / Disclaimer
Copyright: All rights reserved. No part of this publication may be translated into other languages, reproduced or utilized in any form or by any means, electronic or mechanical, including photocopying, recording, microcopying, or by any information storage and retrieval system, without permission in writing from the publisher.
Drug Dosage: The authors and the publisher have exerted every effort to ensure that drug selection and dosage set forth in this text are in accord with current recommendations and practice at the time of publication. However, in view of ongoing research, changes in government regulations, and the constant flow of information relating to drug therapy and drug reactions, the reader is urged to check the package insert for each drug for any changes in indications and dosage and for added warnings and precautions. This is particularly important when the recommended agent is a new and/or infrequently employed drug.
Disclaimer: The statements, opinions and data contained in this publication are solely those of the individual authors and contributors and not of the publishers and the editor(s). The appearance of advertisements or/and product references in the publication is not a warranty, endorsement, or approval of the products or services advertised or of their effectiveness, quality or safety. The publisher and the editor(s) disclaim responsibility for any injury to persons or property resulting from any ideas, methods, instructions or products referred to in the content or advertisements.