Comprehensive Physiology Wiley Online Library

Renal Tubular Handling of Glucose and Fructose in Health and Disease

Full Article on Wiley Online Library



Abstract

The proximal tubule of the kidney is programmed to reabsorb all filtered glucose and fructose. Glucose is taken up by apical sodium‐glucose cotransporters SGLT2 and SGLT1 whereas SGLT5 and potentially SGLT4 and GLUT5 have been implicated in apical fructose uptake. The glucose taken up by the proximal tubule is typically not metabolized but leaves via the basolateral facilitative glucose transporter GLUT2 and is returned to the systemic circulation or used as an energy source by distal tubular segments after basolateral uptake via GLUT1. The proximal tubule generates new glucose in metabolic acidosis and the postabsorptive phase, and fructose serves as an important substrate. In fact, under physiological conditions and intake, fructose taken up by proximal tubules is primarily utilized for gluconeogenesis. In the diabetic kidney, glucose is retained and gluconeogenesis enhanced, the latter in part driven by fructose. This is maladaptive as it sustains hyperglycemia. Moreover, renal glucose retention is coupled to sodium retention through SGLT2 and SGLT1, which induces secondary deleterious effects. SGLT2 inhibitors are new anti‐hyperglycemic drugs that can protect the kidneys and heart from failing independent of kidney function and diabetes. Dietary excess of fructose also induces tubular injury. This can be magnified by kidney formation of fructose under pathological conditions. Fructose metabolism is linked to urate formation, which partially accounts for fructose‐induced tubular injury, inflammation, and hemodynamic alterations. Fructose metabolism favors glycolysis over mitochondrial respiration as urate suppresses aconitase in the tricarboxylic acid cycle, and has been linked to potentially detrimental aerobic glycolysis (Warburg effect). © 2022 American Physiological Society. Compr Physiol 12:2995‐3044, 2022.

Figure 1. Figure 1. Glucose reabsorption in the kidney. (A) Under normoglycemia, SGLT2 in the early proximal tubule reabsorbs approximately 97% of filtered glucose. The remaining approximately 3% of glucose is reabsorbed by SGLT1 in the late proximal tubule, such that urine is nearly free of glucose. SGLT2 inhibition shifts glucose reabsorption downstream and unmasks the glucose reabsorption capacity of SGLT1 (≈40% of filtered glucose, depending on glucose load; see numbers in parentheses). (B) Cell model of glucose transport: The basolateral Na+‐K+‐ATPase lowers cytosolic Na+ concentrations and generates a negative interior voltage, thereby providing the driving force for Na+‐coupled glucose uptake through SGLT2 and SGLT1 across the apical membrane. The facilitative glucose transporter GLUT2 mediates glucose transport across the basolateral membrane down its chemical gradient. Basolateral GLUT1 may contribute to reabsorb glucose or take glucose up from peritubular space. Na+‐glucose cotransport is electrogenic and accompanied by paracellular Cl reabsorption or transcellular K+ secretion to stabilize membrane potential; K+ channels KCNE1/unknown α subunit and KCNE1/KCNQ1 in early and late proximal tubule, respectively. Modified, with permission, from Vallon V, 2011 391.
Figure 2. Figure 2. Tubular glucose reabsorption can be saturated. Tubular reabsorption of glucose increases linearly with the filtered glucose load up to the point when reabsorption reaches its maximum (Tmax glucose) and glucose starts to appear in urine. Theoretically in humans, a Tmax of approximately 350 mg/min and normal GFR would result in a plasma glucose threshold of approximately 280 mg/dL. The Tmax, however, varies between individual nephrons and, therefore, low‐level spilling of glucose into the urine initiates at modestly elevated plasma glucose levels of approximately 180 to 200 mg/dL in a healthy adult (see “Splay”). Normoglycemia is defined as fasted plasma glucose levels <100 mg/dL (<5.5 mM). SGLT2 inhibition reduces the renal glucose reabsorption to the transport capacity of SGLT1, that is, it reduces the renal glucose threshold (≈55–65 mg/dL) and Tmax (≈70 mg/min). Modified, with permission, from Vallon V, 2020 389.
Figure 3. Figure 3. Defining the contribution of SGLT2 and SGLT1 to renal glucose reabsorption. (A) Left two panels: free‐flow collections of tubular fluid were performed by micropuncture to establish a profile for fractional reabsorption of glucose versus fractional reabsorption of fluid along accessible proximal tubules at the kidney surface. Glucose reabsorption is prevented in the early proximal tubule in mice lacking SGLT2 (Sglt2−/−), but enhanced in the later proximal tubule, suggesting compensation by SGLT1. Right panel: In renal inulin clearance studies, the reduction in fractional renal glucose reabsorption in Sglt2−/− mice correlated with the amount of filtered glucose. (B) In metabolic cage studies, the SGLT2 inhibitor empagliflozin dose‐dependently increased glucose excretion in WT mice. The response curve was shifted leftward and the maximum response doubled in Sglt1−/− mice. The difference between the 2 dose‐response curves reflects glucose reabsorption via SGLT1 in WT mice. Glucosuria is initiated in WT mice when SGLT1‐mediated glucose uptake is maximal (red arrow). The difference between curves was maintained for all higher doses (same length of vertical green lines), indicating selectivity of the drug for SGLT2 versus SGLT1 in this dose range. (C) Using genetic knockout models and pharmacologic tools in renal inulin clearance studies indicated that the glucose reabsorption preserved during SGLT2 knockout or inhibition (≈40%) is mediated by SGLT1. The SGLT2 inhibitor empagliflozin was applied at low and high doses to establish free plasma concentrations (similar to concentrations in glomerular filtrate) close to IC50 for mouse SGLT2 (≈1–2 nM) or 10‐fold higher. Reused, with permission, from Rieg T, et al., 2014 313; Vallon V, et al., 2011 403.
Figure 4. Figure 4. Glucose and fructose metabolism. Most of the filtered glucose is taken up by SGLT2 in the S1 segment of the proximal tubule and leaves via basolateral GLUT2 (not shown). This is because the glycolytic potential is rather low in proximal tubules compared with further distal parts of the nephron and collecting duct system. In accordance, hexokinase (HK), the gateway enzyme of glucose metabolism that phosphorylates glucose to glucose‐6‐phosphate (Glc6P), is less active in proximal tubules relative to the rest of nephron and collecting duct. Within the proximal tubule, the highest expression and activity of HK (I, II) is in S2/S3 segments. Other key enzymes of glycolysis include phosphofructokinase (PFK) and pyruvate kinase (PKM). In contrast to glucose, fructose, which is primarily taken up by SGLT5 in S2/S3 segments, is readily phosphorylated by fructokinase (known as ketohexokinase, KHK) to produce fructose 1‐phosphate (Fru1P). Fru1P is subsequently cleaved by aldolase B (AldoB) into dihydroxyacetone phosphate (DHAP) and glyceraldehyde (GA). DHAP and GA feed into gluconeogenesis via fructose 1,6‐biphosphate (Fru1,6BP) or into glycolysis via glyceraldehyde‐3‐phosphate (G3P). G3P enters the glycolytic pathway distal to PFK and the formation of Fru1,6BP. While PFK is the most heavily regulated enzyme and is considered as the gating step of glycolysis, fructose metabolism bypasses this enzyme and lacks a negative regulatory step. In parallel, fructokinase activation sequesters a phosphate, so that intracellular phosphate and ATP levels are transiently reduced. The rapid reduction of phosphate consequently activates AMP deaminase (AMPD), which cleaves AMP to IMP. The decline in phosphate levels is attenuated by the relatively slower metabolism of Fru1P by AldoB. The latter is slowed down further by the increase in IMP, which inhibits AldoB. IMP metabolism drives urate formation. Fructose metabolism is linked to the pentose phosphate pathway (PPP) to generate nucleotides and amino acids, but also to lipid generation like triglycerides and cholesterol, and to lactate formation as an alternative energy form. Urate suppresses aconitase in TCA cycle, thereby favoring glycolysis over mitochondrial oxidative phosphorylation (OXPHOS), similar to Warburg effect. For gluconeogenesis, DHAP, G3P, and Fru1,6BP are metabolized toward glucose (red arrows). TK, triose kinase.
Figure 5. Figure 5. Coordination of glucose transport and gluconeogenesis in the proximal tubule. (1) Insulin is a physiological stimulator of SGLT2, which may serve to maximize renal glucose reabsorption capacity in situations of increased blood glucose levels, for example, following a meal. (2) At the same time, enhanced Na+‐glucose uptake and insulin suppress renal gluconeogenesis. (3) The latter, in contrast, is stimulated in the postabsorptive phase (fasting) by increased catecholamine and reduced insulin levels and involves primarily lactate as a precursor. (4) The newly formed glucose is delivered to the systemic circulation by basolateral GLUT2. (5) In metabolic acidosis, the increase in gluconeogenesis from glutamine is linked to the formation of (i) ammonium (NH4+), a renally excreted acid equivalent, and (ii) new bicarbonate, which is taken up into the circulation. The Na+‐H+‐exchanger NHE3 contributes to apical H+/NH4+ secretion and Na+/bicarbonate reabsorption. (6) The newly formed glucose can be used as fuel for proximal tubule H+ secretion or, after intercellular transfer, for intercalated cell H+ secretion. (7) SGLT2 and NHE3 are both stimulated by insulin to enhance Na+ and glucose reabsorption and their functions may be positively linked through the scaffolding protein MAP17. ?, indicates pathways that need further confirmation. Modified, with permission, from Vallon V, 2020 389.
Figure 6. Figure 6. Fructose from either diet or endogenous production can be utilized for survival. Fructose is natural fruits that stimulates fat accumulation, thereby contributing to the storage of energy that can be used when a lack of food is encountered. The freshwater Pacu fish actively feast on ripe fruits fallen into the river and become fat in the rainy season. Long‐distance migrating birds eat fruits prior to their migration to increase their fat stores. Hibernating mammals, such as bears and the ground squirrel, actively eat natural fruits to accumulate fat as an internal storage of energy for next winter season. Fresh fruits were the main dietary staple for early primates during the evolution of mankind. Moreover, pregnant women and other female mammals have the ability to produce endogenous fructose in the placenta, where fructose is used for fetal organ development during pregnancy. Naked mole rats can produce fructose endogenously in several organs for their survival under hypoxic condition. Adapted, with permission, from Johnson RJ, et al., 2019 161; Junk WJ, 1985 163.
Figure 7. Figure 7. Fructose transporters and metabolism in proximal tubular cells. There are several fructose transporters located at the apical membrane of proximal tubular epithelial cells, primarily in S2/S3 segments, including SGLT4/5 and GLUT5 as observed in rodents and humans. The rat sodium‐dependent glucose transporter‐1 (rNaGLT1) has been localized to the convoluted and the straight portions of proximal tubules in rats. In turn, GLUT2 is a facilitative transporter for fructose and glucose exit across the basolateral membrane. The proximal tubular cells are equipped with several enzymes for fructose metabolism, including fructokinase/ketohexokinase (FK/KHK), aldolase B (AldoB), and fructose 1,6 bisphosphatase (FBPase). Under fasting condition, a low level of fructose 2,6 biphosphate (Fru2,6BP) favors metabolism by FBPase over phosphofructokinase‐1 (PFK‐1) thereby metabolizing fructose 1,6 biphosphate (Fru1,6BP) toward gluconeogenesis. In turn, under satiation, a high level of Fru2,6BP activates PFK‐1 and promotes glycolysis. DHAP, dihydroxyacetone phosphate; G3P; glyceraldehyde 3‐phosphate; G6Pase, glucose 6‐phosphatase; GA, glyceraldehyde; HK, hexokinase; TK, triose kinase. Adapted, with permission, from Grempler R, et al., 2012 123; Fukuzawa T, et al., 2013.
Figure 8. Figure 8. Fructosuria in mice lacking Sglt5. (A) Plasma fructose concentrations measured under anesthesia after 3 h of fasting. Open circles represent individual mice. (B and C) Wild‐type (WT) mice and Sglt5‐deficient mice (Sglt5−/−) given plain water or 30% fructose water were maintained in metabolic cages and 24‐h urine samples collected. Despite similar plasma fructose concentrations, absolute urinary fructose excretion is significantly greater in Sglt5−/− vs WT mice given plain water, and the difference is further enhanced when given fructose water. Data are presented as means ± S.E.M (n = 8–10/group). +++, P < 0.001 versus WT mice given plain water. ***, P < 0.001 versus WT mice given fructose water. ## and ###, P < 0.01 and P < 0.001 versus respective plain water controls. Adapted, with permission, from Fukuzawa T, et al., 2013 95.
Figure 9. Figure 9. Cellular processes in the early proximal tubule linked to SGLT2 and its inhibition. Hyperglycemia enhances filtered glucose and, via SGLT2, the reabsorption of glucose and Na+ (1). Diabetes can increase SGLT2 expression (2); proposed mechanisms include tubular growth, Ang II, and HNF‐1α, which may respond to basolateral hyperglycemia sensed by GLUT2. Hyperinsulinemia and tubular growth upregulate proximal tubular transport systems, including SGLT2, NHE3, URAT1, and Na‐K‐ATPase (3). The apical transporters may be functionally coupled via scaffolding proteins, such as MAP17 (4). The resulting proximal tubular Na+ retention enhances the GFR via tubuloglomerular feedback, which by increasing brush border torque can further increase transporter density in the luminal membrane. The increase in intracellular glucose may lower SGLT2 expression via negative feedback (5). Diabetes, in part due to the associated acidosis, can enhance gluconeogenesis (6). Gluconeogenesis can be inhibited by tubular injury, hyperinsulinemia, and enhanced glucose uptake via SGLT2 (6). HNF‐1α and HNF‐3β upregulate GLUT2 (7) and thereby the basolateral exit of glucose and maintains hyperglycemia (8). Excessive SGLT2‐mediated glucose uptake may trigger apical translocation of GLUT2 (9). Hypoxia due to diabetes‐induced hyperreabsorption or kidney injury may induce HIF1α, which enhances basolateral glucose uptake via GLUT1, induces a metabolic shift to glycolysis and inhibits apical transport (10). Induction of TGF‐β1 and tubular growth may be particularly sensitive to basolateral glucose uptake via GLUT1 (11). Excessive intracellular glucose may also stimulate mTORC1 and attenuate autophagy (12). TGF‐β1 enhances cyclin‐dependent kinase inhibitors p21 and p27 and together with mTORC1 activation promotes tubular senescence, which is linked to inflammation and fibrosis. SGLT2 inhibition attenuates these deleterious effects linked to excessive intracellular glucose and hyperreabsorption. SGLT2 inhibition can also enhance gluconeogenesis, in part by lowering hyperinsulinemia. Gluconeogenesis enhances removal of intermediates from TCA cycle (cataplerosis) thereby facilitating the feeding of fatty acids and ketone bodies into the TCA cycle (anaplerosis), and enhancing oxidative phosphorylation (OxPhos) and ATP generation (13). This is associated with enhanced kidney delivery of fatty acids and ketone bodies in response to SGLT2 inhibition. Abbreviations: Ang II, angiotensin II; GFR, glomerular filtration rate; GLUT, facilitative glucose transporter; HIF‐1α, hypoxia‐inducible factor 1 alpha; HNF, hepatic nuclear factor; MAP17, 17‐kDa membrane‐associated protein; NHE3, Na‐H‐exchanger 3; OA, organic anion; TGF‐β1, transforming growth factor β1; URAT1, urate transporter 1. ?, indicates pathways that need further confirmation. Adapted, with permission, from Vallon V, 2020 389.
Figure 10. Figure 10. Basic differences in glucose versus fructose metabolism. Glucose metabolism in the kidney primarily occurs downstream of the proximal tubule and is determined by oxygen levels. (A) In the presence of sufficient oxygen (indicated by black arrow), glycolysis links to mitochondrial respiration/oxidative phosphorylation (OXPHOS) to efficiently produce high amounts of ATP. However, under low oxygen conditions (indicated by red arrow), mitochondrial respiration is disconnected from glycolysis, and lactate is produced. (B) In comparison, basic fructose metabolism is associated with glycolysis rather than mitochondrial respiration under either normoxia or hypoxia (indicated by blue arrow). A potential mechanism is that urate, a by product of fructose metabolism, suppresses aconitase (Aco), which converts citrate into iso‐citrate as part of the TCA cycle, and disconnects fructose metabolism from mitochondrial respiration.
Figure 11. Figure 11. A proposed deleterious role for SGLT1‐mediated reabsorption during recovery from ischemia‐reperfusion (IR)‐induced acute kidney injury. IR initially suppresses SGLT2 and SGLT1‐mediated reabsorption in the early and later proximal tubule, respectively, which is associated with glucosuria. Early recovery of SGLT1 expression and SGLT1‐mediated sodium reabsorption in late proximal tubule/outer medulla sustain IR‐induced hypoxia. This sustains cell injury in the outer medulla and the inhibition of NKCC2‐mediated NaCl reabsorption in the TAL, which impairs urine concentration and enhances Na‐Cl‐K delivery to macula densa ([Na‐Cl‐K]MD). The latter reduces renin expression and lowers GFR via tubuloglomerular feedback. The reduction in GFR enhances plasma creatinine and urea, the latter contributing to enhanced plasma osmolality. The sustained hypoxia and cell injury further enhance mitochondrial dysfunction, inflammation, and fibrosis, which can spread to the cortex and further suppress tubular function. Sustained suppression of SGLT2 maintains a high glucose load to downstream SGLT1, which may enhance the detrimental influence of SGLT1. Modified, with permission, from Nespoux J, et al., 2019 267.
Figure 12. Figure 12. Tubuloglomerular feedback, SGLT2 inhibition, and SGLT1 as a glucose sensor in the macula densa. (A) and (B) The tubuloglomerular feedback (TGF) establishes an inverse relationship between the delivery and concentration of Na‐Cl‐K at the macula densa and the single nephron GFR (SNGFR) of the same nephron. The operating point typically resides in the steepest part of the curve. In the diabetic kidney, a primary increase in proximal tubular reabsorption lowers Na‐Cl‐K delivery to the macula densa, which increases SNGFR through the physiology of TGF. SGLT2 inhibition attenuates proximal tubular reabsorption and increases Na‐Cl‐K delivery to the macula densa and lowers SNGFR through TGF. (C) (1 + 2) The macula densa senses an increase in luminal Na‐Cl‐K delivery by a NKCC2‐dependent mechanism, which then enhances the basolateral release of ATP. (3) ATP is converted by endonucleotidases CD73/39 to adenosine (ADO). (4) ADO activates the adenosine A1 receptor in vascular smooth muscle cells (VSMC) of the afferent arteriole to increase cytosolic Ca2+ and induce vasoconstriction and lower GFR. (5) ADO can also activate adenosine A2 receptors on VSMC of the efferent arteriole to reduce cytosolic Ca2+ and induce vasodilation. (6) Both effects contribute to the TGF mechanism and lower glomerular capillary pressure (PGC). (7) Due to upstream tubular hyperreabsorption, diabetes lowers Na‐Cl‐K delivery to the macula densa. SGLT2 inhibition attenuates the hyperreabsorption, increase Na‐Cl‐K delivery to the macula densa, and lowers GFR and PGC. (8) An increased Na‐Cl‐K delivery also activates nitric oxide synthase NOS1 in the macula densa. (9) The formed nitric oxide (NO) diffuses across the interstitium and dilates the afferent arteriole, thereby partially offsetting the afferent arteriolar vasoconstrictor tone of TGF. (10) When glucose delivery to the macula densa is increased, SGLT1 in the luminal membrane takes up glucose, a process that is linked to the phosphorylation, activation, and increased expression of NOS1 in the macula densa. The resulting enhanced NO tone dilates the afferent arteriole and enhances GFR. This can contribute to diabetes‐induced hyperfiltration, but also attenuate the reduction in GFR by SGLT2 inhibition. (11) On the other hand, SGLT2 inhibition can reduce macula densa NOS1/NO tone by inducing volume depletion. (12) ?, whether enhanced macula densa NO formation can also dilate the efferent arteriole remains to be determined. (C) Modified, with permission, from Vallon V, 2020 389.
Figure 13. Figure 13. The tubular hypothesis of diabetic glomerular hyperfiltration. (A) and (B) In vivo micropuncture studies in rats with superficial glomeruli were performed in nondiabetic and streptozotocin‐diabetic rats 404. Small amounts of blue dye were injected into Bowman space to determine nephron configuration, including the first proximal tubular loop and the early distal tubule close to the macula densa. Tubular fluid was collected close to the macula densa to determine the tubuloglomerular feedback signal ([Na‐Cl‐K]MD) and single nephron glomerular filtration rate (SNGFR; by inulin clearance). Bowman space was punctured to determine the hydrostatic pressure (PBow). Measurements were performed under control conditions and following application of the SGLT2/SGLT1 inhibitor phlorizin into the early proximal tubule, that is, without changing systemic blood glucose levels. Basal measurements (con) revealed that glomerular hyperfiltration in diabetes was associated with reductions in [Na‐Cl‐K]MD and PBow. Adding phlorizin (P) had a small effect in nondiabetic rats, but normalized [Na‐Cl‐K]MD, PBow, and SNGFR in diabetes. (C) Kidneys are programmed to retain glucose. As a consequence, diabetes induces a primary hyperreabsorption in proximal tubules involving enhanced Na+‐glucose cotransport and tubular growth. The concomitant enhanced reabsorption of sodium causes glomerular hyperfiltration through tubuloglomerular feedback ([Na‐Cl‐K]MD) and reducing tubular back pressure (PBow) thereby limiting sodium and volume retention. SGLT2 contributes to the tubular hyperreabsorption and as a consequence, SGLT2 inhibition mitigates these changes and lowers glomerular hyperfiltration. Modified, with permission, from Vallon V, and Thomson SC, 2017 410.
Figure 14. Figure 14. Proposed mechanisms of kidney protection by SGLT2 inhibition. SGLT2 inhibition counteracts the diabetes‐induced hyperreabsorption of glucose and Na+ in the early proximal tubule and lowers blood glucose levels. This also increases the NaCl and K concentration ([Na‐Cl‐K]MD) and fluid delivery (V) to the macula densa, which lowers glomerular filtration rate (GFR) through the physiology of tubuloglomerular feedback (1) and by increasing hydrostatic pressure in Bowman's space (PBow) (2). The GFR‐lowering effect of tubuloglomerular feedback includes afferent arteriole constriction (via adenosine A1 receptor) and potentially efferent arteriole dilation (via adenosine A2 receptor), which both reduce glomerular capillary pressure (PGC). Lowering of GFR reduces tubular transport work (3), thereby lowering cortical oxygen demand (QO2) (4) and increasing cortical oxygen tension (PO2) (5). Lowering GFR (6) and hyperglycemia (7) attenuates filtration of tubulo‐toxic compounds, including albumin, and reduces tubular growth and kidney inflammation. Tubular transport work is further reduced by lowering blood glucose and by cellular SGLT2 blockade itself, which reduces tubular glucotoxicity and has also been linked to inhibition of the Na‐H‐exchanger NHE3 (8). SGLT2 inhibition shifts glucose reabsorption downstream where SGLT1 compensates and reduces the risk of hypoglycemia (9). Shifting glucose and Na+ reabsorption downstream to S3 and mTAL segments increases QO2 (10) and lowers PO2 in the outer medulla (OM) (5). Furthermore, lower medullary PO2 may activate hypoxia‐inducible factor (HIF) and enhance erythropoietin (EPO) release (11). The latter increases hematocrit (Hct) (12) and improves O2 delivery to kidney medulla and cortex (13) and the heart (14). Enhanced delivery of NaCl and fluid downstream of early proximal tubule may enhance responsiveness to atrial natriuretic peptide (ANP) and diuretics (15). The diuretic, natriuretic and kaliuretic effects of SGLT2 inhibition lower the risk of hyperkalemia and further increase Hct (16) and reduce extracellular (ECV) and interstitial (ISV) volume and blood pressure (17). These effects, which are also evident by compensatory upregulation of renin and vasopressin levels (18), can help protect the failing kidney and heart (19). The increased cortical oxygen availability together with lesser hyperglycemia, tubular glucotoxicity, filtered albumin, and tubulointerstitial inflammation improves the integrity of the tubular and endothelial system, thereby allowing to maintain a higher tubular transport capacity and GFR in the long term (20). The glucosuric effect lowers therapeutic and/or endogenous insulin levels and increases glucagon concentrations (21). This induces compensatory lipolysis and hepatic gluconeogenesis and ketogenesis. SGLT2 inhibitors are uricosuric, potentially involving urate transporter 1 (URAT1) inhibition and their glucosuric and insulin‐lowering effect (22). These metabolic adaptations reduce urate levels, the hypoglycemia risk, and body and organ fat mass, which together with the resulting mild ketosis have the potential to further protect the kidney and heart (19, 23). The proposed effects are to a significant extend independent of hyperglycemia. Other abbreviations: NO, nitric oxide; UNaClV, urinary salt excretion; UV, urinary flow rate. Modified, with permission, from Vallon V, 2020 389.
Figure 15. Figure 15. Glomerular hemodynamic effects of SGLT2 inhibitor superimposed on a model of tubuloglomerular feedback (TGF), which incorporates effects on pre‐ and postglomerular resistances. Idealized TGF curves are shown for glomerular filtration rate (GFR) and PGC. The PGC curve lies to the right of the GFR curve owing to differences in the way that pre‐ and postglomerular resistances react across the range of inputs, as shown in the bottom portion of the figure. Within a group of nephrons, the position of operating points along the respective TGF curves will form a distribution. The figure shows the effects on GFR and PGC when identical increases in macula densa delivery are imposed by SGLT2 inhibitor for two nephrons drawn from this distribution (nephrons A and B). For nephron A, the TGF response to SGLT2 blocker included a large decrease in GFR and small decrease in PGC. For nephron B, there was a smaller decline in GFR and a larger decline in PGC. The inverse relationship between decreases in whole kidney GFR and decreases in average PGC for that animal is expected if animals with the smaller decreases in GFR yet larger decreases in PGC had more nephrons operating near the elbow of their respective TGF curves and vice versa. In other words, the effect of SGLT2 inhibition on GFR can over or underestimate effects on PGC depending on the location of the operating point. Modified, with permission, from Thomson SC, and Vallon V, 2021 375.
Figure 16. Figure 16. The integrated effects of SGLT1 in the diabetic kidney. (A) SGLT1 is expressed in the luminal membrane of the macula densa (MD) in human kidney. (B) Blue arrows indicate positive interactions. Hyperglycemia enhances filtered glucose and induces tubular growth. This increases Na+‐glucose cotransport, thereby maintaining hyperglycemia and reducing urinary Na+ and fluid excretion, with a larger contribution of SGLT2 versus SGLT1. Lesser urinary Na+ and fluid excretion increase effective circulating volume (ECV) and blood pressure (BP). Tubular hyperreabsorption lowers tubular backpressure in Bowman space (PBow) and the NaCl delivery and concentration at the MD, both increasing glomerular filtration rate (GFR) to restore urinary Na+ and fluid excretion. An increase in glucose delivery to the MD indicates saturated upstream Na+‐glucose cotransport. This is sensed by SGLT1 in the MD and, by stimulating MD nitric oxide synthase 1 (NOS1), further increases GFR to compensate for maximized Na+‐glucose cotransport. At the same time, SGLT1‐mediated glucose sensing may trigger tubular growth to enhance tubular glucose transport capacity. SGLT1 inhibition has a relatively small effect on diabetic tubular hyperreabsorption and thus induces little natriuresis and diuresis. Through inhibition of MD‐NOS1 upregulation and lowering of hyperfiltration, however, SGLT1 inhibition induces a relatively larger antinatriuretic and antidiuretic effect. As a consequence, SGLT1 inhibition can increase ECV with the resulting suppression in renin and increase in BP aim to restore renal Na+ and fluid excretion and ECV. (C) Diabetic Akita mice show enhanced MD NOS1 expression and higher GFR versus controls. Gene knockout of SGLT1 (Sglt1−/−) blunted both effects. (D) The classic TGF mechanism explains the increase in GFR when filtered glucose is increased up to the transport maximum for glucose (TM glucose). A further increase in filtered glucose can enhance MD NaCl and fluid delivery by the osmotic diuresis due to nonreabsorbed glucose, which would lower GFR. MD glucose sensing provides an additional stimulus to raise GFR and maintain hyperfiltration. EGM, extraglomerular mesangium, TALH, thick ascending limb. ?, indicate pathways that need further confirmation. Adapted, with permission, from Song P, et al., 2019 355.
Figure 17. Figure 17. Mechanisms by which excessive fructose causes kidney diseases. Under physiological conditions, fructose links to gluconeogenesis, and may protect from hypoxia and maintain systemic glucose concentrations, and preserves renal function. In turn, endogenous fructose formation is enhanced under several pathological conditions, including diabetes, ischemia, dehydration, senescence, and pressure overload. Either excessive dietary fructose or aberrant endogenous fructose generation stimulates the pathological pathway to cause tubulointerstitial damage and endothelial dysfunction in association with urate production and inflammation. Fructose metabolism links to the Warburg effect, which favors an unbalanced increase of glycolysis with suppressed mitochondrial respiration and stimulation of lactate production under aerobic conditions. These processes could be involved in the development of acute kidney injury (AKI), diabetic nephropathy, chronic kidney disease (CKD), senescence, or deleterious consequences of dehydration. Recurrent dehydration has been proposed to induce renal injury via a fructokinase‐dependent mechanism, likely from the generation of endogenous fructose via the polyol pathway. Adapted, with permission, from Roncal Jimenez CA, et al., 2014 317.


Figure 1. Glucose reabsorption in the kidney. (A) Under normoglycemia, SGLT2 in the early proximal tubule reabsorbs approximately 97% of filtered glucose. The remaining approximately 3% of glucose is reabsorbed by SGLT1 in the late proximal tubule, such that urine is nearly free of glucose. SGLT2 inhibition shifts glucose reabsorption downstream and unmasks the glucose reabsorption capacity of SGLT1 (≈40% of filtered glucose, depending on glucose load; see numbers in parentheses). (B) Cell model of glucose transport: The basolateral Na+‐K+‐ATPase lowers cytosolic Na+ concentrations and generates a negative interior voltage, thereby providing the driving force for Na+‐coupled glucose uptake through SGLT2 and SGLT1 across the apical membrane. The facilitative glucose transporter GLUT2 mediates glucose transport across the basolateral membrane down its chemical gradient. Basolateral GLUT1 may contribute to reabsorb glucose or take glucose up from peritubular space. Na+‐glucose cotransport is electrogenic and accompanied by paracellular Cl reabsorption or transcellular K+ secretion to stabilize membrane potential; K+ channels KCNE1/unknown α subunit and KCNE1/KCNQ1 in early and late proximal tubule, respectively. Modified, with permission, from Vallon V, 2011 391.


Figure 2. Tubular glucose reabsorption can be saturated. Tubular reabsorption of glucose increases linearly with the filtered glucose load up to the point when reabsorption reaches its maximum (Tmax glucose) and glucose starts to appear in urine. Theoretically in humans, a Tmax of approximately 350 mg/min and normal GFR would result in a plasma glucose threshold of approximately 280 mg/dL. The Tmax, however, varies between individual nephrons and, therefore, low‐level spilling of glucose into the urine initiates at modestly elevated plasma glucose levels of approximately 180 to 200 mg/dL in a healthy adult (see “Splay”). Normoglycemia is defined as fasted plasma glucose levels <100 mg/dL (<5.5 mM). SGLT2 inhibition reduces the renal glucose reabsorption to the transport capacity of SGLT1, that is, it reduces the renal glucose threshold (≈55–65 mg/dL) and Tmax (≈70 mg/min). Modified, with permission, from Vallon V, 2020 389.


Figure 3. Defining the contribution of SGLT2 and SGLT1 to renal glucose reabsorption. (A) Left two panels: free‐flow collections of tubular fluid were performed by micropuncture to establish a profile for fractional reabsorption of glucose versus fractional reabsorption of fluid along accessible proximal tubules at the kidney surface. Glucose reabsorption is prevented in the early proximal tubule in mice lacking SGLT2 (Sglt2−/−), but enhanced in the later proximal tubule, suggesting compensation by SGLT1. Right panel: In renal inulin clearance studies, the reduction in fractional renal glucose reabsorption in Sglt2−/− mice correlated with the amount of filtered glucose. (B) In metabolic cage studies, the SGLT2 inhibitor empagliflozin dose‐dependently increased glucose excretion in WT mice. The response curve was shifted leftward and the maximum response doubled in Sglt1−/− mice. The difference between the 2 dose‐response curves reflects glucose reabsorption via SGLT1 in WT mice. Glucosuria is initiated in WT mice when SGLT1‐mediated glucose uptake is maximal (red arrow). The difference between curves was maintained for all higher doses (same length of vertical green lines), indicating selectivity of the drug for SGLT2 versus SGLT1 in this dose range. (C) Using genetic knockout models and pharmacologic tools in renal inulin clearance studies indicated that the glucose reabsorption preserved during SGLT2 knockout or inhibition (≈40%) is mediated by SGLT1. The SGLT2 inhibitor empagliflozin was applied at low and high doses to establish free plasma concentrations (similar to concentrations in glomerular filtrate) close to IC50 for mouse SGLT2 (≈1–2 nM) or 10‐fold higher. Reused, with permission, from Rieg T, et al., 2014 313; Vallon V, et al., 2011 403.


Figure 4. Glucose and fructose metabolism. Most of the filtered glucose is taken up by SGLT2 in the S1 segment of the proximal tubule and leaves via basolateral GLUT2 (not shown). This is because the glycolytic potential is rather low in proximal tubules compared with further distal parts of the nephron and collecting duct system. In accordance, hexokinase (HK), the gateway enzyme of glucose metabolism that phosphorylates glucose to glucose‐6‐phosphate (Glc6P), is less active in proximal tubules relative to the rest of nephron and collecting duct. Within the proximal tubule, the highest expression and activity of HK (I, II) is in S2/S3 segments. Other key enzymes of glycolysis include phosphofructokinase (PFK) and pyruvate kinase (PKM). In contrast to glucose, fructose, which is primarily taken up by SGLT5 in S2/S3 segments, is readily phosphorylated by fructokinase (known as ketohexokinase, KHK) to produce fructose 1‐phosphate (Fru1P). Fru1P is subsequently cleaved by aldolase B (AldoB) into dihydroxyacetone phosphate (DHAP) and glyceraldehyde (GA). DHAP and GA feed into gluconeogenesis via fructose 1,6‐biphosphate (Fru1,6BP) or into glycolysis via glyceraldehyde‐3‐phosphate (G3P). G3P enters the glycolytic pathway distal to PFK and the formation of Fru1,6BP. While PFK is the most heavily regulated enzyme and is considered as the gating step of glycolysis, fructose metabolism bypasses this enzyme and lacks a negative regulatory step. In parallel, fructokinase activation sequesters a phosphate, so that intracellular phosphate and ATP levels are transiently reduced. The rapid reduction of phosphate consequently activates AMP deaminase (AMPD), which cleaves AMP to IMP. The decline in phosphate levels is attenuated by the relatively slower metabolism of Fru1P by AldoB. The latter is slowed down further by the increase in IMP, which inhibits AldoB. IMP metabolism drives urate formation. Fructose metabolism is linked to the pentose phosphate pathway (PPP) to generate nucleotides and amino acids, but also to lipid generation like triglycerides and cholesterol, and to lactate formation as an alternative energy form. Urate suppresses aconitase in TCA cycle, thereby favoring glycolysis over mitochondrial oxidative phosphorylation (OXPHOS), similar to Warburg effect. For gluconeogenesis, DHAP, G3P, and Fru1,6BP are metabolized toward glucose (red arrows). TK, triose kinase.


Figure 5. Coordination of glucose transport and gluconeogenesis in the proximal tubule. (1) Insulin is a physiological stimulator of SGLT2, which may serve to maximize renal glucose reabsorption capacity in situations of increased blood glucose levels, for example, following a meal. (2) At the same time, enhanced Na+‐glucose uptake and insulin suppress renal gluconeogenesis. (3) The latter, in contrast, is stimulated in the postabsorptive phase (fasting) by increased catecholamine and reduced insulin levels and involves primarily lactate as a precursor. (4) The newly formed glucose is delivered to the systemic circulation by basolateral GLUT2. (5) In metabolic acidosis, the increase in gluconeogenesis from glutamine is linked to the formation of (i) ammonium (NH4+), a renally excreted acid equivalent, and (ii) new bicarbonate, which is taken up into the circulation. The Na+‐H+‐exchanger NHE3 contributes to apical H+/NH4+ secretion and Na+/bicarbonate reabsorption. (6) The newly formed glucose can be used as fuel for proximal tubule H+ secretion or, after intercellular transfer, for intercalated cell H+ secretion. (7) SGLT2 and NHE3 are both stimulated by insulin to enhance Na+ and glucose reabsorption and their functions may be positively linked through the scaffolding protein MAP17. ?, indicates pathways that need further confirmation. Modified, with permission, from Vallon V, 2020 389.


Figure 6. Fructose from either diet or endogenous production can be utilized for survival. Fructose is natural fruits that stimulates fat accumulation, thereby contributing to the storage of energy that can be used when a lack of food is encountered. The freshwater Pacu fish actively feast on ripe fruits fallen into the river and become fat in the rainy season. Long‐distance migrating birds eat fruits prior to their migration to increase their fat stores. Hibernating mammals, such as bears and the ground squirrel, actively eat natural fruits to accumulate fat as an internal storage of energy for next winter season. Fresh fruits were the main dietary staple for early primates during the evolution of mankind. Moreover, pregnant women and other female mammals have the ability to produce endogenous fructose in the placenta, where fructose is used for fetal organ development during pregnancy. Naked mole rats can produce fructose endogenously in several organs for their survival under hypoxic condition. Adapted, with permission, from Johnson RJ, et al., 2019 161; Junk WJ, 1985 163.


Figure 7. Fructose transporters and metabolism in proximal tubular cells. There are several fructose transporters located at the apical membrane of proximal tubular epithelial cells, primarily in S2/S3 segments, including SGLT4/5 and GLUT5 as observed in rodents and humans. The rat sodium‐dependent glucose transporter‐1 (rNaGLT1) has been localized to the convoluted and the straight portions of proximal tubules in rats. In turn, GLUT2 is a facilitative transporter for fructose and glucose exit across the basolateral membrane. The proximal tubular cells are equipped with several enzymes for fructose metabolism, including fructokinase/ketohexokinase (FK/KHK), aldolase B (AldoB), and fructose 1,6 bisphosphatase (FBPase). Under fasting condition, a low level of fructose 2,6 biphosphate (Fru2,6BP) favors metabolism by FBPase over phosphofructokinase‐1 (PFK‐1) thereby metabolizing fructose 1,6 biphosphate (Fru1,6BP) toward gluconeogenesis. In turn, under satiation, a high level of Fru2,6BP activates PFK‐1 and promotes glycolysis. DHAP, dihydroxyacetone phosphate; G3P; glyceraldehyde 3‐phosphate; G6Pase, glucose 6‐phosphatase; GA, glyceraldehyde; HK, hexokinase; TK, triose kinase. Adapted, with permission, from Grempler R, et al., 2012 123; Fukuzawa T, et al., 2013.


Figure 8. Fructosuria in mice lacking Sglt5. (A) Plasma fructose concentrations measured under anesthesia after 3 h of fasting. Open circles represent individual mice. (B and C) Wild‐type (WT) mice and Sglt5‐deficient mice (Sglt5−/−) given plain water or 30% fructose water were maintained in metabolic cages and 24‐h urine samples collected. Despite similar plasma fructose concentrations, absolute urinary fructose excretion is significantly greater in Sglt5−/− vs WT mice given plain water, and the difference is further enhanced when given fructose water. Data are presented as means ± S.E.M (n = 8–10/group). +++, P < 0.001 versus WT mice given plain water. ***, P < 0.001 versus WT mice given fructose water. ## and ###, P < 0.01 and P < 0.001 versus respective plain water controls. Adapted, with permission, from Fukuzawa T, et al., 2013 95.


Figure 9. Cellular processes in the early proximal tubule linked to SGLT2 and its inhibition. Hyperglycemia enhances filtered glucose and, via SGLT2, the reabsorption of glucose and Na+ (1). Diabetes can increase SGLT2 expression (2); proposed mechanisms include tubular growth, Ang II, and HNF‐1α, which may respond to basolateral hyperglycemia sensed by GLUT2. Hyperinsulinemia and tubular growth upregulate proximal tubular transport systems, including SGLT2, NHE3, URAT1, and Na‐K‐ATPase (3). The apical transporters may be functionally coupled via scaffolding proteins, such as MAP17 (4). The resulting proximal tubular Na+ retention enhances the GFR via tubuloglomerular feedback, which by increasing brush border torque can further increase transporter density in the luminal membrane. The increase in intracellular glucose may lower SGLT2 expression via negative feedback (5). Diabetes, in part due to the associated acidosis, can enhance gluconeogenesis (6). Gluconeogenesis can be inhibited by tubular injury, hyperinsulinemia, and enhanced glucose uptake via SGLT2 (6). HNF‐1α and HNF‐3β upregulate GLUT2 (7) and thereby the basolateral exit of glucose and maintains hyperglycemia (8). Excessive SGLT2‐mediated glucose uptake may trigger apical translocation of GLUT2 (9). Hypoxia due to diabetes‐induced hyperreabsorption or kidney injury may induce HIF1α, which enhances basolateral glucose uptake via GLUT1, induces a metabolic shift to glycolysis and inhibits apical transport (10). Induction of TGF‐β1 and tubular growth may be particularly sensitive to basolateral glucose uptake via GLUT1 (11). Excessive intracellular glucose may also stimulate mTORC1 and attenuate autophagy (12). TGF‐β1 enhances cyclin‐dependent kinase inhibitors p21 and p27 and together with mTORC1 activation promotes tubular senescence, which is linked to inflammation and fibrosis. SGLT2 inhibition attenuates these deleterious effects linked to excessive intracellular glucose and hyperreabsorption. SGLT2 inhibition can also enhance gluconeogenesis, in part by lowering hyperinsulinemia. Gluconeogenesis enhances removal of intermediates from TCA cycle (cataplerosis) thereby facilitating the feeding of fatty acids and ketone bodies into the TCA cycle (anaplerosis), and enhancing oxidative phosphorylation (OxPhos) and ATP generation (13). This is associated with enhanced kidney delivery of fatty acids and ketone bodies in response to SGLT2 inhibition. Abbreviations: Ang II, angiotensin II; GFR, glomerular filtration rate; GLUT, facilitative glucose transporter; HIF‐1α, hypoxia‐inducible factor 1 alpha; HNF, hepatic nuclear factor; MAP17, 17‐kDa membrane‐associated protein; NHE3, Na‐H‐exchanger 3; OA, organic anion; TGF‐β1, transforming growth factor β1; URAT1, urate transporter 1. ?, indicates pathways that need further confirmation. Adapted, with permission, from Vallon V, 2020 389.


Figure 10. Basic differences in glucose versus fructose metabolism. Glucose metabolism in the kidney primarily occurs downstream of the proximal tubule and is determined by oxygen levels. (A) In the presence of sufficient oxygen (indicated by black arrow), glycolysis links to mitochondrial respiration/oxidative phosphorylation (OXPHOS) to efficiently produce high amounts of ATP. However, under low oxygen conditions (indicated by red arrow), mitochondrial respiration is disconnected from glycolysis, and lactate is produced. (B) In comparison, basic fructose metabolism is associated with glycolysis rather than mitochondrial respiration under either normoxia or hypoxia (indicated by blue arrow). A potential mechanism is that urate, a by product of fructose metabolism, suppresses aconitase (Aco), which converts citrate into iso‐citrate as part of the TCA cycle, and disconnects fructose metabolism from mitochondrial respiration.


Figure 11. A proposed deleterious role for SGLT1‐mediated reabsorption during recovery from ischemia‐reperfusion (IR)‐induced acute kidney injury. IR initially suppresses SGLT2 and SGLT1‐mediated reabsorption in the early and later proximal tubule, respectively, which is associated with glucosuria. Early recovery of SGLT1 expression and SGLT1‐mediated sodium reabsorption in late proximal tubule/outer medulla sustain IR‐induced hypoxia. This sustains cell injury in the outer medulla and the inhibition of NKCC2‐mediated NaCl reabsorption in the TAL, which impairs urine concentration and enhances Na‐Cl‐K delivery to macula densa ([Na‐Cl‐K]MD). The latter reduces renin expression and lowers GFR via tubuloglomerular feedback. The reduction in GFR enhances plasma creatinine and urea, the latter contributing to enhanced plasma osmolality. The sustained hypoxia and cell injury further enhance mitochondrial dysfunction, inflammation, and fibrosis, which can spread to the cortex and further suppress tubular function. Sustained suppression of SGLT2 maintains a high glucose load to downstream SGLT1, which may enhance the detrimental influence of SGLT1. Modified, with permission, from Nespoux J, et al., 2019 267.


Figure 12. Tubuloglomerular feedback, SGLT2 inhibition, and SGLT1 as a glucose sensor in the macula densa. (A) and (B) The tubuloglomerular feedback (TGF) establishes an inverse relationship between the delivery and concentration of Na‐Cl‐K at the macula densa and the single nephron GFR (SNGFR) of the same nephron. The operating point typically resides in the steepest part of the curve. In the diabetic kidney, a primary increase in proximal tubular reabsorption lowers Na‐Cl‐K delivery to the macula densa, which increases SNGFR through the physiology of TGF. SGLT2 inhibition attenuates proximal tubular reabsorption and increases Na‐Cl‐K delivery to the macula densa and lowers SNGFR through TGF. (C) (1 + 2) The macula densa senses an increase in luminal Na‐Cl‐K delivery by a NKCC2‐dependent mechanism, which then enhances the basolateral release of ATP. (3) ATP is converted by endonucleotidases CD73/39 to adenosine (ADO). (4) ADO activates the adenosine A1 receptor in vascular smooth muscle cells (VSMC) of the afferent arteriole to increase cytosolic Ca2+ and induce vasoconstriction and lower GFR. (5) ADO can also activate adenosine A2 receptors on VSMC of the efferent arteriole to reduce cytosolic Ca2+ and induce vasodilation. (6) Both effects contribute to the TGF mechanism and lower glomerular capillary pressure (PGC). (7) Due to upstream tubular hyperreabsorption, diabetes lowers Na‐Cl‐K delivery to the macula densa. SGLT2 inhibition attenuates the hyperreabsorption, increase Na‐Cl‐K delivery to the macula densa, and lowers GFR and PGC. (8) An increased Na‐Cl‐K delivery also activates nitric oxide synthase NOS1 in the macula densa. (9) The formed nitric oxide (NO) diffuses across the interstitium and dilates the afferent arteriole, thereby partially offsetting the afferent arteriolar vasoconstrictor tone of TGF. (10) When glucose delivery to the macula densa is increased, SGLT1 in the luminal membrane takes up glucose, a process that is linked to the phosphorylation, activation, and increased expression of NOS1 in the macula densa. The resulting enhanced NO tone dilates the afferent arteriole and enhances GFR. This can contribute to diabetes‐induced hyperfiltration, but also attenuate the reduction in GFR by SGLT2 inhibition. (11) On the other hand, SGLT2 inhibition can reduce macula densa NOS1/NO tone by inducing volume depletion. (12) ?, whether enhanced macula densa NO formation can also dilate the efferent arteriole remains to be determined. (C) Modified, with permission, from Vallon V, 2020 389.


Figure 13. The tubular hypothesis of diabetic glomerular hyperfiltration. (A) and (B) In vivo micropuncture studies in rats with superficial glomeruli were performed in nondiabetic and streptozotocin‐diabetic rats 404. Small amounts of blue dye were injected into Bowman space to determine nephron configuration, including the first proximal tubular loop and the early distal tubule close to the macula densa. Tubular fluid was collected close to the macula densa to determine the tubuloglomerular feedback signal ([Na‐Cl‐K]MD) and single nephron glomerular filtration rate (SNGFR; by inulin clearance). Bowman space was punctured to determine the hydrostatic pressure (PBow). Measurements were performed under control conditions and following application of the SGLT2/SGLT1 inhibitor phlorizin into the early proximal tubule, that is, without changing systemic blood glucose levels. Basal measurements (con) revealed that glomerular hyperfiltration in diabetes was associated with reductions in [Na‐Cl‐K]MD and PBow. Adding phlorizin (P) had a small effect in nondiabetic rats, but normalized [Na‐Cl‐K]MD, PBow, and SNGFR in diabetes. (C) Kidneys are programmed to retain glucose. As a consequence, diabetes induces a primary hyperreabsorption in proximal tubules involving enhanced Na+‐glucose cotransport and tubular growth. The concomitant enhanced reabsorption of sodium causes glomerular hyperfiltration through tubuloglomerular feedback ([Na‐Cl‐K]MD) and reducing tubular back pressure (PBow) thereby limiting sodium and volume retention. SGLT2 contributes to the tubular hyperreabsorption and as a consequence, SGLT2 inhibition mitigates these changes and lowers glomerular hyperfiltration. Modified, with permission, from Vallon V, and Thomson SC, 2017 410.


Figure 14. Proposed mechanisms of kidney protection by SGLT2 inhibition. SGLT2 inhibition counteracts the diabetes‐induced hyperreabsorption of glucose and Na+ in the early proximal tubule and lowers blood glucose levels. This also increases the NaCl and K concentration ([Na‐Cl‐K]MD) and fluid delivery (V) to the macula densa, which lowers glomerular filtration rate (GFR) through the physiology of tubuloglomerular feedback (1) and by increasing hydrostatic pressure in Bowman's space (PBow) (2). The GFR‐lowering effect of tubuloglomerular feedback includes afferent arteriole constriction (via adenosine A1 receptor) and potentially efferent arteriole dilation (via adenosine A2 receptor), which both reduce glomerular capillary pressure (PGC). Lowering of GFR reduces tubular transport work (3), thereby lowering cortical oxygen demand (QO2) (4) and increasing cortical oxygen tension (PO2) (5). Lowering GFR (6) and hyperglycemia (7) attenuates filtration of tubulo‐toxic compounds, including albumin, and reduces tubular growth and kidney inflammation. Tubular transport work is further reduced by lowering blood glucose and by cellular SGLT2 blockade itself, which reduces tubular glucotoxicity and has also been linked to inhibition of the Na‐H‐exchanger NHE3 (8). SGLT2 inhibition shifts glucose reabsorption downstream where SGLT1 compensates and reduces the risk of hypoglycemia (9). Shifting glucose and Na+ reabsorption downstream to S3 and mTAL segments increases QO2 (10) and lowers PO2 in the outer medulla (OM) (5). Furthermore, lower medullary PO2 may activate hypoxia‐inducible factor (HIF) and enhance erythropoietin (EPO) release (11). The latter increases hematocrit (Hct) (12) and improves O2 delivery to kidney medulla and cortex (13) and the heart (14). Enhanced delivery of NaCl and fluid downstream of early proximal tubule may enhance responsiveness to atrial natriuretic peptide (ANP) and diuretics (15). The diuretic, natriuretic and kaliuretic effects of SGLT2 inhibition lower the risk of hyperkalemia and further increase Hct (16) and reduce extracellular (ECV) and interstitial (ISV) volume and blood pressure (17). These effects, which are also evident by compensatory upregulation of renin and vasopressin levels (18), can help protect the failing kidney and heart (19). The increased cortical oxygen availability together with lesser hyperglycemia, tubular glucotoxicity, filtered albumin, and tubulointerstitial inflammation improves the integrity of the tubular and endothelial system, thereby allowing to maintain a higher tubular transport capacity and GFR in the long term (20). The glucosuric effect lowers therapeutic and/or endogenous insulin levels and increases glucagon concentrations (21). This induces compensatory lipolysis and hepatic gluconeogenesis and ketogenesis. SGLT2 inhibitors are uricosuric, potentially involving urate transporter 1 (URAT1) inhibition and their glucosuric and insulin‐lowering effect (22). These metabolic adaptations reduce urate levels, the hypoglycemia risk, and body and organ fat mass, which together with the resulting mild ketosis have the potential to further protect the kidney and heart (19, 23). The proposed effects are to a significant extend independent of hyperglycemia. Other abbreviations: NO, nitric oxide; UNaClV, urinary salt excretion; UV, urinary flow rate. Modified, with permission, from Vallon V, 2020 389.


Figure 15. Glomerular hemodynamic effects of SGLT2 inhibitor superimposed on a model of tubuloglomerular feedback (TGF), which incorporates effects on pre‐ and postglomerular resistances. Idealized TGF curves are shown for glomerular filtration rate (GFR) and PGC. The PGC curve lies to the right of the GFR curve owing to differences in the way that pre‐ and postglomerular resistances react across the range of inputs, as shown in the bottom portion of the figure. Within a group of nephrons, the position of operating points along the respective TGF curves will form a distribution. The figure shows the effects on GFR and PGC when identical increases in macula densa delivery are imposed by SGLT2 inhibitor for two nephrons drawn from this distribution (nephrons A and B). For nephron A, the TGF response to SGLT2 blocker included a large decrease in GFR and small decrease in PGC. For nephron B, there was a smaller decline in GFR and a larger decline in PGC. The inverse relationship between decreases in whole kidney GFR and decreases in average PGC for that animal is expected if animals with the smaller decreases in GFR yet larger decreases in PGC had more nephrons operating near the elbow of their respective TGF curves and vice versa. In other words, the effect of SGLT2 inhibition on GFR can over or underestimate effects on PGC depending on the location of the operating point. Modified, with permission, from Thomson SC, and Vallon V, 2021 375.


Figure 16. The integrated effects of SGLT1 in the diabetic kidney. (A) SGLT1 is expressed in the luminal membrane of the macula densa (MD) in human kidney. (B) Blue arrows indicate positive interactions. Hyperglycemia enhances filtered glucose and induces tubular growth. This increases Na+‐glucose cotransport, thereby maintaining hyperglycemia and reducing urinary Na+ and fluid excretion, with a larger contribution of SGLT2 versus SGLT1. Lesser urinary Na+ and fluid excretion increase effective circulating volume (ECV) and blood pressure (BP). Tubular hyperreabsorption lowers tubular backpressure in Bowman space (PBow) and the NaCl delivery and concentration at the MD, both increasing glomerular filtration rate (GFR) to restore urinary Na+ and fluid excretion. An increase in glucose delivery to the MD indicates saturated upstream Na+‐glucose cotransport. This is sensed by SGLT1 in the MD and, by stimulating MD nitric oxide synthase 1 (NOS1), further increases GFR to compensate for maximized Na+‐glucose cotransport. At the same time, SGLT1‐mediated glucose sensing may trigger tubular growth to enhance tubular glucose transport capacity. SGLT1 inhibition has a relatively small effect on diabetic tubular hyperreabsorption and thus induces little natriuresis and diuresis. Through inhibition of MD‐NOS1 upregulation and lowering of hyperfiltration, however, SGLT1 inhibition induces a relatively larger antinatriuretic and antidiuretic effect. As a consequence, SGLT1 inhibition can increase ECV with the resulting suppression in renin and increase in BP aim to restore renal Na+ and fluid excretion and ECV. (C) Diabetic Akita mice show enhanced MD NOS1 expression and higher GFR versus controls. Gene knockout of SGLT1 (Sglt1−/−) blunted both effects. (D) The classic TGF mechanism explains the increase in GFR when filtered glucose is increased up to the transport maximum for glucose (TM glucose). A further increase in filtered glucose can enhance MD NaCl and fluid delivery by the osmotic diuresis due to nonreabsorbed glucose, which would lower GFR. MD glucose sensing provides an additional stimulus to raise GFR and maintain hyperfiltration. EGM, extraglomerular mesangium, TALH, thick ascending limb. ?, indicate pathways that need further confirmation. Adapted, with permission, from Song P, et al., 2019 355.


Figure 17. Mechanisms by which excessive fructose causes kidney diseases. Under physiological conditions, fructose links to gluconeogenesis, and may protect from hypoxia and maintain systemic glucose concentrations, and preserves renal function. In turn, endogenous fructose formation is enhanced under several pathological conditions, including diabetes, ischemia, dehydration, senescence, and pressure overload. Either excessive dietary fructose or aberrant endogenous fructose generation stimulates the pathological pathway to cause tubulointerstitial damage and endothelial dysfunction in association with urate production and inflammation. Fructose metabolism links to the Warburg effect, which favors an unbalanced increase of glycolysis with suppressed mitochondrial respiration and stimulation of lactate production under aerobic conditions. These processes could be involved in the development of acute kidney injury (AKI), diabetic nephropathy, chronic kidney disease (CKD), senescence, or deleterious consequences of dehydration. Recurrent dehydration has been proposed to induce renal injury via a fructokinase‐dependent mechanism, likely from the generation of endogenous fructose via the polyol pathway. Adapted, with permission, from Roncal Jimenez CA, et al., 2014 317.
References
 1.Ackermann TF, Boini KM, Volkl H, Bhandaru M, Bareiss PM, Just L, Vallon V, Amann K, Kuhl D, Feng Y, Hammes HP, Lang F. SGK1‐sensitive renal tubular glucose reabsorption in diabetes. Am J Physiol Renal Physiol 296: F859‐F866, 2009.
 2.Agius L, Peak M. Intracellular binding of glucokinase in hepatocytes and translocation by glucose, fructose and insulin. Biochem J 296 (Pt 3): 785‐796, 1993.
 3.Ali T, Khan I, Simpson W, Prescott G, Townend J, Smith W, Macleod A. Incidence and outcomes in acute kidney injury: A comprehensive population‐based study. J Am Soc Nephrol 18: 1292‐1298, 2007.
 4.Alleyne GA, Scullard GH. Renal metabolic response to acid base changes. I. Enzymatic control of ammoniagenesis in the rat. J Clin Invest 48: 364‐370, 1969.
 5.Andres‐Hernando A, Jensen TJ, Kuwabara M, Orlicky DJ, Cicerchi C, Li N, Roncal‐Jimenez CA, Garcia GE, Ishimoto T, Maclean PS, Bjornstad P, Sanchez‐Lozada LG, Kanbay M, Nakagawa T, Johnson RJ, Lanaspa MA. Vasopressin mediates fructose‐induced metabolic syndrome by activating the V1b receptor. JCI Insight 6: e140848, 2021.
 6.Andres‐Hernando A, Li N, Cicerchi C, Inaba S, Chen W, Roncal‐Jimenez C, Le MT, Wempe MF, Milagres T, Ishimoto T, Fini M, Nakagawa T, Johnson RJ, Lanaspa MA. Protective role of fructokinase blockade in the pathogenesis of acute kidney injury in mice. Nat Commun 8: 14181, 2017.
 7.Andres‐Hernando A, Orlicky DJ, Kuwabara M, Ishimoto T, Nakagawa T, Johnson RJ, Lanaspa MA. Deletion of fructokinase in the liver or in the intestine reveals differential effects on sugar‐induced metabolic dysfunction. Cell Metab 32 (1): 117‐127, 2020.
 8.Aoyama M, Isshiki K, Kume S, Chin‐Kanasaki M, Araki H, Araki S, Koya D, Haneda M, Kashiwagi A, Maegawa H, Uzu T. Fructose induces tubulointerstitial injury in the kidney of mice. Biochem Biophys Res Commun 419: 244‐249, 2012.
 9.Ash SR, Cuppage FE. Shift toward anaerobic glycolysis in the regenerating rat kidney. Am J Pathol 60: 385‐402, 1970.
 10.Asipu A, Hayward BE, O'Reilly J, Bonthron DT. Properties of normal and mutant recombinant human ketohexokinases and implications for the pathogenesis of essential fructosuria. Diabetes 52: 2426‐2432, 2003.
 11.Augustin R, Carayannopoulos MO, Dowd LO, Phay JE, Moley JF, Moley KH. Identification and characterization of human glucose transporter‐like protein‐9 (GLUT9): Alternative splicing alters trafficking. J Biol Chem 279: 16229‐16236, 2004.
 12.Baartscheer A, Schumacher CA, Wust RC, Fiolet JW, Stienen GJ, Coronel R, Zuurbier CJ. Empagliflozin decreases myocardial cytoplasmic Na(+) through inhibition of the cardiac Na(+)/H(+) exchanger in rats and rabbits. Diabetologia 60: 568‐573, 2017.
 13.Bacon JS, Bell DJ. The identification of fructose as a constituent of the foetal blood of the sheep. Biochem J 40: xlii, 1946.
 14.Baena M, Sanguesa G, Davalos A, Latasa MJ, Sala‐Vila A, Sanchez RM, Roglans N, Laguna JC, Alegret M. Fructose, but not glucose, impairs insulin signaling in the three major insulin‐sensitive tissues. Sci Rep 6: 26149, 2016.
 15.Bairlein F. How to get fat: Nutritional mechanisms of seasonal fat accumulation in migratory songbirds. Naturwissenschaften 89: 1‐10, 2002.
 16.Bakris G, Oshima M, Mahaffey KW, Agarwal R, Cannon CP, Capuano G, Charytan DM, de Zeeuw D, Edwards R, Greene T, Heerspink HJL, Levin A, Neal B, Oh R, Pollock C, Rosenthal N, Wheeler DC, Zhang H, Zinman B, Jardine MJ, Perkovic V. Effects of canagliflozin in patients with baseline eGFR <30 ml/min per 1.73 m(2): Subgroup analysis of the randomized CREDENCE trial. Clin J Am Soc Nephrol 15: 1705‐1714, 2020.
 17.Balen D, Ljubojevic M, Breljak D, Brzica H, Zlender V, Koepsell H, Sabolic I. Revised immunolocalization of the Na+‐D‐glucose cotransporter SGLT1 in rat organs with an improved antibody. Am J Physiol Cell Physiol 295: C475‐C489, 2008.
 18.Bankir L, Yang B. New insights into urea and glucose handling by the kidney, and the urine concentrating mechanism. Kidney Int 81: 1179‐1198, 2012.
 19.Barfuss DW, Schafer JA. Differences in active and passive glucose transport along the proximal nephron. Am J Physiol 241: F322‐F332, 1981.
 20.Barklay H, Haas P, Huggett ASG, King G, Rowley D. The sugar of the foetal blood, the amniotic and allantoic fluids. J Physiol 109: 98‐102, 1949.
 21.Barnett AH, Mithal A, Manassie J, Jones R, Rattunde H, Woerle HJ, Broedl UC. Efficacy and safety of empagliflozin added to existing antidiabetes treatment in patients with type 2 diabetes and chronic kidney disease: A randomised, double‐blind, placebo‐controlled trial. Lancet Diabetes Endocrinol 2: 369‐384, 2014.
 22.Barone S, Fussell SL, Singh AK, Lucas F, Xu J, Kim C, Wu X, Yu Y, Amlal H, Seidler U, Zuo J, Soleimani M. Slc2a5 (Glut5) is essential for the absorption of fructose in the intestine and generation of fructose‐induced hypertension. J Biol Chem 284: 5056‐5066, 2009.
 23.Basile DP, Anderson MD, Sutton TA. Pathophysiology of acute kidney injury. Compr Physiol 2: 1303‐1353, 2012.
 24.Benoy MP, Elliott KA. The metabolism of lactic and pyruvic acids in normal and tumour tissues: Synthesis of carbohydrate. Biochem J 31: 1268‐1275, 1937.
 25.Bessho R, Takiyama Y, Takiyama T, Kitsunai H, Takeda Y, Sakagami H, Ota T. Hypoxia‐inducible factor‐1alpha is the therapeutic target of the SGLT2 inhibitor for diabetic nephropathy. Sci Rep 9: 14754, 2019.
 26.Biava C, Grossman A, West M. Ultrastructural observations on renal glycogen in normal and pathologic human kidneys. Lab Invest 15: 330‐356, 1966.
 27.Bjorkman O, Felig P. Role of the kidney in the metabolism of fructose in 60‐hour fasted humans. Diabetes 31: 516‐520, 1982.
 28.Blantz RC, Peterson OW, Gushwa L, Tucker BJ. Effect of modest hyperglycemia on tubuloglomerular feedback activity. Kidney Int Suppl 12: S206‐S212, 1982.
 29.Blantz RC, Vallon V. Tubuloglomerular feedback responses of the downstream efferent resistance: Unmasking a role for adenosine? Kidney Int 71: 837‐839, 2007.
 30.Bomback AS, Katz R, He K, Shoham DA, Burke GL, Klemmer PJ. Sugar‐sweetened beverage consumption and the progression of chronic kidney disease in the multi‐ethnic study of atherosclerosis (MESA). Am J Clin Nutr 90: 1172‐1178, 2009.
 31.Bonventre JV, Yang L. Cellular pathophysiology of ischemic acute kidney injury. J Clin Invest 121: 4210‐4221, 2011.
 32.Borges‐Junior FA, Silva Dos Santos D, Benetti A, Polidoro JZ, Wisnivesky ACT, Crajoinas RO, Antonio EL, Jensen L, Caramelli B, Malnic G, Tucci PJ, ACC G. Empagliflozin inhibits proximal tubule NHE3 activity, preserves GFR, and restores euvolemia in nondiabetic rats with induced heart failure. J Am Soc Nephrol Apr 12:ASN.2020071029, 2021. DOI: 10.2020071681/ASN.2020071029.
 33.Bowman RH. Gluconeogenesis in the isolated perfused rat kidney. J Biol Chem 245: 1604‐1612, 1970.
 34.Brasen JC, Burford JL, McDonough AA, Holstein‐Rathlou NH, Peti‐Peterdi J. Local pH domains regulate NHE3‐mediated Na(+) reabsorption in the renal proximal tubule. Am J Physiol Renal Physiol 307: F1249‐F1262, 2014.
 35.Brezis M, Rosen S. Hypoxia of the renal medulla–its implications for disease. N Engl J Med 332: 647‐655, 1995.
 36.Brown KS, Kalinowski SS, Megill JR, Durham SK, Mookhtiar KA. Glucokinase regulatory protein may interact with glucokinase in the hepatocyte nucleus. Diabetes 46: 179‐186, 1997.
 37.Brownlee M. Biochemistry and molecular cell biology of diabetic complications. Nature 414: 813‐820, 2001.
 38.Brymora A, Flisinski M, Johnson RJ, Goszka G, Stefanska A, Manitius J. Low‐fructose diet lowers blood pressure and inflammation in patients with chronic kidney disease. Nephrol Dial Transplant 27: 608‐612, 2012.
 39.Burch HB, Choi S, Dence CN, Alvey TR, Cole BR, Lowry OH. Metabolic effects of large fructose loads in different parts of the rat nephron. J Biol Chem 255: 8239‐8244, 1980.
 40.Burch HB, Lowry OH, Meinhardt L, Max P Jr, Chyu K. Effect of fructose, dihydroxyacetone, glycerol, and glucose on metabolites and related compounds in liver and kidney. J Biol Chem 245: 2092‐2102, 1970.
 41.Burch HB, Narins RG, Chu C, Fagioli S, Choi S, McCarthy W, Lowry OH. Distribution along the rat nephron of three enzymes of gluconeogenesis in acidosis and starvation. Am J Physiol 235: F246‐F253, 1978.
 42.Burckhardt BC, Burckhardt G. Transport of organic anions across the basolateral membrane of proximal tubule cells. Rev Physiol Biochem Pharmacol 146: 95‐158, 2003.
 43.Cabral PD, Hong NJ, Hye Khan MA, Ortiz PA, Beierwaltes WH, Imig JD, Garvin JL. Fructose stimulates Na/H exchange activity and sensitizes the proximal tubule to angiotensin II. Hypertension 63: e68‐e73, 2014.
 44.Cai T, Ke Q, Fang Y, Wen P, Chen H, Yuan Q, Luo J, Zhang Y, Sun Q, Lv Y, Zen K, Jiang L, Zhou Y, Yang J. Sodium‐glucose cotransporter 2 inhibition suppresses HIF‐1alpha‐mediated metabolic switch from lipid oxidation to glycolysis in kidney tubule cells of diabetic mice. Cell Death Dis 11: 390, 2020.
 45.Caliceti C, Calabria D, Roda A, Cicero AFG. Fructose intake, serum uric acid, and cardiometabolic disorders: A critical review. Nutrients 9: 395, 2017.
 46.Cammisotto PG, Londono I, Gingras D, Bendayan M. Control of glycogen synthase through ADIPOR1‐AMPK pathway in renal distal tubules of normal and diabetic rats. Am J Physiol Renal Physiol 294: F881‐F889, 2008.
 47.Carey HV, Andrews MT, Martin SL. Mammalian hibernation: Cellular and molecular responses to depressed metabolism and low temperature. Physiol Rev 83: 1153‐1181, 2003.
 48.Cassis P, Locatelli M, Cerullo D, Corna D, Buelli S, Zanchi C, Villa S, Morigi M, Remuzzi G, Benigni A, Zoja C. SGLT2 inhibitor dapagliflozin limits podocyte damage in proteinuric nondiabetic nephropathy. JCI Insight 3, 2018.
 49.Chamberlin ME, LeFurgey A, Mandel LJ. Suspension of medullary thick ascending limb tubules from the rabbit kidney. Am J Physiol 247: F955‐F964, 1984.
 50.Chang YK, Choi H, Jeong JY, Na KR, Lee KW, Lim BJ, Choi DE. Dapagliflozin, SGLT2 inhibitor, attenuates renal ischemia‐reperfusion injury. PLoS One 11: e0158810, 2016.
 51.Chen H, Busse LW. Novel therapies for acute kidney injury. Kidney Int Rep 2: 785‐799, 2017.
 52.Chen J, Williams S, Ho S, Loraine H, Hagan D, Whaley JM, Feder JN. Quantitative PCR tissue expression profiling of the human SGLT2 gene and related family members. Diabetes Ther 1: 57‐92, 2010.
 53.Cherney DZ, Perkins BA, Soleymanlou N, Maione M, Lai V, Lee A, Fagan NM, Woerle HJ, Johansen OE, Broedl UC, Von Eynatten M. Renal hemodynamic effect of sodium‐glucose cotransporter 2 inhibition in patients with type 1 diabetes mellitus. Circulation 129: 587‐597, 2014.
 54.Chin E, Zamah AM, Landau D, Gronbcek H, Flyvbjerg A, LeRoith D, Bondy CA. Changes in facilitative glucose transporter messenger ribonucleic acid levels in the diabetic rat kidney. Endocrinology 138: 1267‐1275, 1997.
 55.Chin E, Zhou J, Bondy C. Anatomical and developmental patterns of facilitative glucose transporter gene expression in the rat kidney. J Clin Invest 91: 1810‐1815, 1993.
 56.Chino Y, Samukawa Y, Sakai S, Nakai Y, Yamaguchi JI, Nakanishi T, Tamai I. SGLT2 inhibitor lowers serum uric acid through alteration of uric acid transport activity in renal tubule by increased glycosuria. Biopharm Drug Dispos 35: 391‐404, 2014.
 57.Choi YJ, Yoon Y, Lee KY, Hien TT, Kang KW, Kim KC, Lee J, Lee MY, Lee SM, Kang DH, Lee BH. Uric acid induces endothelial dysfunction by vascular insulin resistance associated with the impairment of nitric oxide synthesis. FASEB J 28: 3197‐3204, 2014.
 58.Cirillo P, Gersch MS, Mu W, Scherer PM, Kim KM, Gesualdo L, Henderson GN, Johnson RJ, Sautin YY. Ketohexokinase‐dependent metabolism of fructose induces proinflammatory mediators in proximal tubular cells. J Am Soc Nephrol 20: 545‐553, 2009.
 59.Coady MJ, El TA, Santer R, Bissonnette P, Sasseville LJ, Calado J, Lussier Y, Dumayne C, Bichet DG, Lapointe JY. MAP17 is a necessary activator of renal Na+/Glucose cotransporter SGLT2. J Am Soc Nephrol 28: 85‐93, 2017.
 60.Coffee EM, Tolan DR. Gluconeogenesis. In: LEE B, Scaglia F, editors. Inborn Errors in Metabolism: From Neonatal Screening to Metabolic Pathways. New York: Oxford University Press, 2014, p. 68‐91.
 61.Cohn C, Kolinsky M. Effect of blood sugar levels and insulin lack on gluconeogenesis by the kidney of the dog. Am J Physiol 156: 345‐348, 1949.
 62.Collazo R, Fan L, Hu MC, Zhao H, Wiederkehr MR, Moe OW. Acute regulation of Na+/H+ exchanger NHE3 by parathyroid hormone via NHE3 phosphorylation and dynamin‐dependent endocytosis. J Biol Chem 275: 31601‐31608, 2000.
 63.Conjard A, Martin M, Guitton J, Baverel G, Ferrier B. Gluconeogenesis from glutamine and lactate in the isolated human renal proximal tubule: Longitudinal heterogeneity and lack of response to adrenaline. Biochem J 360: 371‐377, 2001.
 64.Darshi M, Onishi A, Kim JJ, Pham J, Van Espen BF, Murphy A, Heerspink HJl, Vallon V, Sharma K. Metabolic reprogramming in diabetic kidney disease can be restored via SGLT2 inhibition. J Am Soc Nephrol 439: 439 (Abstract), 2017.
 65.Dekkers CCJ, Petrykiv S, Laverman GD, Cherney DZ, Gansevoort RT, Heerspink HJL. Effects of the SGLT‐2 inhibitor dapagliflozin on glomerular and tubular injury markers. Diabetes Obes Metab 20: 1988‐1993, 2018.
 66.Diggle CP, Shires M, Leitch D, Brooke D, Carr IM, Markham AF, Hayward BE, Asipu A, Bonthron DT. Ketohexokinase: Expression and localization of the principal fructose‐metabolizing enzyme. J Histochem Cytochem 57: 763‐774, 2009.
 67.Ding H, Jiang L, Xu J, Bai F, Zhou Y, Yuan Q, Luo J, Zen K, Yang J. Inhibiting aerobic glycolysis suppresses renal interstitial fibroblast activation and renal fibrosis. Am J Physiol Renal Physiol 313: F561‐F575, 2017.
 68.Doblado M, Moley KH. Facilitative glucose transporter 9, a unique hexose and urate transporter. Am J Physiol Endocrinol Metab 297: E831‐E835, 2009.
 69.Dominguez JH, Camp K, Maianu L, Feister H, Garvey WT. Molecular adaptations of GLUT1 and GLUT2 in renal proximal tubules of diabetic rats. Am J Physiol 266: F283‐F290, 1994.
 70.Dominguez JH, Camp K, Maianu L, Garvey WT. Glucose transporters of rat proximal tubule: Differential expression and subcellular distribution. Am J Physiol 262: F807‐F812, 1992.
 71.Douard V, Ferraris RP. Regulation of the fructose transporter GLUT5 in health and disease. Am J Physiol Endocrinol Metab 295: E227‐E237, 2008.
 72.Douard V, Ferraris RP. The role of fructose transporters in diseases linked to excessive fructose intake. J Physiol 591: 401‐414, 2013.
 73.Drewnowsk KD, Craig MR, Digiovanni SR, McCarty JM, Moorman AF, Lamers WH, Schoolwerth AC. PEPCK mRNA localization in proximal tubule and gene regulation during metabolic acidosis. J Physiol Pharmacol 53: 3‐20, 2002.
 74.Ehrenkranz JR, Lewis NG, Kahn CR, Roth J. Phlorizin: A review. Diabetes Metab Res Rev 21: 31‐38, 2005.
 75.Eickhoff MK, Dekkers CCJ, Kramers BJ, Laverman GD, Frimodt‐Moller M, Jorgensen NR, Faber J, Danser AHJ, Gansevoort RT, Rossing P, Persson F, Heerspink HJL. Effects of dapagliflozin on volume status when added to renin‐angiotensin system inhibitors. J Clin Med 8: 799, 2019.
 76.Eskandari S, Wright EM, Loo DD. Kinetics of the reverse mode of the Na+/glucose cotransporter. J Membr Biol 204: 23‐32, 2005.
 77.Faham S, Watanabe A, Besserer GM, Cascio D, Specht A, Hirayama BA, Wright EM, Abramson J. The crystal structure of a sodium galactose transporter reveals mechanistic insights into Na+/sugar symport. Science 321: 810‐814, 2008.
 78.Farber SJ, Berger EY, Earle DP. Effect of diabetes and insulin on the maximum capacity of the renal tubules to reabsorb glucose. J Clin Invest 30: 125‐129, 1951.
 79.Farsijani NM, Liu Q, Kobayashi H, Davidoff O, Sha F, Fandrey J, Ikizler TA, O'Connor PM, Haase VH. Renal epithelium regulates erythropoiesis via HIF‐dependent suppression of erythropoietin. J Clin Invest 126: 1425‐1437, 2016.
 80.Fattah H, Shigeoka A, Huang W, Patel R, Kasimsetty S, Singh P, McKay DB, Vallon V. Diverse gene expression patterns of renal transporters in AKI. J Am Soc Nephrol 29: 1045 (Abstract), 2018.
 81.Fattah H, Vallon V. The potential role of SGLT2 inhibitors in the treatment of type 1 diabetes mellitus. Drugs 78: 717‐726, 2018.
 82.Ferrannini E. Sodium‐glucose co‐transporters and their inhibition: Clinical physiology. Cell Metab 26: 27‐38, 2017.
 83.Ferrannini E, Mark M, Mayoux E. CV protection in the EMPA‐REG OUTCOME trial: A "Thrifty Substrate" Hypothesis. Diabetes Care 39: 1108‐1114, 2016.
 84.Ferrannini E, Muscelli E, Frascerra S, Baldi S, Mari A, Heise T, Broedl UC, Woerle HJ. Metabolic response to sodium‐glucose cotransporter 2 inhibition in type 2 diabetic patients. J Clin Invest 124: 499‐508, 2014.
 85.Ferrannini E, Pereira‐Moreira R, Seghieri M, Rebelos E, Souza AL, Chueire VB, Arvia C, Muscelli E. Insulin enhances renal glucose excretion: Relation to insulin sensitivity and sodium‐glucose cotransport. BMJ Open Diabetes Res Care 8: e001178, 2020.
 86.Finch CE, Stanford CB. Meat‐adaptive genes and the evolution of slower aging in humans. Q Rev Biol 79: 3‐50, 2004.
 87.Fiorentino M, Grandaliano G, Gesualdo L, Castellano G. Acute kidney injury to chronic kidney disease transition. Contrib Nephrol 193: 45‐54, 2018.
 88.Fischer KF, Lees JA, Newman JH. Hypoglycemia in hospitalized patients. Causes and outcomes. N Engl J Med 315: 1245‐1250, 1986.
 89.Foote C, Perkovic V, Neal B. Effects of SGLT2 inhibitors on cardiovascular outcomes. Diab Vasc Dis Res 9: 117‐123, 2012.
 90.Freitas HS, Anhe GF, Melo KF, Okamoto MM, Oliveira‐Souza M, Bordin S, Machado UF. Na(+) ‐glucose transporter‐2 messenger ribonucleic acid expression in kidney of diabetic rats correlates with glycemic levels: Involvement of hepatocyte nuclear factor‐1alpha expression and activity. Endocrinology 149: 717‐724, 2008.
 91.Freitas HS, Schaan BD, David‐Silva A, Sabino‐Silva R, Okamoto MM, Alves‐Wagner AB, Mori RC, Machado UF. SLC2A2 gene expression in kidney of diabetic rats is regulated by HNF‐1alpha and HNF‐3beta. Mol Cell Endocrinol 305: 63‐70, 2009.
 92.Frohnert PP, Hohmann B, Zwiebel R, Baumann K. Free flow micropuncture studies of glucose transport in the rat nephron. Pflugers Arch 315: 66‐85, 1970.
 93.Fu Y, Breljak D, Onishi A, Batz F, Patel R, Huang W, Song P, Freeman B, Mayoux E, Koepsell H, Anzai N, Nigam SK, Sabolic I, Vallon V. The organic anion transporter OAT3 enhances the glucosuric effect of the SGLT2 inhibitor empagliflozin. Am J Physiol Renal Physiol 315: F386‐F394, 2018.
 94.Fu Y, Gerasimova M, Mayoux E, Masuda T, Vallon V. SGLT2 inhibitor empagliflozin increases renal NHE3 phosphorylation in diabetic Akita mice: Possible implications for the prevention of glomerular hyperfiltration. Diabetes 63 (supplement 1): A132, 2014.
 95.Fukuzawa T, Fukazawa M, Ueda O, Shimada H, Kito A, Kakefuda M, Kawase Y, Wada NA, Goto C, Fukushima N, Jishage K, Honda K, King GL, Kawabe Y. SGLT5 reabsorbs fructose in the kidney but its deficiency paradoxically exacerbates hepatic steatosis induced by fructose. PLoS One 8: e56681, 2013.
 96.Gaby AR. Adverse effects of dietary fructose. Altern Med Rev 10: 294‐306, 2005.
 97.Gall JM, Wong V, Pimental DR, Havasi A, Wang Z, Pastorino JG, Bonegio RG, Schwartz JH, Borkan SC. Hexokinase regulates Bax‐mediated mitochondrial membrane injury following ischemic stress. Kidney Int 79: 1207‐1216, 2011.
 98.Gallo LA, Ward MS, Fotheringham AK, Zhuang A, Borg DJ, Flemming NB, Harvie BM, Kinneally TL, Yeh SM, McCarthy DA, Koepsell H, Vallon V, Pollock C, Panchapakesan U, Forbes JM. Once daily administration of the SGLT2 inhibitor, empagliflozin, attenuates markers of renal fibrosis without improving albuminuria in diabetic db/db mice. Sci Rep 6: 26428, 2016.
 99.Gallo LA, Wright EM, Vallon V. Probing SGLT2 as a therapeutic target for diabetes: Basic physiology and consequences. Diab Vasc Dis Res 12: 78‐89, 2015.
 100.Geerlings S, Fonseca V, Castro‐Diaz D, List J, Parikh S. Genital and urinary tract infections in diabetes: Impact of pharmacologically‐induced glucosuria. Diabetes Res Clin Pract 103: 373‐381, 2014.
 101.Gembardt F, Bartaun C, Jarzebska N, Mayoux E, Todorov VT, Hohenstein B, Hugo C. The SGLT2 inhibitor empagliflozin ameliorates early features of diabetic nephropathy in BTBR ob/ob type 2 diabetic mice with and without hypertension. Am J Physiol Renal Physiol 307: F317‐F325, 2014.
 102.Gerich JE. Role of the kidney in normal glucose homeostasis and in the hyperglycaemia of diabetes mellitus: Therapeutic implications. Diabet Med 27: 136‐142, 2010.
 103.Gerich JE, Meyer C, Woerle HJ, Stumvoll M. Renal gluconeogenesis: Its importance in human glucose homeostasis. Diabetes Care 24: 382‐391, 2001.
 104.Gersch MS, Mu W, Cirillo P, Reungjui S, Zhang L, Roncal C, Sautin YY, Johnson RJ, Nakagawa T. Fructose, but not dextrose, accelerates the progression of chronic kidney disease. Am J Physiol Renal Physiol 293: F1256‐F1261, 2007.
 105.Gerstein HC, Miller ME, Byington RP, Goff DC Jr, Bigger JT, Buse JB, Cushman WC, Genuth S, Ismail‐Beigi F, Grimm RH Jr, Probstfield JL, Simons‐Morton DG, Friedewald WT. Effects of intensive glucose lowering in type 2 diabetes. N Engl J Med 358: 2545‐2559, 2008.
 106.Ghazi S, Bourgeois S, Gomariz A, Bugarski M, Haenni D, Martins JR, Nombela‐Arrieta C, Unwin RJ, Wagner CA, Hall AM, Craigie E. Multiparametric imaging reveals that mitochondria‐rich intercalated cells in the kidney collecting duct have a very high glycolytic capacity. FASEB J 34: 8510‐8525, 2020.
 107.Ghezzi C, Gorraitz E, Hirayama BA, Loo DD, Grempler R, Mayoux E, Wright EM. Fingerprints of hSGLT5 sugar and cation selectivity. Am J Physiol Cell Physiol 306: C864‐C870, 2014.
 108.Ghezzi C, Hirayama BA, Gorraitz E, Loo DD, Liang Y, Wright EM. SGLT2 inhibitors act from the extracellular surface of the cell membrane. Physiol Rep 2: pii: e12058, 2014.
 109.Ghezzi C, Wright EM. Regulation of the human Na+ dependent glucose cotransporter hSGLT2. Am J Physiol Cell Physiol 303: C348‐C354, 2012.
 110.Gilbert RE, Thorpe KE. Acute kidney injury with sodium‐glucose co‐transporter‐2 inhibitors: A meta‐analysis of cardiovascular outcome trials. Diabetes Obes Metab 21: 1996‐2000, 2019.
 111.Glaser J, Lemery J, Rajagopalan B, Diaz HF, Garcia‐Trabanino R, Taduri G, Madero M, Amarasinghe M, Abraham G, Anutrakulchai S, Jha V, Stenvinkel P, Roncal‐Jimenez C, Lanaspa MA, Correa‐Rotter R, Sheikh‐Hamad D, Burdmann EA, Andres‐Hernando A, Milagres T, Weiss I, Kanbay M, Wesseling C, Sanchez‐Lozada LG, Johnson RJ. Climate change and the emergent epidemic of CKD from heat stress in rural communities: The case for heat stress nephropathy. Clin J Am Soc Nephrol 11: 1472‐1483, 2016.
 112.Glushakova O, Kosugi T, Roncal C, Mu W, Heinig M, Cirillo P, Sanchez‐Lozada LG, Johnson RJ, Nakagawa T. Fructose induces the inflammatory molecule ICAM‐1 in endothelial cells. J Am Soc Nephrol 19: 1712‐1720, 2008.
 113.Goestemeyer AK, Marks J, Srai SK, Debnam ES, Unwin RJ. GLUT2 protein at the rat proximal tubule brush border membrane correlates with protein kinase C (PKC)‐betal and plasma glucose concentration. Diabetologia 50: 2209‐2217, 2007.
 114.Goncalves MD, Lu C, Tutnauer J, Hartman TE, Hwang SK, Murphy CJ, Pauli C, Morris R, Taylor S, Bosch K, Yang S, Wang Y, Van Riper J, Lekaye HC, Roper J, Kim Y, Chen Q, Gross SS, Rhee KY, Cantley LC, Yun J. High‐fructose corn syrup enhances intestinal tumor growth in mice. Science 363: 1345‐1349, 2019.
 115.Gonzalez‐Vicente A, Cabral PD, Hong NJ, Asirwatham J, Saez F, Garvin JL. Fructose reabsorption by rat proximal tubules: Role of Na(+)‐linked cotransporters and the effect of dietary fructose. Am J Physiol Renal Physiol 316: F473‐F480, 2019.
 116.Gonzalez‐Vicente A, Cabral PD, Hong NJ, Asirwatham J, Yang N, Berthiaume JM, Dominici FP, Garvin JL. Dietary fructose enhances the ability of low concentrations of angiotensin II to stimulate proximal tubule Na(+) reabsorption. Nutrients 9: 885, 2017.
 117.Gonzalez‐Vicente A, Garvin JL, Hopfer U. Transcriptome signature for dietary fructose‐specific changes in rat renal cortex: A quantitative approach to physiological relevance. PLoS One 13: e0201293, 2018.
 118.Goodwin RF. Division of the common mammals into two groups according to the concentration of fructose in the blood of the foetus. J Physiol 132: 146‐156, 1956.
 119.Goran MI, Martin AA, Alderete TL, Fujiwara H, Fields DA. Fructose in breast milk is positively associated with infant body composition at 6 months of age. Nutrients 9, 146, 2017.
 120.Gorboulev V, Schurmann A, Vallon V, Kipp H, Jaschke A, Klessen D, Friedrich A, Scherneck S, Rieg T, Cunard R, Veyhl‐Wichmann M, Srinivasan A, Balen D, Breljak D, Rexhepaj R, Parker HE, Gribble FM, Reimann F, Lang F, Wiese S, Sabolic I, Sendtner M, Koepsell H. Na(+)‐D‐glucose cotransporter SGLT1 is pivotal for intestinal glucose absorption and glucose‐dependent incretin secretion. Diabetes 61: 187‐196, 2012.
 121.Gordish KL, Kassem KM, Ortiz PA, Beierwaltes WH. Moderate (20%) fructose‐enriched diet stimulates salt‐sensitive hypertension with increased salt retention and decreased renal nitric oxide. Physiol Rep 5: e13162, 2017.
 122.Goyal SN, Reddy NM, Patil KR, Nakhate KT, Ojha S, Patil CR, Agrawal YO. Challenges and issues with streptozotocin‐induced diabetes ‐ A clinically relevant animal model to understand the diabetes pathogenesis and evaluate therapeutics. Chem Biol Interact 244: 49‐63, 2016.
 123.Grempler R, Augustin R, Froehner S, Hildebrandt T, Simon E, Mark M, Eickelmann P. Functional characterisation of human SGLT‐5 as a novel kidney‐specific sodium‐dependent sugar transporter. FEBS Lett 586: 248‐253, 2012.
 124.Guder WG, Ross BD. Enzyme distribution along the nephron. Kidney Int 26: 101‐111, 1984.
 125.Guder WG, Schmidt U. The localization of gluconeogenesis in rat nephron. Determination of phosphoenolpyruvate carboxykinase in microdissected tubules. Hoppe Seylers Z Physiol Chem 355: 273‐278, 1974.
 126.Guyton AC. Blood pressure control–special role of the kidneys and body fluids. Science 252: 1813‐1816, 1991.
 127.Gyimesi G, Pujol‐Gimenez J, Kanai Y, Hediger MA. Sodium‐coupled glucose transport, the SLC5 family, and therapeutically relevant inhibitors: From molecular discovery to clinical application. Pflugers Arch 472: 1177‐1206, 2020.
 128.Hallow KM, Gebremichael Y, Helmlinger G, Vallon V. Primary proximal tubule hyperreabsorption and impaired tubular transport counterregulation determine glomerular hyperfiltration in diabetes: A modeling analysis. Am J Physiol Renal Physiol 312: F819‐F835, 2017.
 129.Hallow KM, Helmlinger G, Greasley PJ, McMurray JJV, Boulton DW. Why do SGLT2 inhibitors reduce heart failure hospitalization? A differential volume regulation hypothesis. Diabetes Obes Metab 20: 479‐487, 2018.
 130.Han HJ, Lee YJ, Park SH, Lee JH, Taub M. High glucose‐induced oxidative stress inhibits Na+/glucose cotransporter activity in renal proximal tubule cells. Am J Physiol Renal Physiol 288: F988‐F996, 2005.
 131.Hannou SA, Haslam DE, McKeown NM, Herman MA. Fructose metabolism and metabolic disease. J Clin Invest 128: 545‐555, 2018.
 132.Hansell P, Welch WJ, Blantz RC, Palm F. Determinants of kidney oxygen consumption and their relationship to tissue oxygen tension in diabetes and hypertension. Clin Exp Pharmacol Physiol 40: 123‐137, 2013.
 133.Hansen HH, Jelsing J, Hansen CF, Hansen G, Vrang N, Mark M, Klein T, Mayoux E. The sodium glucose cotransporter type 2 inhibitor empagliflozin preserves beta‐cell mass and restores glucose homeostasis in the male zucker diabetic fatty rat. J Pharmacol Exp Ther 350: 657‐664, 2014.
 134.Hayward BE, Bonthron DT. Structure and alternative splicing of the ketohexokinase gene. Eur J Biochem 257: 85‐91, 1998.
 135.Hediger MA, Coady MJ, Ikeda TS, Wright EM. Expression cloning and cDNA sequencing of the Na+/glucose co‐transporter. Nature 330: 379‐381, 1987.
 136.Heerspink HJ, Desai M, Jardine M, Balis D, Meininger G, Perkovic V. Canagliflozin slows progression of renal function decline independently of glycemic effects. J Am Soc Nephrol 28: 368‐375, 2018.
 137.Heerspink HJL, Stefansson BV, Correa‐Rotter R, Chertow GM, Greene T, Hou FF, Mann JFE, McMurray JJV, Lindberg M, Rossing P, Sjostrom CD, Toto RD, Langkilde AM, Wheeler DC, Committees D‐CT, and Investigators. Dapagliflozin in patients with chronic kidney disease. N Engl J Med 383: 1436‐1446, 2020.
 138.Heise T, Seewaldt‐Becker E, Macha S, Hantel S, Pinnetti S, Seman L, Woerle HJ. Safety, tolerability, pharmacokinetics and pharmacodynamics following 4 weeks' treatment with empagliflozin once daily in patients with type 2 diabetes. Diabetes Obes Metab 15: 613‐621, 2013.
 139.Hers HG, Van Schaftingen E. Fructose 2,6‐bisphosphate 2 years after its discovery. Biochem J 206: 1‐12, 1982.
 140.Heyman SN, Rosen S, Brezis M. The renal medulla: Life at the edge of anoxia. Blood Purif 15: 232‐242, 1997.
 141.Higgins DF, Kimura K, Bernhardt WM, Shrimanker N, Akai Y, Hohenstein B, Saito Y, Johnson RS, Kretzler M, Cohen CD, Eckardt KU, Iwano M, Haase VH. Hypoxia promotes fibrogenesis in vivo via HIF‐1 stimulation of epithelial‐to‐mesenchymal transition. J Clin Invest 117: 3810‐3820, 2007.
 142.Hinden L, Udi S, Drori A, Gammal A, Nemirovski A, Hadar R, Baraghithy S, Permyakova A, Geron M, Cohen M, Tsytkin‐Kirschenzweig S, Riahi Y, Leibowitz G, Nahmias Y, Priel A, Tam J. Modulation of renal GLUT2 by the cannabinoid‐1 receptor: Implications for the treatment of diabetic nephropathy. J Am Soc Nephrol 29: 434‐448, 2018.
 143.Hitchcock MW. Fructose in the sheep foetus. J Physiol 108: 117‐126, 1949.
 144.Holck P, Rasch R. Structure and segmental localization of glycogen in the diabetic rat kidney. Diabetes 42: 891‐900, 1993.
 145.Holman GD. Structure, function and regulation of mammalian glucose transporters of the SLC2 family. Pflugers Arch 472: 1155‐1175, 2020.
 146.Holstein‐Rathlou NH, Marsh DJ. Oscillations of tubular pressure, flow, and distal chloride concentration in rats. Am J Physiol 256: F1007‐F1014, 1989.
 147.Horiba N, Masuda S, Ohnishi C, Takeuchi D, Okuda M, Inui K. Na(+)‐dependent fructose transport via rNaGLT1 in rat kidney. FEBS Lett 546: 276‐280, 2003.
 148.Hotta N, Kawamori R, Fukuda M, Shigeta Y. Long‐term clinical effects of epalrestat, an aldose reductase inhibitor, on progression of diabetic neuropathy and other microvascular complications: Multivariate epidemiological analysis based on patient background factors and severity of diabetic neuropathy. Diabet Med 29: 1529‐1533, 2012.
 149.Huggett AS. Carbohydrate metabolism in the placenta and foetus. Br Med Bull 17: 122‐126, 1961.
 150.Hummel CS, Lu C, Loo DD, Hirayama BA, Voss AA, Wright EM. Glucose transport by human renal Na+/D‐glucose cotransporters SGLT1 and SGLT2. Am J Physiol Cell Physiol 300: C14‐C21, 2011.
 151.Iles RA, Cohen RD, Rist AH, Baron PG. The mechanism of inhibition by acidosis of gluconeogenesis from lactate in rat liver. Biochem J 164: 185‐191, 1977.
 152.Inzucchi SE, Zinman B, Fitchett D, Wanner C, Ferrannini E, Schumacher M, Schmoor C, Ohneberg K, Johansen OE, George JT, Hantel S, Bluhmki E, Lachin JM. How does empagliflozin reduce cardiovascular mortality? Insights from a mediation analysis of the EMPA‐REG OUTCOME trial. Diabetes Care 41: 356‐363, 2018.
 153.Jang C, Hui S, Lu W, Cowan AJ, Morscher RJ, Lee G, Liu W, Tesz GJ, Birnbaum MJ, Rabinowitz JD. The small intestine converts dietary fructose into glucose and organic acids. Cell Metab 27: 351‐361 e353, 2018.
 154.Jauniaux E, Hempstock J, Teng C, Battaglia FC, Burton GJ. Polyol concentrations in the fluid compartments of the human conceptus during the first trimester of pregnancy: Maintenance of redox potential in a low oxygen environment. J Clin Endocrinol Metab 90: 1171‐1175, 2005.
 155.Jensen PK, Kristensen KS, Rasch R, Persson AEG. Decreased sensitivity of the tubuloglomerular feedback mechanism in experimental diabetic rats. In: Persson AEG, Boberg U, editors. The Juxtaglomerular Apparatus. Amsterdam New York: Elsevier, 1988, p. 333‐338.
 156.Johnson RJ, Andrews P, Benner SA, Oliver W. Theodore E. Woodward award. The evolution of obesity: Insights from the mid‐Miocene. Trans Am Clin Climatol Assoc 121: 295‐305; discussion 305‐298, 2010.
 157.Johnson RJ, Bakris GL, Borghi C, Chonchol MB, Feldman D, Lanaspa MA, Merriman TR, Moe OW, Mount DB, Sanchez Lozada LG, Stahl E, Weiner DE, Chertow GM. Hyperuricemia, acute and chronic kidney disease, hypertension, and cardiovascular disease: Report of a scientific workshop organized by the National Kidney Foundation. Am J Kidney Dis, 2018.
 158.Johnson RJ, Nakagawa T, Sanchez‐Lozada LG, Shafiu M, Sundaram S, Le M, Ishimoto T, Sautin YY, Lanaspa MA. Sugar, uric acid, and the etiology of diabetes and obesity. Diabetes 62: 3307‐3315, 2013.
 159.Johnson RJ, Sanchez‐Lozada LG, Nakagawa T. The effect of fructose on renal biology and disease. J Am Soc Nephrol 21: 2036‐2039, 2010.
 160.Johnson RJ, Segal MS, Sautin Y, Nakagawa T, Feig DI, Kang DH, Gersch MS, Benner S, Sanchez‐Lozada LG. Potential role of sugar (fructose) in the epidemic of hypertension, obesity and the metabolic syndrome, diabetes, kidney disease, and cardiovascular disease. Am J Clin Nutr 86: 899‐906, 2007.
 161.Johnson RJ, Stenvinkel P, Andrews P, Sanchez‐Lozada LG, Nakagawa T, Gaucher E, Andres‐Hernando A, Rodriguez‐Iturbe B, Jimenez CR, Garcia G, Kang DH, Tolan DR, Lanaspa MA. Fructose metabolism as a common evolutionary pathway of survival associated with climate change, food shortage and droughts. J Intern Med 287: 252‐262, 2020.
 162.Johnston PA, Rennke H, Levinsky NG. Recovery of proximal tubular function from ischemic injury. Am J Physiol 246: F159‐F166, 1984.
 163.Junk WJ. Temporary fat storage, an adaptation of some fish species to the waterlevel fluctuations and related environmental changes of the Amazon river. Amazoniana 9: 315‐351, 1985.
 164.Jurczak MJ, Lee HY, Birkenfeld AL, Jornayvaz FR, Frederick DW, Pongratz RL, Zhao X, Moeckel GW, Samuel VT, Whaley JM, Shulman GI, Kibbey RG. SGLT2 deletion improves glucose homeostasis and preserves pancreatic beta‐cell function. Diabetes 60: 890‐898, 2011.
 165.Kamran M, Peterson RG, Dominguez JH. Overexpression of GLUT2 gene in renal proximal tubules of diabetic Zucker rats. J Am Soc Nephrol 8: 943‐948, 1997.
 166.Kanai Y, Lee WS, You G, Brown D, Hediger MA. The human kidney low affinity Na+/glucose cotransporter SGLT2. Delineation of the major renal reabsorptive mechanism for D‐glucose. J Clin Invest 93: 397‐404, 1994.
 167.Kang DH, Nakagawa T, Feng L, Watanabe S, Han L, Mazzali M, Truong L, Harris R, Johnson RJ. A role for uric acid in the progression of renal disease. J Am Soc Nephrol 13: 2888‐2897, 2002.
 168.Kang HM, Ahn SH, Choi P, Ko YA, Han SH, Chinga F, Park AS, Tao J, Sharma K, Pullman J, Bottinger EP, Goldberg IJ, Susztak K. Defective fatty acid oxidation in renal tubular epithelial cells has a key role in kidney fibrosis development. Nat Med 21: 37‐46, 2015.
 169.Kang J, Dai XS, Yu TB, Wen B, Yang ZW. Glycogen accumulation in renal tubules, a key morphological change in the diabetic rat kidney. Acta Diabetol 42: 110‐116, 2005.
 170.Karalius VP, Shoham DA. Dietary sugar and artificial sweetener intake and chronic kidney disease: A review. Adv Chronic Kidney Dis 20: 157‐164, 2013.
 171.Kato ET, Silverman MG, Mosenzon O, Zelniker TA, Cahn A, Furtado RHM, Kuder J, Murphy SA, Bhatt DL, Leiter LA, McGuire DK, Wilding JPH, Bonaca MP, Ruff CT, Desai AS, Goto S, Johansson PA, Gause‐Nilsson I, Johanson P, Langkilde AM, Raz I, Sabatine MS, Wiviott SD. Effect of dapagliflozin on heart failure and mortality in type 2 diabetes mellitus. Circulation 139: 2528‐2536, 2019.
 172.Kawasaki T, Akanuma H, Yamanouchi T. Increased fructose concentrations in blood and urine in patients with diabetes. Diabetes Care 25: 353‐357, 2002.
 173.Kayano T, Burant CF, Fukumoto H, Gould GW, Fan YS, Eddy RL, Byers MG, Shows TB, Seino S, Bell GI. Human facilitative glucose transporters. Isolation, functional characterization, and gene localization of cDNAs encoding an isoform (GLUT5) expressed in small intestine, kidney, muscle, and adipose tissue and an unusual glucose transporter pseudogene‐like sequence (GLUT6). J Biol Chem 265: 13276‐13282, 1990.
 174.Kellett GL, Brot‐Laroche E, Mace OJ, Leturque A. Sugar absorption in the intestine: The role of GLUT2. Annu Rev Nutr 28: 35‐54, 2008.
 175.Khosla UM, Zharikov S, Finch JL, Nakagawa T, Roncal C, Mu W, Krotova K, Block ER, Prabhakar S, Johnson RJ. Hyperuricemia induces endothelial dysfunction. Kidney Int 67: 1739‐1742, 2005.
 176.Khunti K, Davies M, Majeed A, Thorsted BL, Wolden ML, Paul SK. Hypoglycemia and risk of cardiovascular disease and all‐cause mortality in insulin‐treated people with type 1 and type 2 diabetes: A cohort study. Diabetes Care 38: 316‐322, 2015.
 177.Kida K, Nakajo S, Kamiya F, Toyama Y, Nishio T, Nakagawa H. Renal net glucose release in vivo and its contribution to blood glucose in rats. J Clin Invest 62: 721‐726, 1978.
 178.Kidokoro K, Cherney DZI, Bozovic A, Nagasu H, Satoh M, Kanda E, Sasaki T, Kashihara N. Evaluation of glomerular hemodynamic function by empagliflozin in diabetic mice using in vivo imaging. Circulation 140: 303‐315, 2019.
 179.Kim J, Song G, Wu G, Bazer FW. Functional roles of fructose. Proc Natl Acad Sci U S A 109: E1619‐E1628, 2012.
 180.Kimura T, Takahashi M, Yan K, Sakurai H. Expression of SLC2A9 isoforms in the kidney and their localization in polarized epithelial cells. PLoS One 9: e84996, 2014.
 181.Kitada K, Nakano D, Ohsaki H, Hitomi H, Minamino T, Yatabe J, Felder RA, Mori H, Masaki T, Kobori H, Nishiyama A. Hyperglycemia causes cellular senescence via a SGLT2‐ and p21‐dependent pathway in proximal tubules in the early stage of diabetic nephropathy. J Diabetes Complications 28: 604‐611, 2014.
 182.Klein KL, Wang MS, Torikai S, Davidson WD, Kurokawa K. Substrate oxidation by isolated single nephron segments of the rat. Kidney Int 20: 29‐35, 1981.
 183.Kocinsky HS, Dynia DW, Wang T, Aronson PS. NHE3 phosphorylation at serines 552 and 605 does not directly affect NHE3 activity. Am J Physiol Renal Physiol 293: F212‐F218, 2007.
 184.Kocinsky HS, Girardi AC, Biemesderfer D, Nguyen T, Mentone S, Orlowski J, Aronson PS. Use of phospho‐specific antibodies to determine the phosphorylation of endogenous Na+/H+ exchanger NHE3 at PKA consensus sites. Am J Physiol Renal Physiol 289: F249‐F258, 2005.
 185.Koepsell H. Glucose transporters in the small intestine in health and disease. Pflugers Arch 472: 1207‐1248, 2020.
 186.Koepsell H, Vallon V. A special issue on glucose transporters in health and disease. Pflugers Arch 472: 1107‐1109, 2020.
 187.Kogot‐Levin A, Hinden L, Riahi Y, Israeli T, Tirosh B, Cerasi E, Mizrachi EB, Tam J, Mosenzon O, Leibowitz G. Proximal tubule mTORC1 is a central player in the pathophysiology of diabetic nephropathy and its correction by SGLT2 inhibitors. Cell Rep 32: 107954, 2020.
 188.Kohan DE, Fioretto P, Johnsson K, Parikh S, Ptaszynska A, Ying L. The effect of dapagliflozin on renal function in patients with type 2 diabetes. J Nephrol 29: 391‐400, 2016.
 189.Komers R, Lindsley JN, Oyama TT, Allison KM, Anderson S. Role of neuronal nitric oxide synthase (NOS1) in the pathogenesis of renal hemodynamic changes in diabetes. Am J Physiol Renal Physiol 279: F573‐F583, 2000.
 190.Komoroski B, Vachharajani N, Boulton D, Kornhauser D, Geraldes M, Li L, Pfister M. Dapagliflozin, a novel SGLT2 inhibitor, induces dose‐dependent glucosuria in healthy subjects. Clin Pharmacol Ther 85: 520‐526, 2009.
 191.Kowalski GM, Bruce CR. The regulation of glucose metabolism: Implications and considerations for the assessment of glucose homeostasis in rodents. Am J Physiol Endocrinol Metab 307: E859‐E871, 2014.
 192.Kranhold JF, Loh D, Morris RC Jr. Renal fructose‐metabolizing enzymes: Significance in hereditary fructose intolerance. Science 165: 402‐403, 1969.
 193.Krebs HA, Lund P. Formation of glucose from hexoses, pentoses, polyols and related substances in kidney cortex. Biochem J 98: 210‐214, 1966.
 194.Kuhre RE, Ghiasi SM, Adriaenssens AE, Wewer Albrechtsen NJ, Andersen DB, Aivazidis A, Chen L, Mandrup‐Poulsen T, Orskov C, Gribble FM, Reimann F, Wierup N, Tyrberg B, Holst JJ. No direct effect of SGLT2 activity on glucagon secretion. Diabetologia 62: 1011‐1023, 2019.
 195.Lambers Heerspink HJ, de Zeeuw D, Wie L, Leslie B, List J. Dapagliflozin a glucose‐regulating drug with diuretic properties in subjects with type 2 diabetes. Diabetes Obes Metab 15: 853‐862, 2013.
 196.Lan R, Geng H, Singha PK, Saikumar P, Bottinger EP, Weinberg JM, Venkatachalam MA. Mitochondrial pathology and glycolytic shift during proximal tubule atrophy after ischemic AKI. J Am Soc Nephrol 27: 3356‐3367, 2016.
 197.Lanaspa MA, Ishimoto T, Cicerchi C, Tamura Y, Roncal‐Jimenez CA, Chen W, Tanabe K, Andres‐Hernando A, Orlicky DJ, Finol E, Inaba S, Li N, Rivard CJ, Kosugi T, Sanchez‐Lozada LG, Petrash JM, Sautin YY, Ejaz AA, Kitagawa W, Garcia GE, Bonthron DT, Asipu A, Diggle CP, Rodriguez‐Iturbe B, Nakagawa T, Johnson RJ. Endogenous fructose production and fructokinase activation mediate renal injury in diabetic nephropathy. J Am Soc Nephrol 25: 2526‐2538, 2014.
 198.Lanaspa MA, Kuwabara M, Andres‐Hernando A, Li N, Cicerchi C, Jensen T, Orlicky DJ, Roncal‐Jimenez CA, Ishimoto T, Nakagawa T, Rodriguez‐Iturbe B, MacLean PS, Johnson RJ. High salt intake causes leptin resistance and obesity in mice by stimulating endogenous fructose production and metabolism. Proc Natl Acad Sci U S A 115: 3138‐3143, 2018.
 199.Lanaspa MA, Sanchez‐Lozada LG, Choi YJ, Cicerchi C, Kanbay M, Roncal‐Jimenez CA, Ishimoto T, Li N, Marek G, Duranay M, Schreiner G, Rodriguez‐Iturbe B, Nakagawa T, Kang DH, Sautin YY, Johnson RJ. Uric acid induces hepatic steatosis by generation of mitochondrial oxidative stress: Potential role in fructose‐dependent and ‐independent fatty liver. J Biol Chem 287: 40732‐40744, 2012.
 200.Lanaspa MA, Sanchez‐Lozada LG, Cicerchi C, Li N, Roncal‐Jimenez CA, Ishimoto T, Le M, Garcia GE, Thomas JB, Rivard CJ, Andres‐Hernando A, Hunter B, Schreiner G, Rodriguez‐Iturbe B, Sautin YY, Johnson RJ. Uric acid stimulates fructokinase and accelerates fructose metabolism in the development of fatty liver. PLoS One 7: e47948, 2012.
 201.Lang F, Gorlach A, Vallon V. Targeting SGK1 in diabetes. Expert Opin Ther Targets 13: 1303‐1311, 2009.
 202.Layton AT, Vallon V. SGLT2 inhibition in a kidney with reduced nephron number: Modeling and analysis of solute transport and metabolism. Am J Physiol Renal Physiol 314: F969‐F984, 2018.
 203.Layton AT, Vallon V, Edwards A. Modeling oxygen consumption in the proximal tubule: Effects of NHE and SGLT2 inhibition. Am J Physiol Renal Physiol 308: F1343‐F1357, 2015.
 204.Layton AT, Vallon V, Edwards A. Predicted consequences of diabetes and SGLT inhibition on transport and oxygen consumption along a rat nephron. Am J Physiol Renal Physiol 310: F1269‐F1283, 2016.
 205.Lee JW, Chou CL, Knepper MA. Deep sequencing in microdissected renal tubules identifies nephron segment‐specific transcriptomes. J Am Soc Nephrol 26: 2669‐2677, 2015.
 206.Lee WS, Kanai Y, Wells RG, Hediger MA. The high affinity Na+/glucose cotransporter. Re‐evaluation of function and distribution of expression. J Biol Chem 269: 12032‐12039, 1994.
 207.Lee YH, Kim SH, Kang JM, Heo JH, Kim DJ, Park SH, Sung M, Kim J, Oh J, Yang DH, Lee SH, Lee SY. Empagliflozin attenuates diabetic tubulopathy by improving mitochondrial fragmentation and autophagy. Am J Physiol Renal Physiol 317: F767‐F780, 2019.
 208.Legouis D, Ricksten SE, Faivre A, Verissimo T, Gariani K, Verney C, Galichon P, Berchtold L, Feraille E, Fernandez M, Placier S, Koppitch K, Hertig A, Martin PY, Naesens M, Pugin J, McMahon AP, Cippa PE, de Seigneux S. Altered proximal tubular cell glucose metabolism during acute kidney injury is associated with mortality. Nat Metab 2: 732‐743, 2020.
 209.Li J, Liu H, Takagi S, Nitta K, Kitada M, Srivastava SP, Takagaki Y, Kanasaki K, Koya D. Renal protective effects of empagliflozin via inhibition of EMT and aberrant glycolysis in proximal tubules. JCI Insight 5: e129034, 2020.
 210.Li J, Neal B, Perkovic V, de Zeeuw D, Neuen BL, Arnott C, Simpson R, Oh R, Mahaffey KW, Heerspink HJL. Mediators of the effects of canagliflozin on kidney protection in patients with type 2 diabetes. Kidney Int 98: 769‐777, 2020.
 211.Li J, Woodward M, Perkovic V, Figtree GA, Heerspink HJL, Mahaffey KW, de Zeeuw D, Vercruysse F, Shaw W, Matthews DR, Neal B. Mediators of the effects of canagliflozin on heart failure in patients with type 2 diabetes. JACC Heart Fail 8: 57‐66, 2020.
 212.Li Z, Agrawal V, Ramratnam M, Sharma RK, D'Auria S, Sincoular A, Jakubiak M, Music ML, Kutschke WJ, Huang XN, Gifford L, Ahmad F. Cardiac sodium‐glucose co‐transporter 1 (SGLT1) is a novel mediator of ischemia/reperfusion injury. Cardiovasc Res 115: 1646‐1658, 2019.
 213.Limbutara K, Chou CL, Knepper MA. Quantitative proteomics of all 14 renal tubule segments in rat. J Am Soc Nephrol, 2020. DOI: 10.1681/ASN.2020010071.
 214.Lin J, Curhan GC. Associations of sugar and artificially sweetened soda with albuminuria and kidney function decline in women. Clin J Am Soc Nephrol 6: 160‐166, 2011.
 215.Linden KC, DeHaan CL, Zhang Y, Glowacka S, Cox AJ, Kelly DJ, Rogers S. Renal expression and localization of the facilitative glucose transporters GLUT1 and GLUT12 in animal models of hypertension and diabetic nephropathy. Am J Physiol Renal Physiol 290: F205‐F213, 2006.
 216.Litvinov D, Selvarajan K, Garelnabi M, Brophy L, Parthasarathy S. Anti‐atherosclerotic actions of azelaic acid, an end product of linoleic acid peroxidation, in mice. Atherosclerosis 209: 449‐454, 2010.
 217.Liu H, Takagaki Y, Kumagai A, Kanasaki K, Koya D. The PKM2 activator TEPP‐46 suppresses kidney fibrosis via inhibition of the EMT program and aberrant glycolysis associated with suppression of HIF‐1alpha accumulation. J Diabetes Investig 12: 697‐709, 2021.
 218.Liu R, Carretero OA, Ren Y, Garvin JL. Increased intracellular pH at the macula densa activates nNOS during tubuloglomerular feedback. Kidney Int 67: 1837‐1843, 2005.
 219.Locatelli M, Zoja C, Zanchi C, Corna D, Villa S, Bolognini S, Novelli R, Perico L, Remuzzi G, Benigni A, Cassis P. Manipulating Sirtuin 3 pathway ameliorates renal damage in experimental diabetes. Sci Rep 10: 8418, 2020.
 220.Loeschke K, Baumann K, Renschler H, Ullrich KJ. Differentiation of the active and passive components of D‐glucose transport in the proximal tubule of rat kidney. Pflugers Arch 305: 118‐138, 1969.
 221.Lu YH, Chang YP, Li T, Han F, Li CJ, Li XY, Xue M, Cheng Y, Meng ZY, Han Z, Sun B, Chen LM. Empagliflozin attenuates hyperuricemia by upregulation of ABCG2 via AMPK/AKT/CREB signaling pathway in type 2 diabetic mice. Int J Biol Sci 16: 529‐542, 2020.
 222.Ly JP, Onay T, Sison K, Sivaskandarajah G, Sabbisetti V, Li L, Bonventre JV, Flenniken A, Paragas N, Barasch JM, Adamson SL, Osborne L, Rossant J, Schnermann J, Quaggin SE. The sweet pee model for Sglt2 mutation. J Am Soc Nephrol 22: 113‐123, 2011.
 223.Lynch MR, Tran MT, Parikh SM. PGC1alpha in the kidney. Am J Physiol Renal Physiol 314: F1‐F8, 2018.
 224.Lytvyn Y, Bjornstad P, Katz A, Singh SK, Godoy LC, Chung LT, Vinovskis CL, Pyle L, Roussel R, Perkins BA, Cherney D. SGLT2 inhibition increases serum copeptin in young adults with type 1 diabetes. Diabetes Metab, 2019.
 225.Lytvyn Y, Skrtic M, Yang GK, Yip PM, Perkins BA, Cherney DZ. Glycosuria‐mediated urinary uric acid excretion in patients with uncomplicated type 1 diabetes mellitus. Am J Physiol Renal Physiol 308: F77‐F83, 2015.
 226.Macdonald FR, Peel JE, Jones HB, Mayers RM, Westgate L, Whaley JM, Poucher SM. The novel sodium glucose transporter 2 inhibitor dapagliflozin sustains pancreatic function and preserves islet morphology in obese, diabetic rats. Diabetes Obes Metab 12: 1004‐1012, 2010.
 227.Macdonald I, Keyser A, Pacy D. Some effects, in man, of varying the load of glucose, sucrose, fructose, or sorbitol on various metabolites in blood. Am J Clin Nutr 31: 1305‐1311, 1978.
 228.Madunic IV, Breljak D, Karaica D, Koepsell H, Sabolic I. Expression profiling and immunolocalization of Na(+)‐D‐glucose‐cotransporter 1 in mice employing knockout mice as specificity control indicate novel locations and differences between mice and rats. Pflugers Arch 469: 1545‐1565, 2017.
 229.Maenpaa PH, Raivio KO, Kekomaki MP. Liver adenine nucleotides: Fructose‐induced depletion and its effect on protein synthesis. Science 161: 1253‐1254, 1968.
 230.Magee GM, Bilous RW, Cardwell CR, Hunter SJ, Kee F, Fogarty DG. Is hyperfiltration associated with the future risk of developing diabetic nephropathy? A meta‐analysis. Diabetologia 52: 691‐697, 2009.
 231.Malek M, Nematbakhsh M. Renal ischemia/reperfusion injury; from pathophysiology to treatment. J Renal Inj Prev 4: 20‐27, 2015.
 232.Marks J, Carvou NJ, Debnam ES, Srai SK, Unwin RJ. Diabetes increases facilitative glucose uptake and GLUT2 expression at the rat proximal tubule brush border membrane. J Physiol 553: 137‐145, 2003.
 233.Martin MG, Turk E, Lostao MP, Kerner C, Wright EM. Defects in Na+/glucose cotransporter (SGLT1) trafficking and function cause glucose‐galactose malabsorption. Nat Genet 12: 216‐220, 1996.
 234.Masuda T, Muto S, Fukuda K, Watanabe M, Ohara K, Koepsell H, Vallon V, Nagata D. Osmotic diuresis by SGLT2 inhibition stimulates vasopressin‐induced water reabsorption to maintain body fluid volume. Physiol Rep 8: e14360, 2020.
 235.Masuda T, Watanabe Y, Fukuda K, Watanabe M, Onishi A, Ohara K, Imai T, Koepsell H, Muto S, Vallon V, Nagata D. Unmasking a sustained negative effect of SGLT2 inhibition on body fluid volume in the rat. Am J Physiol Renal Physiol 315: F653‐F664, 2018.
 236.Mate A, de la Hermosa MA, Barfull A, Planas JM, Vazquez CM. Characterization of D‐fructose transport by rat kidney brush‐border membrane vesicles: Changes in hypertensive rats. Cell Mol Life Sci 58: 1961‐1967, 2001.
 237.Mathupala SP, Ko YH, Pedersen PL. Hexokinase II: Cancer's double‐edged sword acting as both facilitator and gatekeeper of malignancy when bound to mitochondria. Oncogene 25: 4777‐4786, 2006.
 238.Matsuo H, Chiba T, Nagamori S, Nakayama A, Domoto H, Phetdee K, Wiriyasermkul P, Kikuchi Y, Oda T, Nishiyama J, Nakamura T, Morimoto Y, Kamakura K, Sakurai Y, Nonoyama S, Kanai Y, Shinomiya N. Mutations in glucose transporter 9 gene SLC2A9 cause renal hypouricemia. Am J Hum Genet 83: 744‐751, 2008.
 239.Mavrias DA, Mayer RJ. Metabolism of fructose in the small intestine. 1. The effect of fructose feeding on fructose transport and metabolism in rat small intestine. Biochim Biophys Acta 291: 531‐537, 1973.
 240.McMurray JJV, Solomon SD, Inzucchi SE, Kober L, Kosiborod MN, Martinez FA, Ponikowski P, Sabatine MS, Anand IS, Belohlavek J, Bohm M, Chiang CE, Chopra VK, de Boer RA, Desai AS, Diez M, Drozdz J, Dukat A, Ge J, Howlett JG, Katova T, Kitakaze M, Ljungman CEA, Merkely B, Nicolau JC, O'Meara E, Petrie MC, Vinh PN, Schou M, Tereshchenko S, Verma S, Held C, DeMets DL, Docherty KF, Jhund PS, Bengtsson O, Sjostrand M, Langkilde AM. Dapagliflozin in patients with heart failure and reduced ejection fraction. N Engl J Med 381: 1995‐2008, 2019.
 241.Mellanby K. Metabolic water and desiccation. Nature 150: 21‐21, 1942.
 242.Merovci A, Solis‐Herrera C, Daniele G, Eldor R, Fiorentino TV, Tripathy D, Xiong J, Perez Z, Norton L, Abdul‐Ghani MA, DeFronzo RA. Dapagliflozin improves muscle insulin sensitivity but enhances endogenous glucose production. J Clin Invest 124: 509‐514, 2014.
 243.Meyer C, Stumvoll M, Nadkarni V, Dostou J, Mitrakou A, Gerich J. Abnormal renal and hepatic glucose metabolism in type 2 diabetes mellitus. J Clin Invest 102: 619‐624, 1998.
 244.Michelakis ED, Webster L, Mackey JR. Dichloroacetate (DCA) as a potential metabolic‐targeting therapy for cancer. Br J Cancer 99: 989‐994, 2008.
 245.Mirtschink P, Krishnan J, Grimm F, Sarre A, Horl M, Kayikci M, Fankhauser N, Christinat Y, Cortijo C, Feehan O, Vukolic A, Sossalla S, Stehr SN, Ule J, Zamboni N, Pedrazzini T, Krek W. HIF‐driven SF3B1 induces KHK‐C to enforce fructolysis and heart disease. Nature 522: 444‐449, 2015.
 246.Moen MF, Zhan M, Hsu VD, Walker LD, Einhorn LM, Seliger SL, Fink JC. Frequency of hypoglycemia and its significance in chronic kidney disease. Clin J Am Soc Nephrol 4: 1121‐1127, 2009.
 247.Mogensen CE. Maximum tubular reabsorption capacity for glucose and renal hemodynamics during rapid hypertonic glucose infusion in normal and diabetic subjects. Scand J Clin Lab Invest 28: 101‐109, 1971.
 248.Molitoris BA, Kinne R. Ischemia induces surface membrane dysfunction. Mechanism of altered Na+‐dependent glucose transport. J Clin Invest 80: 647‐654, 1987.
 249.Monami M, Nardini C, Mannucci E. Efficacy and safety of sodium glucose co‐transport‐2 inhibitors in type 2 diabetes: A meta‐analysis of randomized clinical trials. Diabetes Obes Metab 16: 457‐466, 2014.
 250.Mudaliar S, Alloju S, Henry RR. Can a shift in fuel energetics explain the beneficial cardiorenal outcomes in the EMPA‐REG OUTCOME study? A unifying hypothesis. diabetes care 39: 1115‐1122, 2016.
 251.Mueckler M, Thorens B. The SLC2 (GLUT) family of membrane transporters. Mol Aspects Med 34: 121‐138, 2013.
 252.Mulder S, Heerspink HJL, Darshi M, Kim JJ, Laverman GD, Sharma K, Pena MJ. Effects of dapagliflozin on urinary metabolites in people with type 2 diabetes. Diabetes Obes Metab, 2019.
 253.Nakagawa T. Uncoupling of the VEGF‐endothelial nitric oxide axis in diabetic nephropathy: An explanation for the paradoxical effects of VEGF in renal disease. Am J Physiol Renal Physiol 292: F1665‐F1672, 2007.
 254.Nakagawa T, Hu H, Zharikov S, Tuttle KR, Short RA, Glushakova O, Ouyang X, Feig DI, Block ER, Herrera‐Acosta J, Patel JM, Johnson RJ. A causal role for uric acid in fructose‐induced metabolic syndrome. Am J Physiol Renal Physiol 290: F625‐F631, 2006.
 255.Nakagawa T, Johnson RJ, Andres‐Hernando A, Roncal‐Jimenez C, Sanchez‐Lozada LG, Tolan DR, Lanaspa MA. Fructose production and metabolism in the kidney. J Am Soc Nephrol 31: 898‐906, 2020.
 256.Nakagawa T, Lanaspa MA, Johnson RJ. The effects of fruit consumption in patients with hyperuricaemia or gout. Rheumatology (Oxford) 58: 1133‐1141, 2019.
 257.Nakagawa T, Lanaspa MA, Millan IS, Fini M, Rivard CJ, Sanchez‐Lozada LG, Andres‐Hernando A, Tolan DR, Johnson RJ. Fructose contributes to the Warburg effect for cancer growth. Cancer Metab 8: 16, 2020.
 258.Nakagawa T, Tanabe K, Croker BP, Johnson RJ, Grant MB, Kosugi T, Li Q. Endothelial dysfunction as a potential contributor in diabetic nephropathy. Nat Rev Nephrol 7: 36‐44, 2011.
 259.Nakagawa T, Tuttle KR, Short RA, Johnson RJ. Hypothesis: Fructose‐induced hyperuricemia as a causal mechanism for the epidemic of the metabolic syndrome. Nat Clin Pract Nephrol 1: 80‐86, 2005.
 260.Nakayama T, Kosugi T, Gersch M, Connor T, Sanchez‐Lozada LG, Lanaspa MA, Roncal C, Perez‐Pozo SE, Johnson RJ, Nakagawa T. Dietary fructose causes tubulointerstitial injury in the normal rat kidney. Am J Physiol Renal Physiol 298: F712‐F720, 2010.
 261.Nangaku M. Chronic hypoxia and tubulointerstitial injury: A final common pathway to end‐stage renal failure. J Am Soc Nephrol 17: 17‐25, 2006.
 262.Nannipieri M, Lanfranchi A, Santerini D, Catalano C, Van de Werve G, Ferrannini E. Influence of long‐term diabetes on renal glycogen metabolism in the rat. Nephron 87: 50‐57, 2001.
 263.Neal B, Perkovic V, Mahaffey KW, de Zeeuw D, Fulcher G, Erondu N, Shaw W, Law G, Desai M, Matthews DR. Canagliflozin and cardiovascular and renal events in type 2 diabetes. N Engl J Med 377: 644‐657, 2017.
 264.Neill O, Fasching A, Pihl L, Patinha D, Franzen S, Palm F. Acute SGLT inhibition normalizes oxygen tension in the renal cortex but causes hypoxia in the renal medulla in anaesthetized control and diabetic rats. Am J Physiol Renal Physiol 309: F227‐F234, 2015.
 265.Neilson EG. The fructose nation. J Am Soc Nephrol 18: 2619‐2621, 2007.
 266.Nespoux J, Patel R, Hudkins KL, Huang W, Freeman B, Kim Y, Koepsell H, Alpers CE, Vallon V. Gene deletion of the Na‐glucose cotransporter SGLT1 ameliorates kidney recovery in a murine model of acute kidney injury induced by ischemia‐reperfusion. Am J Physiol Renal Physiol 316: F1201‐F1210, 2019.
 267.Nespoux J, Patel R, Zhang H, Huang W, Freeman B, Sanders PW, Kim YC, Vallon V. Gene knockout of the Na(+)‐glucose cotransporter SGLT2 in a murine model of acute kidney injury induced by ischemia‐reperfusion. Am J Physiol Renal Physiol 318: F1100‐F1112, 2020.
 268.Nespoux J, Vallon V. Renal effects of SGLT2 inhibitors: An update. Curr Opin Nephrol Hypertens 29: 190‐198, 2019.
 269.Nespoux J, Vallon V. SGLT2 inhibition and kidney protection. Clin Sci (Lond) 132: 1329‐1339, 2018.
 270.Neuen BL, Young T, Heerspink HJL, Neal B, Perkovic V, Billot L, Mahaffey KW, Charytan DM, Wheeler DC, Arnott C, Bompoint S, Levin A, Jardine MJ. SGLT2 inhibitors for the prevention of kidney failure in patients with type 2 diabetes: A systematic review and meta‐analysis. Lancet Diabetes Endocrinol 7: 845‐854, 2019.
 271.Nguyen S, Choi HK, Lustig RH, Hsu CY. Sugar‐sweetened beverages, serum uric acid, and blood pressure in adolescents. J Pediatr 154: 807‐813, 2009.
 272.Niculescu L, Veiga‐da‐Cunha M, Van Schaftingen E. Investigation on the mechanism by which fructose, hexitols and other compounds regulate the translocation of glucokinase in rat hepatocytes. Biochem J 321 (Pt 1): 239‐246, 1997.
 273.Norton L, Shannon CE, Fourcaudot M, Hu C, Wang N, Ren W, Song J, Abdul‐Ghani M, DeFronzo RA, Ren J, Jia W. Sodium‐glucose co‐transporter (SGLT) and glucose transporter (GLUT) expression in the kidney of type 2 diabetic subjects. Diabetes Obes Metab 19: 1322‐1326, 2017.
 274.Nosadini R, Semplicini A, Fioretto P, Lusiani L, Trevisan R, Donadon V, Zanette G, Nicolosi GL, Dall'Aglio V, Zanuttini D, et al. Sodium‐lithium countertransport and cardiorenal abnormalities in essential hypertension. Hypertension 18: 191‐198, 1991.
 275.Novikov A, Fu Y, Huang W, Freeman B, Patel R, van Ginkel C, Koepsell H, Busslinger M, Onishi A, Nespoux J, Vallon V. SGLT2 inhibition and renal urate excretion: Role of luminal glucose, GLUT9, and URAT1. Am J Physiol Renal Physiol 316: F173‐F185, 2019.
 276.Ohmura C, Watada H, Azuma K, Shimizu T, Kanazawa A, Ikeda F, Yoshihara T, Fujitani Y, Hirose T, Tanaka Y, Kawamori R. Aldose reductase inhibitor, epalrestat, reduces lipid hydroperoxides in type 2 diabetes. Endocr J 56: 149‐156, 2009.
 277.Onishi A, Fu Y, Darshi M, Crespo‐Masip M, Huang W, Song P, Patel R, Kim YC, Nespoux J, Freeman B, Soleimani M, Thomson SC, Sharma K, Vallon V. Effect of renal tubule‐specific knockdown of the Na(+)/H(+) exchanger NHE3 in Akita diabetic mice. Am J Physiol Renal Physiol 317: F419‐F434, 2019.
 278.Onishi A, Fu Y, Patel R, Darshi M, Crespo‐Masip M, Huang W, Song P, Freeman B, Kim YC, Soleimani M, Sharma K, Thomson SC, Vallon V. A role for tubular Na(+)/H(+) exchanger NHE3 in the natriuretic effect of the SGLT2 inhibitor empagliflozin. Am J Physiol Renal Physiol 319: F712‐F728, 2020.
 279.Ono S, Kume S, Yasuda‐Yamahara M, Yamahara K, Takeda N, Chin‐Kanasaki M, Araki H, Sekine O, Yokoi H, Mukoyama M, Uzu T, Araki SI, Maegawa H. O‐linked beta‐N‐acetylglucosamine modification of proteins is essential for foot process maturation and survival in podocytes. Nephrol Dial Transplant 32: 1477‐1487, 2017.
 280.Osorio H, Bautista R, Rios A, Franco M, Santamaria J, Escalante B. Effect of treatment with losartan on salt sensitivity and SGLT2 expression in hypertensive diabetic rats. Diabetes Res Clin Pract 86: e46‐e49, 2009.
 281.Owen OE, Felig P, Morgan AP, Wahren J, Cahill GF Jr. Liver and kidney metabolism during prolonged starvation. J Clin Invest 48: 574‐583, 1969.
 282.Owen OE, Kalhan SC, Hanson RW. The key role of anaplerosis and cataplerosis for citric acid cycle function. J Biol Chem 277: 30409‐30412, 2002.
 283.Packer M, Anker SD, Butler J, Filippatos G, Pocock SJ, Carson P, Januzzi J, Verma S, Tsutsui H, Brueckmann M, Jamal W, Kimura K, Schnee J, Zeller C, Cotton D, Bocchi E, Bohm M, Choi DJ, Chopra V, Chuquiure E, Giannetti N, Janssens S, Zhang J, Gonzalez Juanatey JR, Kaul S, Brunner‐La Rocca HP, Merkely B, Nicholls SJ, Perrone S, Pina I, Ponikowski P, Sattar N, Senni M, Seronde MF, Spinar J, Squire I, Taddei S, Wanner C, Zannad F, Investigators EM‐RT. Cardiovascular and renal outcomes with empagliflozin in heart failure. N Engl J Med 383: 1413‐1424, 2020.
 284.Palatini P, Mormino P, Dorigatti F, Santonastaso M, Mos L, De Toni R, Winnicki M, Dal Follo M, Biasion T, Garavelli G, Pessina AC, Group HS. Glomerular hyperfiltration predicts the development of microalbuminuria in stage 1 hypertension: The HARVEST. Kidney Int 70: 578‐584, 2006.
 285.Panchapakesan U, Pegg K, Gross S, Komala MG, Mudaliar H, Forbes J, Pollock C, Mather A. Effects of SGLT2 inhibition in human kidney proximal tubular cells–renoprotection in diabetic nephropathy? PLoS One 8: e54442, 2013.
 286.Papademetriou V, Lovato L, Doumas M, Nylen E, Mottl A, Cohen RM, Applegate WB, Puntakee Z, Yale JF, Cushman WC. Chronic kidney disease and intensive glycemic control increase cardiovascular risk in patients with type 2 diabetes. Kidney Int 87: 649‐659, 2015.
 287.Park TJ, Reznick J, Peterson BL, Blass G, Omerbasic D, Bennett NC, Kuich P, Zasada C, Browe BM, Hamann W, Applegate DT, Radke MH, Kosten T, Lutermann H, Gavaghan V, Eigenbrod O, Begay V, Amoroso VG, Govind V, Minshall RD, Smith ESJ, Larson J, Gotthardt M, Kempa S, Lewin GR. Fructose‐driven glycolysis supports anoxia resistance in the naked mole‐rat. Science 356: 307‐311, 2017.
 288.Pascual JM, Wang D, Lecumberri B, Yang H, Mao X, Yang R, De Vivo DC. GLUT1 deficiency and other glucose transporter diseases. Eur J Endocrinol 150: 627‐633, 2004.
 289.Pereira‐Moreira R, Muscelli E. Effect of Insulin on Proximal Tubules Handling of Glucose: A Systematic Review. J Diabetes Res 2020: 8492467, 2020.
 290.Perkins BA, Cherney DZ, Partridge H, Soleymanlou N, Tschirhart H, Zinman B, Fagan NM, Kaspers S, Woerle HJ, Broedl UC, Johansen OE. Sodium‐Glucose Cotransporter 2 Inhibition and Glycemic Control in Type 1 Diabetes: Results of an 8‐Week Open‐Label Proof‐of‐Concept Trial. Diabetes Care 37: 1480‐1483, 2014.
 291.Perkovic V, Jardine M, Vijapurkar U, Meininger G. Renal effects of canagliflozin in type 2 diabetes mellitus. Curr Med Res Opin 31: 2219‐2231, 2015.
 292.Perkovic V, Jardine MJ, Neal B, Bompoint S, Heerspink HJL, Charytan DM, Edwards R, Agarwal R, Bakris G, Bull S, Cannon CP, Capuano G, Chu PL, de ZD, Greene T, Levin A, Pollock C, Wheeler DC, Yavin Y, Zhang H, Zinman B, Meininger G, Brenner BM, and Mahaffey KW. Canagliflozin and Renal Outcomes in Type 2 Diabetes and Nephropathy. N Engl J Med 380: 2295‐2306, 2019.
 293.Persson P, Hansell P, Palm F. Reduced adenosine A2a receptor‐mediated efferent arteriolar vasodilation contributes to diabetes‐induced glomerular hyperfiltration. Kidney Int 87: 109‐115, 2015.
 294.Pessoa TD, Campos LC, Carraro‐Lacroix L, Girardi AC, Malnic G. Functional role of glucose metabolism, osmotic stress, and sodium‐glucose cotransporter isoform‐mediated transport on Na+/H+ exchanger isoform 3 activity in the renal proximal tubule. J Am Soc Nephrol 25: 2028‐2039, 2014.
 295.Pfaff IL, Vallon V. Protein kinase C beta isoenzymes in diabetic kidneys and their relation to nephroprotective actions of the ACE inhibitor lisinopril. Kidney Blood Press Res 25: 329‐340, 2002.
 296.Pfaff IL, Wagner HJ, Vallon V. Immunolocalization of protein kinase C isoenzymes alpha, beta1 and betaII in rat kidney. J Am Soc Nephrol 10: 1861‐1873, 1999.
 297.Phillips AO, Steadman R, Morrisey K, Williams JD. Polarity of stimulation and secretion of transforming growth factor‐beta 1 by cultured proximal tubular cells. Am J Pathol 150: 1101‐1111, 1997.
 298.Powell DR, DaCosta CM, Gay J, Ding ZM, Smith M, Greer J, Doree D, Jeter‐Jones S, Mseeh F, Rodriguez LA, Harris A, Buhring L, Platt KA, Vogel P, Brommage R, Shadoan MK, Sands AT, Zambrowicz B. Improved glycemic control in mice lacking Sglt1 and Sglt2. Am J Physiol Endocrinol Metab 304: E117‐E130, 2013.
 299.Powell DR, DaCosta CM, Smith M, Doree D, Harris A, Buhring L, Heydorn W, Nouraldeen A, Xiong W, Yalamanchili P, Mseeh F, Wilson A, Shadoan M, Zambrowicz B, Ding ZM. Effect of LX4211 on glucose homeostasis and body composition in preclinical models. J Pharmacol Exp Ther 350: 232‐242, 2014.
 300.Pruijm M, Milani B, Pivin E, Podhajska A, Vogt B, Stuber M, Burnier M. Reduced cortical oxygenation predicts a progressive decline of renal function in patients with chronic kidney disease. Kidney Int 93: 932‐940, 2018.
 301.Qiu H, Novikov A, Vallon V. Ketosis and diabetic ketoacidosis in response to SGLT2 inhibitors: Basic mechanisms and therapeutic perspectives. Diabetes Metab Res Rev 33: 5, 2017.
 302.Quamme GA, Freeman HJ. Evidence for a high‐affinity sodium‐dependent D‐glucose transport system in the kidney. Am J Physiol 253: F151‐F157, 1987.
 303.Quinn PG, Yeagley D. Insulin regulation of PEPCK gene expression: A model for rapid and reversible modulation. Curr Drug Targets Immune Endocr Metabol Disord 5: 423‐437, 2005.
 304.Rahmoune H, Thompson PW, Ward JM, Smith CD, Hong G, Brown J. Glucose transporters in human renal proximal tubular cells isolated from the urine of patients with non‐insulin‐dependent diabetes. Diabetes 54: 3427‐3434, 2005.
 305.Raivio KO, Kekomaki MP, Maenpaa PH. Depletion of liver adenine nucleotides induced by D‐fructose. Dose‐dependence and specificity of the fructose effect. Biochem Pharmacol 18: 2615‐2624, 1969.
 306.Rajasekeran H, Lytvyn Y, Bozovic A, Lovshin JA, Diamandis E, Cattran D, Husain M, Perkins BA, Advani A, Reich HN, Kulasingam V, Cherney DZI. Urinary adenosine excretion in type 1 diabetes. Am J Physiol Renal Physiol 313: F184‐F191, 2017.
 307.Rand EB, Depaoli AM, Davidson NO, Bell GI, Burant CF. Sequence, tissue distribution, and functional characterization of the rat fructose transporter GLUT5. Am J Physiol 264: G1169‐G1176, 1993.
 308.Rasch R. Tubular lesions in streptozotocin‐diabetic rats. Diabetologia 27: 32‐37, 1984.
 309.Rebholz CM, Young BA, Katz R, Tucker KL, Carithers TC, Norwood AF, Correa A. Patterns of beverages consumed and risk of incident kidney disease. Clin J Am Soc Nephrol 14: 49‐56, 2019.
 310.Reinecke RM, Hauser PJ. Arterio venous blood sugar studies on the kidney of the eviscerated dog. Fed Proc 7: 99, 1948.
 311.Ren Y, Garvin JL, Liu R, Carretero OA. Possible mechanism of efferent arteriole (Ef‐Art) tubuloglomerular feedback. Kidney Int 71: 861‐866, 2007.
 312.Rieg T, Masuda T, Gerasimova M, Mayoux E, Platt K, Powell DR, Thomson SC, Koepsell H, Vallon V. Increase in SGLT1‐mediated transport explains renal glucose reabsorption during genetic and pharmacological SGLT2 inhibition in euglycemia. Am J Physiol Renal Physiol 306: F188‐F193, 2014.
 313.Ritchie S, Waugh D. The pathology of Armanni‐Ebstein diabetic nephropathy. Am J Pathol 33: 1035‐1057, 1957.
 314.Rohde R, Deetjen P. Glucose reabsorption in the rat kidney. Micropuncture analysis of tubular glucose concentration during free flow. Pflugers Arch 302: 219‐232, 1968.
 315.Roncal‐Jimenez CA, Ishimoto T, Lanaspa MA, Milagres T, Hernando AA, Jensen T, Miyazaki M, Doke T, Hayasaki T, Nakagawa T, Marumaya S, Long DA, Garcia GE, Kuwabara M, Sanchez‐Lozada LG, Kang DH, Johnson RJ. Aging‐associated renal disease in mice is fructokinase dependent. Am J Physiol Renal Physiol 311: F722‐F730, 2016.
 316.Roncal Jimenez CA, Ishimoto T, Lanaspa MA, Rivard CJ, Nakagawa T, Ejaz AA, Cicerchi C, Inaba S, Le M, Miyazaki M, Glaser J, Correa‐Rotter R, Gonzalez MA, Aragon A, Wesseling C, Sanchez‐Lozada LG, Johnson RJ. Fructokinase activity mediates dehydration‐induced renal injury. Kidney Int 86: 294‐302, 2014.
 317.Rosen S, Epstein FH, Brezis M. Determinants of intrarenal oxygenation: Factors in acute renal failure. Ren Fail 14: 321‐325, 1992.
 318.Saab KR, Kendrick J, Yracheta JM, Lanaspa MA, Pollard M, Johnson RJ. New insights on the risk for cardiovascular disease in African Americans: The role of added sugars. J Am Soc Nephrol 26: 247‐257, 2015.
 319.Sabolic I, Vrhovac I, Eror DB, Gerasimova M, Rose M, Breljak D, Ljubojevic M, Brzica H, Sebastiani A, Thal SC, Sauvant C, Kipp H, Vallon V, Koepsell H. Expression of Na+‐D‐glucose cotransporter SGLT2 in rodents is kidney‐specific and exhibits sex and species differences. Am J Physiol Cell Physiol 302: C1174‐C1188, 2012.
 320.Sala‐Rabanal M, Hirayama BA, Ghezzi C, Liu J, Huang SC, Kepe V, Koepsell H, Yu A, Powell DR, Thorens B, Wright EM, Barrio JR. Revisiting the physiological roles of SGLTs and GLUTs using positron emission tomography in mice. J Physiol 594: 4425‐4438, 2016.
 321.Saldana TM, Basso O, Darden R, Sandler DP. Carbonated beverages and chronic kidney disease. Epidemiology 18: 501‐506, 2007.
 322.Salomon LL, Lanza FL, Smith DE. Renal conversion of fructose to glucose. Am J Physiol 200: 871‐877, 1961.
 323.Sanchez‐Lozada LG, Andres‐Hernando A, Garcia‐Arroyo FE, Cicerchi C, Li N, Kuwabara M, Roncal‐Jimenez CA, Johnson RJ, Lanaspa MA. Uric acid activates aldose reductase and the polyol pathway for endogenous fructose and fat production causing development of fatty liver in rats. J Biol Chem 294: 4272‐4281, 2019.
 324.Sanchez‐Lozada LG, Lanaspa MA, Cristobal‐Garcia M, Garcia‐Arroyo F, Soto V, Cruz‐Robles D, Nakagawa T, Yu MA, Kang DH, Johnson RJ. Uric acid‐induced endothelial dysfunction is associated with mitochondrial alterations and decreased intracellular ATP concentrations. Nephron Experimental nephrology 121: e71‐e78, 2012.
 325.Sanchez‐Lozada LG, Tapia E, Jimenez A, Bautista P, Cristobal M, Nepomuceno T, Soto V, Avila‐Casado C, Nakagawa T, Johnson RJ, Herrera‐Acosta J, Franco M. Fructose‐induced metabolic syndrome is associated with glomerular hypertension and renal microvascular damage in rats. Am J Physiol Renal Physiol 292: F423‐F429, 2007.
 326.Santer R, Calado J. Familial renal glucosuria and SGLT2: From a Mendelian trait to a therapeutic target. Clin J Am Soc Nephrol 5: 133‐141, 2010.
 327.Santer R, Groth S, Kinner M, Dombrowski A, Berry GT, Brodehl J, Leonard JV, Moses S, Norgren S, Skovby F, Schneppenheim R, Steinmann B, Schaub J. The mutation spectrum of the facilitative glucose transporter gene SLC2A2 (GLUT2) in patients with Fanconi‐Bickel syndrome. Hum Genet 110: 21‐29, 2002.
 328.Santer R, Schneppenheim R, Suter D, Schaub J, Steinmann B. Fanconi‐Bickel syndrome–the original patient and his natural history, historical steps leading to the primary defect, and a review of the literature. Eur J Pediatr 157: 783‐797, 1998.
 329.Saponaro C, Muhlemann M, Acosta‐Montalvo A, Piron A, Gmyr V, Delalleau N, Moerman E, Thevenet J, Pasquetti G, Coddeville A, Cnop M, Kerr‐Conte J, Staels B, Pattou F, Bonner C. Inter‐individual heterogeneity of SGLT2 expression and function in human pancreatic islets. Diabetes, 2020.
 330.Sasaki M, Sasako T, Kubota N, Sakurai Y, Takamoto I, Kubota T, Inagi R, Seki G, Goto M, Ueki K, Nangaku M, Jomori T, Kadowaki T. Dual regulation of gluconeogenesis by insulin and glucose in the proximal tubules of the kidney. Diabetes 66: 2339‐2350, 2017.
 331.Satirapoj B, Korkiatpitak P, Supasyndh O. Effect of sodium‐glucose cotransporter 2 inhibitor on proximal tubular function and injury in patients with type 2 diabetes: A randomized controlled trial. Clin Kidney J 12: 326‐332, 2019.
 332.Sato Y, Feig DI, Stack AG, Kang DH, Lanaspa MA, Ejaz AA, Sanchez‐Lozada LG, Kuwabara M, Borghi C, Johnson RJ. The case for uric acid‐lowering treatment in patients with hyperuricaemia and CKD. Nat Rev Nephrol 15: 767‐775, 2019.
 333.Schleicher ED, Weigert C. Role of the hexosamine biosynthetic pathway in diabetic nephropathy. Kidney Int Suppl 77: S13‐S18, 2000.
 334.Schmidt C, Hocherl K, Schweda F, Kurtz A, Bucher M. Regulation of renal sodium transporters during severe inflammation. J Am Soc Nephrol 18: 1072‐1083, 2007.
 335.Schmidt U, Guder WG. Sites of enzyme activity along the nephron. Kidney Int 9: 233‐242, 1976.
 336.Schmidt U, Marosvari I, Dubach UC. Renal metabolism of glucose: Anatomical sites of hexokinase activity in the rat nephron. FEBS Lett 53: 26‐28, 1975.
 337.Schnermann J, Briggs JP. Single nephron comparison of the effect of loop of Henle flow on filtration rate and pressure in control and angiotensin II‐infused rats. Miner Electrolyte Metab 15: 103‐107, 1989.
 338.Schulte K, Kunter U, Moeller MJ. The evolution of blood pressure and the rise of mankind. Nephrol Dial Transplant 30: 713‐723, 2015.
 339.Schurek HJ, Johns O. Is tubuloglomerular feedback a tool to prevent nephron oxygen deficiency? Kidney Int 51: 386‐392, 1997.
 340.Scott TW, Setchell BP, Bassett JM. Characterization and metabolism of ovine foetal lipids. Biochem J 104: 1040‐1047, 1967.
 341.Seidner G, Alvarez MG, Yeh JI, O'Driscoll KR, Klepper J, Stump TS, Wang D, Spinner NB, Birnbaum MJ, De Vivo DC. GLUT‐1 deficiency syndrome caused by haploinsufficiency of the blood‐brain barrier hexose carrier. Nat Genet 18: 188‐191, 1998.
 342.Sha S, Devineni D, Ghosh A, Polidori D, Chien S, Wexler D, Shalayda K, Demarest K, Rothenberg P. Canagliflozin, a novel inhibitor of sodium glucose co‐transporter 2, dose dependently reduces calculated renal threshold for glucose excretion and increases urinary glucose excretion in healthy subjects. Diabetes Obes Metab 13: 669‐672, 2011.
 343.Shanley PF, Brezis M, Spokes K, Silva P, Epstein FH, Rosen S. Transport‐dependent cell injury in the S3 segment of the proximal tubule. Kidney Int 29: 1033‐1037, 1986.
 344.Shen L, Kristensen SL, Bengtsson O, Bohm M, de Boer RA, Docherty KF, Inzucchi SE, Katova T, Kober L, Kosiborod MN, Langkilde AM, Lindholm D, Martinez MFA, O'Meara E, Nicolau JC, Petrie MC, Ponikowski P, Sabatine MS, Schou M, Sjostrand M, Solomon SD, Jhund PS, McMurray JJV. Dapagliflozin in HFrEF patients treated with mineralocorticoid receptor antagonists: An analysis of DAPA‐HF. JACC Heart Fail 9: 254‐264, 2021.
 345.Shepard BD, Pluznick JL. Saving the sweetness: Renal glucose handling in health and disease. Am J Physiol Renal Physiol 313: F55‐F61, 2017.
 346.Shinozaki K, Kashiwagi A, Nishio Y, Okamura T, Yoshida Y, Masada M, Toda N, Kikkawa R. Abnormal biopterin metabolism is a major cause of impaired endothelium‐dependent relaxation through nitric oxide/O2‐ imbalance in insulin‐resistant rat aorta. Diabetes 48: 2437‐2445, 1999.
 347.Shiota M, Galassetti P, Monohan M, Neal DW, Cherrington AD. Small amounts of fructose markedly augment net hepatic glucose uptake in the conscious dog. Diabetes 47: 867‐873, 1998.
 348.Shoham DA, Durazo‐Arvizu R, Kramer H, Luke A, Vupputuri S, Kshirsagar A, Cooper RS. Sugary soda consumption and albuminuria: Results from the National Health and Nutrition Examination Survey, 1999‐2004. PLoS One 3: e3431, 2008.
 349.Singh AK, Amlal H, Haas PJ, Dringenberg U, Fussell S, Barone SL, Engelhardt R, Zuo J, Seidler U, Soleimani M. Fructose‐induced hypertension: Essential role of chloride and fructose absorbing transporters PAT1 and Glut5. Kidney Int 74: 438‐447, 2008.
 350.So A, Thorens B. Uric acid transport and disease. J Clin Invest 120: 1791‐1799, 2010.
 351.Softic S, Meyer JG, Wang GX, Gupta MK, Batista TM, Lauritzen H, Fujisaka S, Serra D, Herrero L, Willoughby J, Fitzgerald K, Ilkayeva O, Newgard CB, Gibson BW, Schilling B, Cohen DE, Kahn CR. Dietary sugars alter hepatic fatty acid oxidation via transcriptional and post‐translational modifications of mitochondrial proteins. Cell Metab 30: 735‐753 e734, 2019.
 352.Solini A, Rossi C, Mazzanti CM, Proietti A, Koepsell H, Ferrannini E. Sodium‐glucose co‐transporter (SGLT)2 and SGLT1 renal expression in patients with type 2 diabetes. Diabetes Obes Metab 19: 1289‐1294, 2017.
 353.Soncrant T, Komnenov D, Beierwaltes WH, Chen H, Wu M, Rossi NF. Bilateral renal cryodenervation decreases arterial pressure and improves insulin sensitivity in fructose‐fed Sprague‐Dawley rats. Am J Physiol Regul Integr Comp Physiol 315: R529‐R538, 2018.
 354.Song P, Huang W, Onishi A, Patel R, Kim Y, van Ginkel C, Fu Y, Freeman B, Koepsell H, Thomson SC, Liu R, Vallon V. Knockout of Na‐glucose‐cotransporter SGLT1 mitigates diabetes‐induced upregulation of nitric oxide synthase‐1 in macula densa and glomerular hyperfiltration. Am J Physiol Renal Physiol 317: F207‐F217, 2019.
 355.Song Z, Roncal‐Jimenez CA, Lanaspa‐Garcia MA, Oppelt SA, Kuwabara M, Jensen T, Milagres T, Andres‐Hernando A, Ishimoto T, Garcia GE, Johnson G, MacLean PS, Sanchez‐Lozada LG, Tolan DR, Johnson RJ. Role of fructose and fructokinase in acute dehydration‐induced vasopressin gene expression and secretion in mice. J Neurophysiol 117: 646‐654, 2017.
 356.Sotak M, Marks J, Unwin RJ. Putative tissue location and function of the SLC5 family member SGLT3. Exp Physiol 102: 5‐13, 2017.
 357.Soty M, Penhoat A, Amigo‐Correig M, Vinera J, Sardella A, Vullin‐Bouilloux F, Zitoun C, Houberdon I, Mithieux G. A gut‐brain neural circuit controlled by intestinal gluconeogenesis is crucial in metabolic health. Mol Metab 4: 106‐117, 2015.
 358.Srivastava SP, Goodwin JE, Kanasaki K, Koya D. Metabolic reprogramming by N‐acetyl‐seryl‐aspartyl‐lysyl‐proline protects against diabetic kidney disease. Br J Pharmacol 177: 3691‐3711, 2020.
 359.Srivastava SP, Li J, Kitada M, Fujita H, Yamada Y, Goodwin JE, Kanasaki K, Koya D. SIRT3 deficiency leads to induction of abnormal glycolysis in diabetic kidney with fibrosis. Cell Death Dis 9: 997, 2018.
 360.Stenvinkel P, Jani AH, Johnson RJ. Hibernating bears (Ursidae): Metabolic magicians of definite interest for the nephrologist. Kidney Int 83: 207‐212, 2013.
 361.Suga T, Kikuchi O, Kobayashi M, Matsui S, Yokota‐Hashimoto H, Wada E, Kohno D, Sasaki T, Takeuchi K, Kakizaki S, Yamada M, Kitamura T. SGLT1 in pancreatic alpha cells regulates glucagon secretion in mice, possibly explaining the distinct effects of SGLT2 inhibitors on plasma glucagon levels. Mol Metab 19: 1‐12, 2019.
 362.Sugawara‐Yokoo M, Suzuki T, Matsuzaki T, Naruse T, Takata K. Presence of fructose transporter GLUT5 in the S3 proximal tubules in the rat kidney. Kidney Int 56: 1022‐1028, 1999.
 363.Sullivan MA, Forbes JM. Glucose and glycogen in the diabetic kidney: Heroes or villains? EBioMedicine 47: 590‐597, 2019.
 364.Sun S, Ning X, Zhang Y, Lu Y, Nie Y, Han S, Liu L, Du R, Xia L, He L, Fan D. Hypoxia‐inducible factor‐1alpha induces Twist expression in tubular epithelial cells subjected to hypoxia, leading to epithelial‐to‐mesenchymal transition. Kidney Int 75: 1278‐1287, 2009.
 365.Sundborn G, Thornley S, Merriman TR, Lang B, King C, Lanaspa MA, Johnson RJ. Are liquid sugars different from solid sugar in their ability to cause metabolic syndrome? Obesity (Silver Spring) 27: 879‐887, 2019.
 366.Swe MT, Pongchaidecha A, Chatsudthipong V, Chattipakorn N, Lungkaphin A. Molecular signaling mechanisms of renal gluconeogenesis in nondiabetic and diabetic conditions. J Cell Physiol 234: 8134‐8151, 2019.
 367.Szablewski L. Distribution of glucose transporters in renal diseases. J Biomed Sci 24: 64, 2017.
 368.Takagi S, Li J, Takagaki Y, Kitada M, Nitta K, Takasu T, Kanasaki K, Koya D. Ipragliflozin improves mitochondrial abnormalities in renal tubules induced by a high‐fat diet. J Diabetes Investig 9: 1025‐1032, 2018.
 369.Tazawa S, Yamato T, Fujikura H, Hiratochi M, Itoh F, Tomae M, Takemura Y, Maruyama H, Sugiyama T, Wakamatsu A, Isogai T, Isaji M. SLC5A9/SGLT4, a new Na+‐dependent glucose transporter, is an essential transporter for mannose, 1,5‐anhydro‐D‐glucitol, and fructose. Life Sci 76: 1039‐1050, 2005.
 370.Thach TT, Wu C, Hwang KY, Lee SJ. Azelaic acid induces mitochondrial biogenesis in skeletal muscle by activation of olfactory receptor 544. Front Physiol 11: 329, 2020.
 371.Thomson S, Vallon V, Blantz RC. Asymmetry of tubuloglomerular feedback effector mechanism with respect to ambient tubular flow. Am J Physiol 271: F1123‐F1130, 1996.
 372.Thomson SC, Deng A, Komine N, Hammes JS, Blantz RC, Gabbai FB. Early diabetes as a model for testing the regulation of juxtaglomerular NOS I. Am J Physiol Renal Physiol 287: F732‐F738, 2004.
 373.Thomson SC, Rieg T, Miracle C, Mansoury H, Whaley J, Vallon V, Singh P. Acute and chronic effects of SGLT2 blockade on glomerular and tubular function in the early diabetic rat. Am J Physiol Regul Integr Comp Physiol 302: R75‐R83, 2012.
 374.Thomson SC, Vallon V. Effects of SGLT2 inhibitor and dietary NaCl on glomerular hemodynamics assessed by micropuncture in diabetic rats. Am J Physiol Renal Physiol 320: F761‐F777, 2021.
 375.Thomson SC, Vallon V. Renal Effects of Sodium‐Glucose Co‐Transporter Inhibitors. Am J Cardiol 124 (Suppl 1): S28‐S35, 2019.
 376.Thorens B, Lodish HF, Brown D. Differential localization of two glucose transporter isoforms in rat kidney. Am J Physiol 259: C286‐C294, 1990.
 377.Tiku A, Johnson DW, Badve SV. Recent evidence on the effect of urate‐lowering treatment on the progression of kidney disease. Curr Opin Nephrol Hypertens 30: 346‐352, 2021.
 378.Tiwari S, Riazi S, Ecelbarger CA. Insulin's impact on renal sodium transport and blood pressure in health, obesity, and diabetes. Am J Physiol Renal Physiol 293: F974‐F984, 2007.
 379.Tojo A, Onozato ML, Fujita T. Role of macula densa neuronal nitric oxide synthase in renal diseases. Med Mol Morphol 39: 2‐7, 2006.
 380.Tolins JP, Shultz PJ, Raij L, Brown DM, Mauer SM. Abnormal renal hemodynamic response to reduced renal perfusion pressure in diabetic rats: Role of NO. Am J Physiol 265: F886‐F895, 1993.
 381.Tomita I, Kume S, Sugahara S, Osawa N, Yamahara K, Yasuda‐Yamahara M, Takeda N, Chin‐Kanasaki M, Kaneko T, Mayoux E, Mark M, Yanagita M, Ogita H, Araki SI, Maegawa H. SGLT2 inhibition mediates protection from diabetic kidney disease by promoting ketone body‐induced mTORC1 inhibition. Cell Metab 32: 404‐419, 2020.
 382.Toyoki D, Shibata S, Kuribayashi‐Okuma E, Xu N, Ishizawa K, Hosoyamada M, Uchida S. Insulin stimulates uric acid reabsorption via regulating urate transporter 1 and ATP‐binding cassette subfamily G member 2. Am J Physiol Renal Physiol 313: F826‐F834, 2017.
 383.Turner RJ, Moran A. Heterogeneity of sodium‐dependent D‐glucose transport sites along the proximal tubule: Evidence from vesicle studies. Am J Physiol 242: F406‐F414, 1982.
 384.Uchida S, Endou H. Substrate specificity to maintain cellular ATP along the mouse nephron. Am J Physiol 255: F977‐F983, 1988.
 385.Umino H, Hasegawa K, Minakuchi H, Muraoka H, Kawaguchi T, Kanda T, Tokuyama H, Wakino S, Itoh H. High basolateral glucose increases sodium‐glucose cotransporter 2 and reduces Sirtuin‐1 in renal tubules through glucose transporter‐2 detection. Sci Rep 8: 6791, 2018.
 386.Uthman L, Baartscheer A, Bleijlevens B, Schumacher CA, Fiolet JWT, Koeman A, Jancev M, Hollmann MW, Weber NC, Coronel R, Zuurbier CJ. Class effects of SGLT2 inhibitors in mouse cardiomyocytes and hearts: Inhibition of Na(+)/H(+) exchanger, lowering of cytosolic Na(+) and vasodilation. Diabetologia 61: 722‐726, 2018.
 387.Uthman L, Nederlof R, Eerbeek O, Baartscheer A, Schumacher C, Buchholtz N, Hollmann MW, Coronel R, Weber NC, Zuurbier CJ. Delayed ischaemic contracture onset by empagliflozin associates with NHE1 inhibition and is dependent on insulin in isolated mouse hearts. Cardiovasc Res 115: 1533‐1545, 2019.
 388.Vallon V. Glucose transporters in the kidney in health and disease. Pflugers Arch 472: 1345‐1370, 2020.
 389.Vallon V. The mechanisms and therapeutic potential of SGLT2 inhibitors in diabetes mellitus. Annu Rev Med 66: 255‐270, 2015.
 390.Vallon V. Molecular determinants of renal glucose transport. Am J Physiol Cell Physiol 300: C6‐C8, 2011.
 391.Vallon V. The proximal tubule in the pathophysiology of the diabetic kidney. Am J Physiol Regul Integr Comp Physiol 300: R1009‐R1022, 2011.
 392.Vallon V. Tubular transport in acute kidney injury: relevance for diagnosis. Prognosis and Intervention. Nephron 134: 160‐166, 2016.
 393.Vallon V, Blantz RC, Thomson S. Glomerular hyperfiltration and the salt paradox in early type 1 diabetes mellitus: A tubulo‐centric view. J Am Soc Nephrol 14: 530‐537, 2003.
 394.Vallon V, Blantz RC, Thomson S. Homeostatic efficiency of tubuloglomerular feedback is reduced in established diabetes mellitus in rats. Am J Physiol 269: F876‐F883, 1995.
 395.Vallon V, Broer S, Nigam SK. Renal handling of organic solutes. In: Yu AS, Chertow G, Luyckx V, Marsden PA, Skorecki K, Taal MW, editors. Brenner & Rector's The Kidney. Philadelphia: Elsevier, 2020, p. 218‐246.
 396.Vallon V, Gerasimova M, Rose MA, Masuda T, Satriano J, Mayoux E, Koepsell H, Thomson SC, Rieg T. SGLT2 inhibitor empagliflozin reduces renal growth and albuminuria in proportion to hyperglycemia and prevents glomerular hyperfiltration in diabetic Akita mice. Am J Physiol Renal Physiol 306: F194‐F204, 2014.
 397.Vallon V, Grahammer F, Richter K, Bleich M, Lang F, Barhanin J, Volkl H, Warth R. Role of KCNE1‐dependent K+ fluxes in mouse proximal tubule. J Am Soc Nephrol 12: 2003‐2011, 2001.
 398.Vallon V, Grahammer F, Volkl H, Sandu CD, Richter K, Rexhepaj R, Gerlach U, Rong Q, Pfeifer K, Lang F. KCNQ1‐dependent transport in renal and gastrointestinal epithelia. Proc Natl Acad Sci U S A 102: 17864‐17869, 2005.
 399.Vallon V, Komers R. Pathophysiology of the diabetic kidney. Compr Physiol 1: 1175‐1232, 2011.
 400.Vallon V, Muhlbauer B, Osswald H. Adenosine and kidney function. Physiol Rev 86: 901‐940, 2006.
 401.Vallon V, Osswald H. Dipyridamole prevents diabetes‐induced alterations of kidney function in rats. Naunyn Schmiedebergs Arch Pharmacol 349: 217‐222, 1994.
 402.Vallon V, Platt KA, Cunard R, Schroth J, Whaley J, Thomson SC, Koepsell H, Rieg T. SGLT2 mediates glucose reabsorption in the early proximal tubule. J Am Soc Nephrol 22: 104‐112, 2011.
 403.Vallon V, Richter K, Blantz RC, Thomson S, Osswald H. Glomerular hyperfiltration in experimental diabetes mellitus: Potential role of tubular reabsorption. J Am Soc Nephrol 10: 2569‐2576, 1999.
 404.Vallon V, Richter K, Huang DY, Rieg T, Schnermann J. Functional consequences at the single‐nephron level of the lack of adenosine A1 receptors and tubuloglomerular feedback in mice. Pflugers Arch 448: 214‐221, 2004.
 405.Vallon V, Rose M, Gerasimova M, Satriano J, Platt KA, Koepsell H, Cunard R, Sharma K, Thomson SC, Rieg T. Knockout of Na‐glucose transporter SGLT2 attenuates hyperglycemia and glomerular hyperfiltration but not kidney growth or injury in diabetes mellitus. Am J Physiol Renal Physiol 304: F156‐F167, 2013.
 406.Vallon V, Schwark JR, Richter K, Hropot M. Role of Na(+)/H(+) exchanger NHE3 in nephron function: Micropuncture studies with S3226, an inhibitor of NHE3. Am J Physiol Renal Physiol 278: F375‐F379, 2000.
 407.Vallon V, Thomson S. Inhibition of local nitric oxide synthase increases homeostatic efficiency of tubuloglomerular feedback. Am J Physiol 269: F892‐F899, 1995.
 408.Vallon V, Thomson SC. Renal function in diabetic disease models: The tubular system in the pathophysiology of the diabetic kidney. Annu Rev Physiol 74: 351‐375, 2012.
 409.Vallon V, Thomson SC. Targeting renal glucose reabsorption to treat hyperglycaemia: The pleiotropic effects of SGLT2 inhibition. Diabetologia 60: 215‐225, 2017.
 410.Vallon V, Thomson SC. The tubular hypothesis of nephron filtration and diabetic kidney disease. Nat Rev Nephrol 16: 317‐336, 2020.
 411.Vallon V, Traynor T, Barajas L, Huang YG, Briggs JP, Schnermann J. Feedback control of glomerular vascular tone in neuronal nitric oxide synthase knockout mice. J Am Soc Nephrol 12: 1599‐1606, 2001.
 412.Vallon V, Unwin R, Inscho EW, Leipziger J, Kishore BK. Extracellular nucleotides and P2 receptors in renal function. Physiol Rev 100: 211‐269, 2020.
 413.Vallon V, Verma S. Effects of SGLT2 inhibitors on kidney and cardiovascular function. Annu Rev Physiol 83: 503‐528, 2021.
 414.van Bommel EJM, Muskiet MHA, van Baar MJB, Tonneijck L, Smits MM, Emanuel AL, Bozovic A, Danser AHJ, Geurts F, Hoorn EJ, Touw DJ, Larsen EL, Poulsen HE, Kramer MHH, Nieuwdorp M, Joles JA, van Raalte DH. The renal hemodynamic effects of the SGLT2 inhibitor dapagliflozin are caused by post‐glomerular vasodilatation rather than pre‐glomerular vasoconstriction in metformin‐treated patients with type 2 diabetes in the randomized, double‐blind RED trial. Kidney Int 97: 202‐212, 2020.
 415.Van Schaftingen E, Detheux M, Veiga da Cunha M. Short‐term control of glucokinase activity: Role of a regulatory protein. FASEB J 8: 414‐419, 1994.
 416.Veiras LC, Girardi ACC, Curry J, Pei L, Ralph DL, Tran A, Castelo‐Branco RC, Pastor‐Soler N, Arranz CT, Yu ASL, McDonough AA. Sexual dimorphic pattern of renal transporters and electrolyte homeostasis. J Am Soc Nephrol 28: 3504‐3517, 2017.
 417.Verma S, Rawat S, Ho KL, Wagg CS, Zhang L, Teoh H, Dyck JE, Uddin GM, Oudit GY, Mayoux E, Lehrke M, Marx N, Lopaschuk GD. Empagliflozin increases cardiac energy production in diabetes: novel translational insights into the heart failure benefits of SGLT2 inhibitors. JACC Basic Transl Sci 3: 575‐587, 2018.
 418.Vrhovac I, Balen ED, Klessen D, Burger C, Breljak D, Kraus O, Radovic N, Jadrijevic S, Aleksic I, Walles T, Sauvant C, Sabolic I, Koepsell H. Localizations of Na‐D‐glucose cotransporters SGLT1 and SGLT2 in human kidney and of SGLT1 in human small intestine, liver, lung, and heart. Pflugers Arch 467: 1881‐1898, 2015.
 419.Walker RW, Goran MI. Laboratory determined sugar content and composition of commerical infant formulas, baby foods and common grocery items targeted to children. Nutrients 7: 5850‐5867, 2015.
 420.Wang J, Shibayama Y, Kobori H, Liu Y, Kobara H, Masaki T, Wang Z, Nishiyama A. High glucose augments angiotensinogen in human renal proximal tubular cells through hepatocyte nuclear factor‐5. PLoS One 12: e0185600, 2017.
 421.Wang XX, Levi J, Luo Y, Myakala K, Herman‐Edelstein M, Qiu L, Wang D, Peng Y, Grenz A, Lucia S, Dobrinskikh E, D'Agati VD, Koepsell H, Kopp JB, Rosenberg A, Levi M. SGLT2 expression is increased in human diabetic nephropathy: SGLT2 inhibition decreases renal lipid accumulation, inflammation and the development of nephropathy in diabetic mice. J Biol Chem 292: 5335‐5348, 2017.
 422.Wanner C, Inzucchi SE, Lachin JM, Fitchett D, von Mattheus EM, Johansen OE, Woerle HJ, Broedl UC, Zinman B. Empagliflozin and progression of kidney disease in type 2 diabetes. N Engl J Med 375: 323‐334, 2016.
 423.Warburg O. On respiratory impairment in cancer cells. Science 124: 269‐270, 1956.
 424.Warburg O. On the origin of cancer cells. Science 123: 309‐314, 1956.
 425.Watanabe S, Kang DH, Feng L, Nakagawa T, Kanellis J, Lan H, Mazzali M, Johnson RJ. Uric acid, hominoid evolution, and the pathogenesis of salt‐sensitivity. Hypertension 40: 355‐360, 2002.
 426.Wei Q, Su J, Dong G, Zhang M, Huo Y, Dong Z. Glycolysis inhibitors suppress renal interstitial fibrosis via divergent effects on fibroblasts and tubular cells. Am J Physiol Renal Physiol 316: F1162‐F1172, 2019.
 427.Weinberg JM, Molitoris BA. Illuminating mitochondrial function and dysfunction using multiphoton technology. J Am Soc Nephrol 20: 1164‐1166, 2009.
 428.Wells RG, Pajor AM, Kanai Y, Turk E, Wright EM, Hediger MA. Cloning of a human kidney cDNA with similarity to the sodium‐glucose cotransporter. Am J Physiol 263: F459‐F465, 1992.
 429.Wen SF. Micropuncture studies of glucose transport in the dog: Mechanism of renal glucosuria. Am J Physiol 231: 468‐475, 1976.
 430.White CE, Piper EL, Noland PR, Daniels LB. Fructose utilization for nucleic acid synthesis in the fetal pig. J Anim Sci 55: 73‐76, 1982.
 431.Wilcox CS, Welch WJ, Murad F, Gross SS, Taylor G, Levi R, Schmidt HH. Nitric oxide synthase in macula densa regulates glomerular capillary pressure. Proc Natl Acad Sci U S A 89: 11993‐11997, 1992.
 432.Witkowska K, Smith KM, Yao SY, Ng AM, O'Neill D, Karpinski E, Young JD, Cheeseman CI. Human SLC2A9a and SLC2A9b isoforms mediate electrogenic transport of urate with different characteristics in the presence of hexoses. Am J Physiol Renal Physiol 303: F527‐F539, 2012.
 433.Wood IS, Trayhurn P. Glucose transporters (GLUT and SGLT): Expanded families of sugar transport proteins. Br J Nutr 89: 3‐9, 2003.
 434.Woods HF, Eggleston LV, Krebs HA. The cause of hepatic accumulation of fructose 1‐phosphate on fructose loading. Biochem J 119: 501‐510, 1970.
 435.Woroniecka KI, Park AS, Mohtat D, Thomas DB, Pullman JM, Susztak K. Transcriptome analysis of human diabetic kidney disease. Diabetes 60: 2354‐2369, 2011.
 436.Wright EM. Glucose transport families SLC5 and SLC50. Mol Aspects Med 34: 183‐196, 2013.
 437.Wright EM. Renal Na(+)‐glucose cotransporters. Am J Physiol Renal Physiol 280: F10‐F18, 2001.
 438.Wright EM, Loo DD, Hirayama BA. Biology of human sodium glucose transporters. Physiol Rev 91: 733‐794, 2011.
 439.Wright EM, Turk E. The sodium/glucose cotransport family SLC5. Pflugers Arch 447: 510‐518, 2004.
 440.Wu C, Hwang SH, Jia Y, Choi J, Kim YJ, Choi D, Pathiraja D, Choi IG, Koo SH, Lee SJ. Olfactory receptor 544 reduces adiposity by steering fuel preference toward fats. J Clin Invest 127: 4118‐4123, 2017.
 441.Yale JF, Bakris G, Cariou B, Yue D, David‐Neto E, Xi L, Figueroa K, Wajs E, Usiskin K, Meininger G. Efficacy and safety of canagliflozin in subjects with type 2 diabetes and chronic kidney disease. Diabetes Obes Metab 15: 463‐473, 2013.
 442.Yamaoka T, Nishimura C, Yamashita K, Itakura M, Yamada T, Fujimoto J, Kokai Y. Acute onset of diabetic pathological changes in transgenic mice with human aldose reductase cDNA. Diabetologia 38: 255‐261, 1995.
 443.Yin XN, Wang J, Cui LF, Fan WX. Enhanced glycolysis in the process of renal fibrosis aggravated the development of chronic kidney disease. Eur Rev Med Pharmacol Sci 22: 4243‐4251, 2018.
 444.You G, Lee WS, Barros EJ, Kanai Y, Huo TL, Khawaja S, Wells RG, Nigam SK, Hediger MA. Molecular characteristics of Na(+)‐coupled glucose transporters in adult and embryonic rat kidney. J Biol Chem 270: 29365‐29371, 1995.
 445.Zapata‐Morales JR, Galicia‐Cruz OG, Franco M, Martinez YM. Hypoxia‐inducible factor‐1alpha (HIF‐1alpha) protein diminishes sodium glucose transport 1 (SGLT1) and SGLT2 protein expression in renal epithelial tubular cells (LLC‐PK1) under hypoxia. J Biol Chem 289: 346‐357, 2014.
 446.Zelniker TA, Wiviott SD, Raz I, Im K, Goodrich EL, Bonaca MP, Mosenzon O, Kato ET, Cahn A, Furtado RHM, Bhatt DL, Leiter LA, McGuire DK, Wilding JPH, Sabatine MS. SGLT2 inhibitors for primary and secondary prevention of cardiovascular and renal outcomes in type 2 diabetes: A systematic review and meta‐analysis of cardiovascular outcome trials. Lancet 393: 31‐39, 2019.
 447.Zenner ZP, Gordish KL, Beierwaltes WH. Free radical scavenging reverses fructose‐induced salt‐sensitive hypertension. Integr Blood Press Control 11: 1‐9, 2018.
 448.Zhang G, Darshi M, Sharma K. The Warburg effect in diabetic kidney disease. Semin Nephrol 38: 111‐120, 2018.
 449.Zhang J, Wei J, Jiang S, Xu L, Wang L, Cheng F, Buggs J, Koepsell H, Vallon V, Liu R. Macula densa SGLT1‐NOS1‐TGF pathway – a new mechanism for glomerular hyperfiltration during hyperglycemia. J Am Soc Nephrol 30: 578‐593, 2019.
 450.Zhang SL, Filep JG, Hohman TC, Tang SS, Ingelfinger JR, Chan JS. Molecular mechanisms of glucose action on angiotensinogen gene expression in rat proximal tubular cells. Kidney Int 55: 454‐464, 1999.
 451.Zhang Y, Nakano D, Guan Y, Hitomi H, Uemura A, Masaki T, Kobara H, Sugaya T, Nishiyama A. A sodium‐glucose cotransporter 2 inhibitor attenuates renal capillary injury and fibrosis by a vascular endothelial growth factor‐dependent pathway after renal injury in mice. Kidney Int 94: 524‐535, 2018.
 452.Zhao S, Jang C, Liu J, Uehara K, Gilbert M, Izzo L, Zeng X, Trefely S, Fernandez S, Carrer A, Miller KD, Schug ZT, Snyder NW, Gade TP, Titchenell PM, Rabinowitz JD, Wellen KE. Dietary fructose feeds hepatic lipogenesis via microbiota‐derived acetate. Nature 579: 586‐591, 2020.
 453.Zierler K. Whole body glucose metabolism. Am J Physiol 276: E409‐E426, 1999.
 454.Zinman B, Wanner C, Lachin JM, Fitchett D, Bluhmki E, Hantel S, Mattheus M, Devins T, Johansen OE, Woerle HJ, Broedl UC, Inzucchi SE. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med 373: 2117‐2128, 2015.

Contact Editor

Submit a note to the editor about this article by filling in the form below.

* Required Field

How to Cite

Volker Vallon, Takahiko Nakagawa. Renal Tubular Handling of Glucose and Fructose in Health and Disease. Compr Physiol 2021, 12: 2995-3044. doi: 10.1002/cphy.c210030