Skip Navigation
Skip to contents

Diabetes Metab J : Diabetes & Metabolism Journal

Search
OPEN ACCESS

Articles

Page Path
HOME > Diabetes Metab J > Volume 46(4); 2022 > Article
Review
Pathophysiology Renoprotective Mechanism of Sodium-Glucose Cotransporter 2 Inhibitors: Focusing on Renal Hemodynamics
Nam Hoon Kimorcid, Nan Hee Kimorcid
Diabetes & Metabolism Journal 2022;46(4):543-551.
DOI: https://doi.org/10.4093/dmj.2022.0209
Published online: July 27, 2022
  • 5,973 Views
  • 645 Download
  • 9 Web of Science
  • 10 Crossref
  • 10 Scopus

Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea

Corresponding author: Nan Hee Kim orcid Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan 15355, Korea E-mail: nhkendo@gamil.com
• Received: June 19, 2022   • Accepted: July 20, 2022

Copyright © 2022 Korean Diabetes Association

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (https://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

  • Diabetic kidney disease (DKD) is a prevalent renal complication of diabetes mellitus that ultimately develops into end-stage kidney disease (ESKD) when not managed appropriately. Substantial risk of ESKD remains even with intensive management of hyperglycemia and risk factors of DKD and timely use of renin-angiotensin-aldosterone inhibitors. Sodium-glucose cotransporter 2 (SGLT2) inhibitors reduce hyperglycemia primarily by inhibiting glucose and sodium reabsorption in the renal proximal tubule. Currently, their effects expand to prevent or delay cardiovascular and renal adverse events, even in those without diabetes. In dedicated renal outcome trials, SGLT2 inhibitors significantly reduced the risk of composite renal adverse events, including the development of ESKD or renal replacement therapy, which led to the positioning of SGLT2 inhibitors as the mainstay of chronic kidney disease management. Multiple mechanisms of action of SGLT2 inhibitors, including hemodynamic, metabolic, and anti-inflammatory effects, have been proposed. Restoration of tubuloglomerular feedback is a plausible explanation for the alteration in renal hemodynamics induced by SGLT2 inhibition and for the associated renal benefit. This review discusses the clinical rationale and mechanism related to the protection SGLT2 inhibitors exert on the kidney, focusing on renal hemodynamic effects.
Diabetic kidney disease (DKD) is one of the most important complications in patients with type 1 and type 2 diabetes mellitus. Diabetes is currently the most common cause of chronic kidney disease (CKD), leading to kidney failure worldwide [1]. In South Korea, nearly 50% of end-stage kidney disease (ESKD) cases are caused by diabetes [2]. Over 30% of people with diabetes have albuminuria or reduced glomerular filtration rate (GFR) [3]. The progress of standard care and the advent of new drugs have substantially decreased the risk of morbidity and mortality in patients with diabetes [4]. Nonetheless, the prevalence of DKD and ESKD is steadily increasing, whereas that of ischemic heart disease and ischemic stroke has gradually decreased over the past decades [5,6]. Given the significantly increased mortality risk related to ESKD [7], preventing DKD with intensive risk factor management is critical. In major clinical trials, the renoprotective effect of renin-angiotensin-aldosterone system (RAAS) inhibitors has been demonstrated in diabetic patients with hypertension [810]. However, for approximately 20 years after that, no single treatment that could delay the progression of DKD has been reported [11,12].
Sodium-glucose cotransporter 1 (SGLT1) is expressed mostly in the intestinal tract and less so in the kidney, whereas SGLT2 is predominantly present in the renal proximal tubule; both SGLTs are responsible for the reabsorption of glucose and sodium [13]. SGLT2 is responsible for 80% to 90% glucose reabsorption in the early proximal tubule, whereas SGLT1 reabsorbs the remaining 10% of the filtered glucose [14]. Some studies have indicated that the mRNA levels and activity of SGLTs are increased in patients with diabetes [15,16]. Therefore, strategies to inhibit SGLT2 have been proposed to lower blood sugar levels in diabetic patients [17,18], and several SGLT2 inhibitors have been introduced. In addition to their glucose-lowering effect, SGLT2 inhibitors have beneficial effects on cardiovascular and renal disease risk factors, such as weight loss and blood pressure reduction [19,20]. Indeed, cardiovascular and dedicated renal outcome trials of SGLT2 inhibitors have demonstrated that these drugs inhibit and delay the development and progression of cardiovascular and renal diseases [2123].
A single effect cannot explain the mechanisms underlying the cardiovascular and renal benefits of SGLT2 inhibitors. Instead, various metabolic and hemodynamic effects appear to be related, and the mechanisms by which SGLT2 inhibitors exert beneficial effects on the renal and cardiovascular systems have been reviewed elsewhere [24,25]. This review discusses the mechanism by which SGLT2 inhibitors exert a renoprotective effect. In particular, we summarize the results of recent studies focusing on changes in renal hemodynamics induced by SGLT2 inhibition.
Table 1 summarizes the renal effects of SGLT2 inhibitors in cardiovascular and renal outcome trials. The cardiovascular outcome trial of empagliflozin ((Empagliflozin) Cardiovascular Outcome Event Trial in Type 2 Diabetes Mellitus Patients [EMPA REG OUTCOME]) demonstrated a significant reduction in the incidence of major adverse cardiovascular events with empagliflozin versus placebo in patients with type 2 diabetes mellitus and established cardiovascular disease (CVD) [26]. In this study, pre-specified renal endpoints, incident or worsening nephropathy (progression to macroalbuminuria, doubling of serum creatinine, initiation of renal replacement therapy, or death from renal disease) also significantly decreased by 39% in the empagliflozin group [27]. Thereafter, in the Canagliflozin Cardiovascular Assessment Study (CANVAS) of canagliflozin and the Dapagliflozin Effect on Cardiovascular Events (DECLARE) trial of dapagliflozin, the risk for prespecified renal endpoints was also significantly reduced in the treatment group compared to placebo by 30% and 39%, respectively [28,29]. Although these studies have limitations in that kidney outcomes were measured as a secondary endpoint, the results exceeded the magnitude of the beneficial effects of angiotensin receptor blockers on the progression of DKD, which was demonstrated by the Reduction of Endpoints in NIDDM with the Angiotensin II Antagonist Losartan Study (RENAAL) and Irbesartan Diabetic Nephropathy Trial (IDNT) (16% and 19%, respectively) [8,10]. Subsequent dedicated renal outcome trials, namely the Canagliflozin and Renal Events in Diabetes with Established Nephropathy Clinical Evaluation (CREDENCE) and Dapagliflozin And Prevention of Adverse outcomes in Chronic Kidney Disease (DAPA CKD) studies, confirmed the effects of canagliflozin and dapagliflozin on primary renal endpoints in advanced CKD patients [10,30]; the risk of renal endpoints was significantly reduced in the CREDENCE study by 30%, and in the DAPA CKD study by 39%. In particular, the DAPA CKD study was conducted in patients with advanced CKD with or without diabetes, and the renoprotective effect of dapagliflozin was observed in both diabetic and non-diabetic patients [31].
The cardiovascular and renal outcome trials described above have consistently shown favorable results for kidney outcomes of SGLT2 inhibitors; therefore, it can be said that SGLT2 inhibitors have a protective role in the DKD progression as a class effect. The results of meta-analysis and subgroup analyses of these studies have several important clinical implications. First, the renal benefit of SGLT2 inhibitors is consistent regardless of CKD stage and albuminuria [21,22,29,30,32,33]. Notably, a similar renal benefit was seen even in patients with stage 1 or 2 CKD or normoalbuminuria as in those with higher stages of CKD or increased albuminuria. The absolute risk reduction for adverse renal events was even greater in patients with normal estimated glomerular filtration rate (eGFR) than in patients with advanced CKD [21,22]. This suggests that SGLT2 inhibitors can be applied at an earlier stage of DKD, significantly affecting early changes in the diabetic kidney, including glomerular hyperfiltration. Second, an initial decline in eGFR (initial “dip”) was observed after SGLT2 inhibitor use in most trials [34,35]. This pattern is similar to the transient eGFR decrease often observed after RAAS inhibitor use, suggesting significant changes in renal hemodynamics after SGLT2 inhibitor treatment initiation. As described later, decreased renal blood flow or renal vascular resistance appears to be responsible for this phenomenon. The initial dip in eGFR is a reversible process. It has been reported that the degree of decline is associated with subsequent kidney function preservation in some [34,36], but not in all trials [37]. In addition, many clinical trials have reported a low risk of acute kidney injury related to SGLT2 inhibitors [22,38], which indirectly shows that the initial dip occurs within a physiological range. Third, it has been demonstrated that SGLT2 inhibitors have beneficial effects not only on renal outcomes but also on heart failure outcomes [3941]. Given that both CKD and heart failure have reduced effective circulating volume, it has been suggested that SGLT2 inhibitors can induce significant and beneficial changes in systemic hemodynamics [42].
The mechanism by which glomerular hyperfiltration occurs in patients with diabetes is not fully understood, but the structural, vascular, and tubular changes associated with hyperglycemia and diabetes seem to be involved [43]. In addition, changes in various growth factors and cytokines in response to hyperglycemia and hyperinsulinemia induce renal hypertrophy [44,45], which is recognized as a causative factor for the occurrence of hyperfiltration.
Hyperfiltration is generally regarded as an early hemodynamic change in patients with diabetes [46] and is found in up to 70% of patients with type 1 and type 2 diabetes mellitus [43]. Afferent renal vasodilation, subsequent glomerular hypertension, and an increased filtration fraction (the ratio between GFR and effective renal plasma flow) have been observed in patients with early diabetes and experimental models of diabetes [47,48]. Tubuloglomerular feedback (TGF) is an important mechanism explaining this phenomenon [49]. During intravascular volume contraction or decreased circulating plasma volume, sodium delivery to the macular densa decreases. This leads to vasoconstriction of the efferent arteriole by renin/angiotensin release and vasodilation of the afferent arteriole by reduced adenosine formation. As a result, intraglomerular pressure is restored, and GFR increases. Therefore, sodium delivery to the distal nephron and the subsequent changes in renin, angiotensin, and adenosine production is central to the TGF model [50].
Under physiologic condition, glucose filtered from the glomerulus is reabsorbed almost completely in the proximal tubule [14]. In patients with diabetes, glucose filtration is increased by hyperglycemia, leading to increased sodium and glucose reabsorption via SGLT upregulation [51]. Increased SGLT1 and SGLT2 expression have been observed in diabetic mouse models and human studies [15,16]. This is supported by the fact that the threshold of glucosuria in diabetic patients is approximately 60 mg/dL higher than that in normal subjects [52]. Consequently, the amount of sodium reaching the macular densa at the juxtamedullary apparatus decreases, which is recognized as a decrease in the effective circulating volume [53,54]. This induces changes in glomerular capillaries, leading to hyperfiltration and glomerular hypertension [55]. In detail, when sodium is transferred to the tubular epithelial cells of the macular densa, the conversion of adenosine triphosphate (ATP) to adenosine diphosphate by the Na+/K+-ATPase decreases, and consequently, the production of adenosine decreases. Adenosine induces arteriolar vasoconstriction through the adenosine type 1 (A1) receptor at the afferent arteriole. Therefore, a decrease in adenosine production inhibits this process and causes relative afferent arteriolar vasodilation. In contrast, a reduction in sodium delivery to the distal tubule in the diabetic kidney induces intrarenal RAAS activation, which results in efferent arteriolar vasoconstriction [55,56]. Taken together, the effects of hyperglycemia in the diabetic kidney influence both afferent and efferent arterioles, resulting in intraglomerular hypertension and hyperfiltration.
According to clinical studies, SGLT2 inhibitors reduce glycosylated hemoglobin by 0.4% to 1.0% in patients with type 2 diabetes mellitus under various trial settings [57]. In addition, SGLT2 inhibitors also control risk factors for CVD and CKD by reducing systolic blood pressure by 2 to 5 mm Hg and body weight by 2 to 4 kg [19,20]. However, compared to glucagon-like peptide-1 receptor agonists, which had a greater effect on reduction in glycosylated hemoglobin and weight loss, SGLT2 inhibitors showed superiority in ESKD prevention [57,58]. This finding implies that the effects of SGLT2 inhibitors on metabolic changes and cardiovascular risk factors do not fully explain their renoprotective effects.
TGF restoration is an important mechanism for explaining renal hemodynamic changes induced by SGLT2 inhibitors. SGLT2 inhibitors normalize the decreased distal sodium delivery to the macular densa in the diabetic kidney by inhibiting sodium reabsorption in the proximal tubule [56]. This increases adenosine production and consequently induces vasoconstriction of the afferent arteriole. As a result, the intraglomerular pressure is reduced, and hyperfiltration is improved. This has been demonstrated in several animal studies and patients with type 1 diabetes mellitus. In streptozotocin-induced diabetic rats, dapagliflozin decreased single-nephron GFR, accompanied by decreased proximal solute reabsorption and increased distal tubule chloride [59]. In patients with type 1 diabetes mellitus with hyperfiltration, empagliflozin treatment significantly reduced renal blood flow and hyperfiltration, accompanied by a decrease in plasma nitric oxide and an increase in renal vascular resistance [60]. In an animal model of type 1 diabetes mellitus, in vivo imaging demonstrated that empagliflozin reduced hyperfiltration via afferent arteriole constriction. In this study, an A1 adenosine receptor blocker counteracted the action of empagliflozin, supporting the hypothesis that SGLT2 inhibition affects renal hemodynamic function via adenosine production [61]. These studies showed that TGF restoration by SGLT2 inhibition does work, but there are limitations in that the studies were conducted primarily under the conditions of type 1 diabetes mellitus and hyperfiltration.
Of note, most large-scaled renal outcome trials have demonstrated the renoprotective effects of SGLT2 inhibitors in patients with type 2 diabetes mellitus and advanced stage of CKD. A possible explanation for the role of hyperfiltration in patients with advanced CKD is that hyperfiltration still occurs at the single nephron level in the late stage of kidney disease progression [62]. This is considered a compensatory mechanism for the reduced nephron number [63]. On the other hand, obesity, which is frequently found in patients with type 2 diabetes mellitus, also contributes to increased tubular sodium reabsorption by increasing abdominal pressure and compression of loops of Henle [64,65]. Therefore, even in advanced CKD, SGLT2 inhibitors may show hemodynamic effects similar to those observed in early CKD stages.
A recent study on patients with type 2 diabetes mellitus provided new insights into the hemodynamic effects of SGLT2 inhibitors [66]. In this study of 44 patients with type 2 diabetes mellitus, dapagliflozin decreased the measured GFR, and filtration fraction compared to gliclazide but did not increase renal vascular resistance. In addition, dapagliflozin treatment increased urinary adenosine and prostaglandin concentrations. Adenosine, a vasoconstrictor in the afferent arteriole, had a conflicting effect (vasodilation) on the efferent arteriole in the presence of RAAS blockade [67]. This means that SGLT2 inhibitors have different renal hemodynamic effects in type 2 diabetes mellitus patients on top of RAAS blockers compared to type 1 diabetes mellitus patients. In other words, efferent arteriole vasodilation through prostaglandin release could be a novel mechanistic explanation. The patients in this study had a baseline mean eGFR of 85 mL/min/1.73 m2 and diabetes duration of 9.8 years, and about 70% of participants were treated with RAAS inhibitors, which suggested that SGLT2 inhibitors may have a different renohemodynamic effect in patients with advanced type 2 diabetes mellitus without hyperfiltration. Another study, including 101 patients with type 2 diabetes mellitus, showed that treatment with empagliflozin and linagliptin versus metformin and insulin glargine did not increase the resistance of afferent arterioles. Still, it decreased efferent arterioles’ resistance and was accompanied by reduced GFR [68]. Table 2, Fig. 1 summarizes renal hemodynamic changes following SGLT2 inhibition.
Recent evidence clearly shows that SGLT2 inhibitors prevent or delay DKD progression. The renoprotective effects of SGLT2 inhibitors appear throughout the course of CKD, from the early to the late stages of the disease. In type 1 diabetes mellitus or under hyperfiltration conditions, restoration of TGF induced by SGLT2 inhibition reduces glomerular hypertension via afferent arteriole vasoconstriction. Recent studies have suggested that the renal hemodynamic effect of SGLT2 inhibitors in older patients with type 2 diabetes mellitus may differ from that in type 1 diabetes mellitus or under hyperfiltration conditions. However, although the route is likely to differ, intraglomerular pressure reduction by SGLT2 inhibition has been consistently observed. This change in renal hemodynamics is one of the important mechanisms explaining the renoprotective effect of SGLT2 inhibitors. It is expected that more evidence in the future will provide a better understanding of the true renal effects of SGLT2 inhibitors.

CONFLICTS OF INTEREST

No potential conflict of interest relevant to this article was reported.

FUNDING

This study was supported by the National Research Foundation of Korea (NRF) (Grant No. 2021R1C1C1005674).

Acknowledgements
None
Fig. 1
Hemodynamic changes following sodium-glucose cotransporter 2 (SGLT2) inhibition in the kidney. ① SGLT2 is responsible for 80% to 90% glucose reabsorption in the early proximal tubule, and SGLT1 reabsorbs the remaining 10% of the filtered glucose under physiologic conditions. ② Under hyperglycemic condition, glucose filtration is increased, which leading to increased sodium and glucose reabsorption via SGLT2 upregulation. SGLT2 inhibitor primarily blocks the action of SGLT2, ③ resulting in increased delivery of sodium and glucose to the distal renal tubule. ④ In type 1 diabetes mellitus or hyperfiltration state, increased sodium delivery to the tubular epithelial cells of the macular densa induces adenosine production which activates adenosine A1 receptor, triggering an increase in cytosolic Ca2+. ⑤ Restoration of tubuloglomerular feedback ultimately results in afferent arteriolar vasoconstriction. ⑥ In type 2 diabetes mellitus on renin-angiotensin-aldosterone system (RAAS) blockade, SGLT2 inhibition may act on renal arterioles in a different way. SGLT2 inhibitors increased production of adenosine and prostaglandin ⑦ which resulting in efferent vasodilation on RAAS blockade.
dmj-2022-0209f1.jpg
dmj-2022-0209f2.jpg
Table 1
Summary of renal effects of SGLT2 inhibitors in the cardiovascular and renal outcome trials
Renal endpoint No. of participants Population Mean eGFR, mL/min/1.73 m2 Renal endpoint HRa of composite renal endpoint HRa of ESKD
EMPA REG OUTCOME (Empagliflozin) Secondary 7,020 T2DM and ASCVD 74.2 Macroalbuminuria, doubling of sCr with eGFR <45 mL/min/1.73 m2, initiation of RRT, death from renal disease 0.61 (0.53–0.70) 0.45 (0.21–0.97)
ANVAS (Canagliflozin) Exploratory 10,142 T2DM, ASCVD or MRF 76.7 ≥40% decrease in eGFR, need for RRT, death from renal cause 0.60 (0.47–0.77) 0.77 (0.30–1.97)
DECLARE (Dapagliflozin) Secondary 17,160 T2DM, ASCVD or MRF 85.4 ≥40% decrease in eGFR to <60 mL/min/1.73 m2, ESKD, death from renal or cardiovascular causes 0.76 (0.67–0.87) 0.31 (0.13–0.79)
CREDENCE (Canagliflozin) Primary 4,401 T2DM, CKD and macroalbuminuria 56.3 Doubling of sCr, ESKD, death from renal or cardiovascular causes 0.70 (0.59–0.82) 0.68 (0.54–0.86)
DAPA CKD (Dapagliflozin) Primary 4,304 With or without T2DM, CKD and albuminuria 43.2 ≥50% decrease in eGFR, ESKD, death from renal or cardiovascular causes 0.61 (0.51–0.72) 0.64 (0.50–0.82)
VERTIS CV (Ertugliflozin) Secondary 8,246 T2DM and ASCVD 76.1 Doubling of sCr, RRT, death from renal causes 0.81 (0.63–1.04) NA
DAPA HF (Dapagliflozin) Secondary 4,744 With or without T2DM, EF ≤40% 66.0 ≥50% decrease in eGFR, ESKD, death from renal causes 0.71 (0.44–1.16) 1.00 (0.50–1.99)
EMPEROR- Reduced (Empagliflozin) Prespecified 3,730 With or without T2DM, EF ≤40% 61.8 ≥40% decrease in eGFR, ESKD 0.50 (0.32–0.77) NA
SCORED (Sotagliflozin) Secondary 10,584 T2DM, CKD, MRF 44.4 ≥50% decrease in eGFR, ESKD 0.71 (0.46–1.08) NA

SGLT2, sodium-glucose cotransporter 2; eGFR, estimated glomerular filtration rate; HR, hazard ratio; ESKD, end-stage kidney disease; EMPA REG OUTCOME, (Empagliflozin) Cardiovascular Outcome Event Trial in Type 2 Diabetes Mellitus Patients; T2DM, type 2 diabetes mellitus; ASCVD, atherosclerotic cardiovascular disease; sCr, serum creatinine; RRT, renal replacement therapy; CANVAS, Canagliflozin Cardiovascular Assessment Study; MRF, multiple risk factors for ASCVD; DECLARE, Dapagliflozin Effect on Cardiovascular Events; CREDENCE, Canagliflozin and Renal Events in Diabetes with Established Nephropathy Clinical Evaluation; CKD, chronic kidney disease; DAPA CKD, Dapagliflozin And Prevention of Adverse outcomes in Chronic Kidney Disease; VERTIS CV, Evaluation of ertugliflozin efficacy and safety cardiovascular outcomes trial; NA, not available; DAPA HF, dapagliflozin and prevention of adverse outcomes in heart failure; EF, ejection fraction; EMPEROR-Reduced, empagliflozin outcome trial in patients with chronic heart failure and a reduced ejection fraction; SCORED, effect of sotagliflozin on cardiovascular and renal events in patients with type 2 diabetes and moderate renal impairment who are at cardiovascular risk.

a Study drug (SGLT2 inhibitors) vs. placebo.

Table 2
Summary of the effects of SGLT2 inhibitors on kidney in diabetes
Diabetes Diabetes with SGLT2 inhibitors
SGLT2 expression
Na+ reabsorption at proximal tubule
Na+ level at macula densa
Adenosine level at macula densa

Vasodilation
Afferent arteriole tone
Vasoconstriction

Vasoconstriction
Efferent arteriole tone
Vasodilation
Intraglomerular pressure
GFR (early change)

SGLT2, sodium-glucose cotransporter 2; GFR, glomerular filtration rate.

  • 1. Atkins RC. The epidemiology of chronic kidney disease. Kidney Int Suppl 2005;94:S14-8.Article
  • 2. The Korean Society of Nephrology: Trends in epidemiologic characteristics of end-stage renal disease from 2020 KORDS (Korean Renal Data System); Available from: https://ksn.or.kr/(cited 2022 Jul 15).
  • 3. Ahn JH, Yu JH, Ko SH, Kwon HS, Kim DJ, Kim JH, et al. Prevalence and determinants of diabetic nephropathy in Korea: Korea National Health and Nutrition Examination Survey. Diabetes Metab J 2014;38:109-19.ArticlePubMedPMC
  • 4. Kim KJ, Kwon TY, Yu S, Seo JA, Kim NH, Choi KM, et al. Ten-year mortality trends for adults with and without diabetes mellitus in South Korea, 2003 to 2013. Diabetes Metab J 2018;42:394-401.ArticlePubMedPMCPDF
  • 5. Lee MJ, Ha KH, Kim DJ, Park I. Trends in the incidence, prevalence, and mortality of end-stage kidney disease in South Korea. Diabetes Metab J 2020;44:933-7.ArticlePubMedPMCPDF
  • 6. Park JH, Ha KH, Kim BY, Lee JH, Kim DJ. Trends in cardiovascular complications and mortality among patients with diabetes in South Korea. Diabetes Metab J 2021;45:120-4.ArticlePubMedPMCPDF
  • 7. Reutens AT. Epidemiology of diabetic kidney disease. Med Clin North Am 2013;97:1-18.ArticlePubMed
  • 8. Brenner BM, Cooper ME, de Zeeuw D, Keane WF, Mitch WE, Parving HH, et al. Effects of losartan on renal and cardiovascular outcomes in patients with type 2 diabetes and nephropathy. N Engl J Med 2001;345:861-9.ArticlePubMed
  • 9. Heart outcomes prevention evaluation study investigators. Effects of ramipril on cardiovascular and microvascular outcomes in people with diabetes mellitus: results of the HOPE study and MICRO-HOPE substudy. Lancet 2000;355:253-9.ArticlePubMed
  • 10. Lewis EJ, Hunsicker LG, Clarke WR, Berl T, Pohl MA, Lewis JB, et al. Renoprotective effect of the angiotensin-receptor antagonist irbesartan in patients with nephropathy due to type 2 diabetes. N Engl J Med 2001;345:851-60.ArticlePubMed
  • 11. Sawaf H, Thomas G, Taliercio JJ, Nakhoul G, Vachharajani TJ, Mehdi A. Therapeutic advances in diabetic nephropathy. J Clin Med 2022;11:378.ArticlePubMedPMC
  • 12. Yamazaki T, Mimura I, Tanaka T, Nangaku M. Treatment of diabetic kidney disease: current and future. Diabetes Metab J 2021;45:11-26.ArticlePubMedPMCPDF
  • 13. Coady MJ, Wallendorff B, Lapointe JY. Characterization of the transport activity of SGLT2/MAP17, the renal low-affinity Na+-glucose cotransporter. Am J Physiol Renal Physiol 2017;313:F467-74.ArticlePubMed
  • 14. Wright EM, Hirayama BA, Loo DF. Active sugar transport in health and disease. J Intern Med 2007;261:32-43.ArticlePubMed
  • 15. Tabatabai NM, Sharma M, Blumenthal SS, Petering DH. Enhanced expressions of sodium-glucose cotransporters in the kidneys of diabetic Zucker rats. Diabetes Res Clin Pract 2009;83:e27-30.ArticlePubMedPMC
  • 16. Norton L, Shannon CE, Fourcaudot M, Hu C, Wang N, Ren W, et al. Sodium-glucose co-transporter (SGLT) and glucose transporter (GLUT) expression in the kidney of type 2 diabetic subjects. Diabetes Obes Metab 2017;19:1322-6.PubMedPDF
  • 17. Abdul-Ghani MA, DeFronzo RA. Inhibition of renal glucose reabsorption: a novel strategy for achieving glucose control in type 2 diabetes mellitus. Endocr Pract 2008;14:782-90.ArticlePubMed
  • 18. Tsujihara K, Hongu M, Saito K, Inamasu M, Arakawa K, Oku A, et al. Na(+)-glucose cotransporter inhibitors as antidiabetics: I. synthesis and pharmacological properties of 4′-dehydroxyphlorizin derivatives based on a new concept. Chem Pharm Bull (Tokyo) 1996;44:1174-80.ArticlePubMed
  • 19. Abdul-Ghani M, Del Prato S, Chilton R, DeFronzo RA. SGLT2 inhibitors and cardiovascular risk: lessons learned from the EMPA-REG outcome study. Diabetes Care 2016;39:717-25.ArticlePubMedPMCPDF
  • 20. Cowie MR, Fisher M. SGLT2 inhibitors: mechanisms of cardiovascular benefit beyond glycaemic control. Nat Rev Cardiol 2020;17:761-72.ArticlePubMedPDF
  • 21. Zelniker TA, Wiviott SD, Raz I, Im K, Goodrich EL, Bonaca MP, et al. 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 2019;393:31-9.ArticlePubMed
  • 22. Neuen BL, Young T, Heerspink HJ, Neal B, Perkovic V, Billot L, et al. SGLT2 inhibitors for the prevention of kidney failure in patients with type 2 diabetes: a systematic review and meta-analysis. Lancet Diabetes Endocrinol 2019;7:845-54.ArticlePubMed
  • 23. Kim HJ, Kim SS, Song SH. Glomerular filtration rate as a kidney outcome of diabetic kidney disease: a focus on new antidiabetic drugs. Korean J Intern Med 2022;37:502-19.ArticlePubMedPMCPDF
  • 24. Sen T, Heerspink HJ. A kidney perspective on the mechanism of action of sodium glucose co-transporter 2 inhibitors. Cell Metab 2021;33:732-9.ArticlePubMed
  • 25. DeFronzo RA, Reeves WB, Awad AS. Pathophysiology of diabetic kidney disease: impact of SGLT2 inhibitors. Nat Rev Nephrol 2021;17:319-34.ArticlePubMedPDF
  • 26. Zinman B, Wanner C, Lachin JM, Fitchett D, Bluhmki E, Hantel S, et al. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med 2015;373:2117-28.ArticlePubMed
  • 27. Wanner C, Inzucchi SE, Lachin JM, Fitchett D, von Eynatten M, Mattheus M, et al. Empagliflozin and progression of kidney disease in type 2 diabetes. N Engl J Med 2016;375:323-34.ArticlePubMed
  • 28. Perkovic V, de Zeeuw D, Mahaffey KW, Fulcher G, Erondu N, Shaw W, et al. Canagliflozin and renal outcomes in type 2 diabetes: results from the CANVAS program randomised clinical trials. Lancet Diabetes Endocrinol 2018;6:691-704.ArticlePubMed
  • 29. Mosenzon O, Wiviott SD, Cahn A, Rozenberg A, Yanuv I, Goodrich EL, et al. Effects of dapagliflozin on development and progression of kidney disease in patients with type 2 diabetes: an analysis from the DECLARE-TIMI 58 randomised trial. Lancet Diabetes Endocrinol 2019;7:606-17.ArticlePubMed
  • 30. Heerspink HJL, Stefansson BV, Correa-Rotter R, Chertow GM, Greene T, Hou FF, et al. Dapagliflozin in patients with chronic kidney disease. N Engl J Med 2020;383:1436-46.ArticlePubMed
  • 31. Wheeler DC, Stefansson BV, Jongs N, Chertow GM, Greene T, Hou FF, et al. Effects of dapagliflozin on major adverse kidney and cardiovascular events in patients with diabetic and non-diabetic chronic kidney disease: a prespecified analysis from the DAPA-CKD trial. Lancet Diabetes Endocrinol 2021;9:22-31.ArticlePubMed
  • 32. Wanner C, Heerspink HJL, Zinman B, Inzucchi SE, Koitka-Weber A, Mattheus M, et al. Empagliflozin and kidney function decline in patients with type 2 diabetes: a slope analysis from the EMPA-REG outcome trial. J Am Soc Nephrol 2018;29:2755-69.ArticlePubMedPMC
  • 33. Levin A, Perkovic V, Wheeler DC, Hantel S, George JT, von Eynatten M, et al. Empagliflozin and cardiovascular and kidney outcomes across KDIGO risk categories: post hoc analysis of a randomized, double-blind, placebo-controlled, multinational trial. Clin J Am Soc Nephrol 2020;15:1433-44.PubMedPMC
  • 34. Heerspink HJ, Cherney DZ. Clinical implications of an acute dip in eGFR after SGLT2 inhibitor initiation. Clin J Am Soc Nephrol 2021;16:1278-80.ArticlePubMedPMC
  • 35. Oh TJ, Moon JY, Hur KY, Ko SH, Kim HJ, Kim T, et al. Sodium-glucose cotransporter-2 inhibitor for renal function preservation in patients with type 2 diabetes mellitus: a Korean Diabetes Association and Korean Society of Nephrology consensus statement. Diabetes Metab J 2020;44:489-97.ArticlePubMedPMCPDF
  • 36. Cherney DZI, Cosentino F, Dagogo-Jack S, McGuire DK, Pratley RE, Frederich R, et al. Initial eGFR changes with ertugliflozin and associations with clinical parameters: analyses from the VERTIS CV trial. Am J Nephrol 2022;1-10.ArticlePubMedPDF
  • 37. Oshima M, Jardine MJ, Agarwal R, Bakris G, Cannon CP, Charytan DM, et al. Insights from CREDENCE trial indicate an acute drop in estimated glomerular filtration rate during treatment with canagliflozin with implications for clinical practice. Kidney Int 2021;99:999-1009.ArticlePubMed
  • 38. Menne J, Dumann E, Haller H, Schmidt BM. Acute kidney injury and adverse renal events in patients receiving SGLT2-inhibitors: a systematic review and meta-analysis. PLoS Med 2019;16:e1002983.ArticlePubMedPMC
  • 39. McMurray JJ, Solomon SD, Inzucchi SE, Kober L, Kosiborod MN, Martinez FA, et al. Dapagliflozin in patients with heart failure and reduced ejection fraction. N Engl J Med 2019;381:1995-2008.PubMed
  • 40. Packer M, Anker SD, Butler J, Filippatos G, Pocock SJ, Carson P, et al. Cardiovascular and renal outcomes with empagliflozin in heart failure. N Engl J Med 2020;383:1413-24.PubMed
  • 41. Anker SD, Butler J, Filippatos G, Ferreira JP, Bocchi E, Bohm M, et al. Empagliflozin in heart failure with a preserved ejection fraction. N Engl J Med 2021;385:1451-61.PubMed
  • 42. van der Aart-van der Beek AB, de Boer RA, Heerspink HJ. Kidney and heart failure outcomes associated with SGLT2 inhibitor use. Nat Rev Nephrol 2022;18:294-306.ArticlePubMedPDF
  • 43. Tonneijck L, Muskiet MH, Smits MM, van Bommel EJ, Heerspink HJ, van Raalte DH, et al. Glomerular hyperfiltration in diabetes: mechanisms, clinical significance, and treatment. J Am Soc Nephrol 2017;28:1023-39.ArticlePubMedPMC
  • 44. Bak M, Thomsen K, Christiansen T, Flyvbjerg A. Renal enlargement precedes renal hyperfiltration in early experimental diabetes in rats. J Am Soc Nephrol 2000;11:1287-92.ArticlePubMed
  • 45. Jung CY, Yoo TH. Pathophysiologic mechanisms and potential biomarkers in diabetic kidney disease. Diabetes Metab J 2022;46:181-97.ArticlePubMedPMCPDF
  • 46. Jerums G, Premaratne E, Panagiotopoulos S, MacIsaac RJ. The clinical significance of hyperfiltration in diabetes. Diabetologia 2010;53:2093-104.ArticlePubMedPDF
  • 47. Vora JP, Dolben J, Dean JD, Thomas D, Williams JD, Owens DR, et al. Renal hemodynamics in newly presenting non-insulin dependent diabetes mellitus. Kidney Int 1992;41:829-35.ArticlePubMed
  • 48. Keller CK, Bergis KH, Fliser D, Ritz E. Renal findings in patients with short-term type 2 diabetes. J Am Soc Nephrol 1996;7:2627-35.ArticlePubMed
  • 49. Thomson SC, Blantz RC. Glomerulotubular balance, tubuloglomerular feedback, and salt homeostasis. J Am Soc Nephrol 2008;19:2272-5.ArticlePubMed
  • 50. Vallon V. Tubuloglomerular feedback and the control of glomerular filtration rate. News Physiol Sci 2003;18:169-74.ArticlePubMed
  • 51. Bank N, Aynedjian HS. Progressive increases in luminal glucose stimulate proximal sodium absorption in normal and diabetic rats. J Clin Invest 1990;86:309-16.ArticlePubMedPMC
  • 52. Ferrannini E, Veltkamp SA, Smulders RA, Kadokura T. Renal glucose handling: impact of chronic kidney disease and sodium-glucose cotransporter 2 inhibition in patients with type 2 diabetes. Diabetes Care 2013;36:1260-5.PubMedPMC
  • 53. Vervoort G, Veldman B, Berden JH, Smits P, Wetzels JF. Glomerular hyperfiltration in type 1 diabetes mellitus results from primary changes in proximal tubular sodium handling without changes in volume expansion. Eur J Clin Invest 2005;35:330-6.ArticlePubMed
  • 54. Pruijm M, Wuerzner G, Maillard M, Bovet P, Renaud C, Bochud M, et al. Glomerular hyperfiltration and increased proximal sodium reabsorption in subjects with type 2 diabetes or impaired fasting glucose in a population of the African region. Nephrol Dial Transplant 2010;25:2225-31.ArticlePubMed
  • 55. 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 1999;10:2569-76.PubMed
  • 56. Vallon V, Thomson SC. Renal function in diabetic disease models: the tubular system in the pathophysiology of the diabetic kidney. Annu Rev Physiol 2012;74:351-75.ArticlePubMedPMC
  • 57. Hsia DS, Grove O, Cefalu WT. An update on sodium-glucose co-transporter-2 inhibitors for the treatment of diabetes mellitus. Curr Opin Endocrinol Diabetes Obes 2017;24:73-9.ArticlePubMedPMC
  • 58. Zelniker TA, Wiviott SD, Raz I, Im K, Goodrich EL, Furtado RHM, et al. Comparison of the effects of glucagon-like peptide receptor agonists and sodium-glucose cotransporter 2 inhibitors for prevention of major adverse cardiovascular and renal outcomes in type 2 diabetes mellitus. Circulation 2019;139:2022-31.ArticlePubMed
  • 59. Thomson SC, Rieg T, Miracle C, Mansoury H, Whaley J, Vallon V, et al. Acute and chronic effects of SGLT2 blockade on glomerular and tubular function in the early diabetic rat. Am J Physiol Regul Integr Comp Physiol 2012;302:R75-83.ArticlePubMedPMC
  • 60. Cherney DZ, Perkins BA, Soleymanlou N, Maione M, Lai V, Lee A, et al. Renal hemodynamic effect of sodium-glucose cotransporter 2 inhibition in patients with type 1 diabetes mellitus. Circulation 2014;129:587-97.ArticlePubMed
  • 61. Kidokoro K, Cherney DZ, Bozovic A, Nagasu H, Satoh M, Kanda E, et al. Evaluation of glomerular hemodynamic function by empagliflozin in diabetic mice using in vivo imaging. Circulation 2019;140:303-15.ArticlePubMed
  • 62. Brenner BM, Lawler EV, Mackenzie HS. The hyperfiltration theory: a paradigm shift in nephrology. Kidney Int 1996;49:1774-7.ArticlePubMed
  • 63. Hostetter TH, Olson JL, Rennke HG, Venkatachalam MA, Brenner BM. Hyperfiltration in remnant nephrons: a potentially adverse response to renal ablation. Am J Physiol 1981;241:F85-93.ArticlePubMed
  • 64. Alonso-Galicia M, Dwyer TM, Herrera GA, Hall JE. Increased hyaluronic acid in the inner renal medulla of obese dogs. Hypertension 1995;25(4 Pt 2):888-92.ArticlePubMed
  • 65. Henegar JR, Bigler SA, Henegar LK, Tyagi SC, Hall JE. Functional and structural changes in the kidney in the early stages of obesity. J Am Soc Nephrol 2001;12:1211-7.ArticlePubMed
  • 66. van Bommel EJ, Muskiet MH, van Baar MJ, Tonneijck L, Smits MM, Emanuel AL, et al. 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 2020;97:202-12.ArticlePubMed
  • 67. Vallon V, Muhlbauer B, Osswald H. Adenosine and kidney function. Physiol Rev 2006;86:901-40.ArticlePubMed
  • 68. Ott C, Kannenkeril D, Jung S, Schnieder R. Combination therapy of empagliflozin and linagliptin vs. metformin and insulin glargine on intra- and renal hemodynamics in type 2 diabetes. ASN Kidney Week 2019 Nov 5 -10. Washington, DC: Available from: https://www.asn-online.org/education/kidneyweek/2019/program-abstract.aspx?controlId=3233843 .

Figure & Data

References

    Citations

    Citations to this article as recorded by  
    • Using intravoxel incoherent motion imaging to evaluate uric acid-induced renal injury and efficacy after treatment
      Zhong-Yuan Cheng, Shang-Ao Gong, Ping-Kang Chen, Zong-Chao Yu, Chen Qiu, Ji-Xin Lin, Jia-Bin Mo, Long Qian, You-Zhen Feng, Xiang-Ran Cai
      British Journal of Radiology.2024; 97(1153): 274.     CrossRef
    • Rethinking eGFR Comparisons in SGLT2 Inhibitor Research
      Yuzuru Ohshiro
      Journal of the American College of Cardiology.2024; 83(9): e87.     CrossRef
    • Baseline eGFR, albuminuria and renal outcomes in patients with SGLT2 inhibitor treatment: an updated meta-analysis
      Yunke Ma, Chu Lin, Xiaoling Cai, Suiyuan Hu, Xingyun Zhu, Fang Lv, Wenjia Yang, Linong Ji
      Acta Diabetologica.2023; 60(3): 435.     CrossRef
    • Effects of sodium-glucose cotransporter 2 inhibitors on renal risk factors in patients with abnormal glucose metabolism: a meta-analysis of randomized controlled trials
      Mengnan Li, Jian Zhang, Guimei Yang, Jiaxin Zhang, Minmin Han, Yi Zhang, Yunfeng Liu
      European Journal of Clinical Pharmacology.2023; 79(6): 859.     CrossRef
    • Age at Mortality in Patients with Type 2 Diabetes Who Underwent Kidney Transplantation: An Analysis of Data from the Korean National Health Insurance and Statistical Information Service, 2006 to 2018
      Sun Ok Song, Eugene Han, Kang Ju Son, Bong-Soo Cha, Byung-Wan Lee
      Journal of Clinical Medicine.2023; 12(9): 3160.     CrossRef
    • Exposure–Response Analysis of the Sodium–Glucose Cotransporter-2 Inhibitors Dapagliflozin and Empagliflozin on Kidney Hemodynamics in Patients with Type 2 Diabetes
      Sjoukje van der Hoek, Jeroen V. Koomen, Erik J. M. van Bommel, Charlotte M. Mosterd, Rosalie A. Scholtes, Anne C. Hesp, Jasper Stevens, Daniel H. van Raalte, Hiddo J. L. Heerspink
      Journal of Personalized Medicine.2023; 13(5): 747.     CrossRef
    • Osteopontin as a Biomarker in Chronic Kidney Disease
      Satyesh K. Sinha, Michael Mellody, Maria Beatriz Carpio, Robert Damoiseaux, Susanne B. Nicholas
      Biomedicines.2023; 11(5): 1356.     CrossRef
    • Increased expression of sodium-glucose cotransporter 2 and O-GlcNAcylation in hepatocytes drives non-alcoholic steatohepatitis
      Hye Jin Chun, Eun Ran Kim, Minyoung Lee, Da Hyun Choi, Soo Hyun Kim, Eugene Shin, Jin-Hong Kim, Jin Won Cho, Dai Hoon Han, Bong-Soo Cha, Yong-ho Lee
      Metabolism.2023; 145: 155612.     CrossRef
    • Synthesis and biological profile of benzoxazolone derivatives
      Parteek Prasher, Tanisqa Mall, Mousmee Sharma
      Archiv der Pharmazie.2023;[Epub]     CrossRef
    • SGLT2 inhibitors prevent LPS-induced M1 macrophage polarization and alleviate inflammatory bowel disease by downregulating NHE1 expression
      Ye Jin Kim, Jonghwa Jin, Dong-Ho Kim, Daehoon Kim, You Mie Lee, Jun-Kyu Byun, Yeon-Kyung Choi, Keun-Gyu Park
      Inflammation Research.2023; 72(10-11): 1981.     CrossRef

    • PubReader PubReader
    • ePub LinkePub Link
    • Cite
      CITE
      export Copy
      Close
      Download Citation
      Download a citation file in RIS format that can be imported by all major citation management software, including EndNote, ProCite, RefWorks, and Reference Manager.

      Format:
      • RIS — For EndNote, ProCite, RefWorks, and most other reference management software
      • BibTeX — For JabRef, BibDesk, and other BibTeX-specific software
      Include:
      • Citation for the content below
      Renoprotective Mechanism of Sodium-Glucose Cotransporter 2 Inhibitors: Focusing on Renal Hemodynamics
      Diabetes Metab J. 2022;46(4):543-551.   Published online July 27, 2022
      Close
    • XML DownloadXML Download
    Figure
    Related articles

    Diabetes Metab J : Diabetes & Metabolism Journal