Acessibilidade / Reportar erro

SGLT2 inhibitors and NLRP3 inflammasome: potential target in diabetic kidney disease

Abstract

Diabetic kidney disease (DKD) remains the leading cause of chronic kidney disease (CKD) worldwide. The pathogenesis of DKD is influenced by functional, histopathological, and immune mechanisms, including NLRP3 inflammasome activity and oxidative stress. The sodium-glucose cotransporter 2 inhibitors (SGLT2i) have shown metabolic benefits and the ability to slow the progression of DKD in several clinical studies over the years. Recent studies suggest that the antidiabetic activity also extends to inhibition of the inflammatory response, including modulation of the NLRP3 inflammasome, reduction of pro-inflammatory markers and reduction of oxidative stress. Here we review the efficacy of SGLT2i in the treatment of CKD and discuss the role of the inflammatory response in the development of DKD, including its relationship to the NLRP3 inflammasome and oxidative stress.

Keywords:
Diabetic Nephropathies; Inflammatory Response; NLR Family; Pyrin Domain-Containing 3 Protein; Inflammasome; Sodium-Glucose Transporter-2 Inhibitors; Renal Insufficiency; Chronic

Resumo

A doença renal do diabetes (DRD) permanece como a principal causa de doença renal crônica (DRC) mundialmente. A patogênese da DRD é influenciada por mecanismos funcionais, histopatológicos e imunológicos, incluindo a atividade do inflamassoma NLRP3 e estresse oxidativo. Os inibidores do cotransportador de sódio-glicose 2 (iSGLT2) demonstraram benefícios metabólicos e a capacidade de retardar a progressão da DRD em diversos estudos clínicos ao longo dos anos. Estudos recentes sugerem que a atividade antidiabética também se estende à inibição da resposta inflamatória, incluindo modulação do inflamassoma NLRP3, redução de marcadores pró-inflamatórios e redução do estresse oxidativo. Aqui, avaliamos a eficácia dos iSGLT2 no tratamento da DRC e discutimos o papel da resposta inflamatória no desenvolvimento da DRD, incluindo sua relação com o inflamassoma NLRP3 e o estresse oxidativo.

Descritores:
Nefropatia Diabética; Resposta Inflamatória; Proteína 3 contendo domínio de pirina da família NLR; Inflamassoma; Inibidores do Transportador de Sódio-Glicose 2; Insuficiência Renal Crônica

Clinical Summary

  • Diabetic kidney disease is the leading cause of chronic kidney disease

  • The NLRP3 inflammasome is a key regulator of the inflammatory response in the development of diabetic kidney disease

  • SGLT2 inhibitors have shown the ability to slow diabetic kidney disease in several clinical trials

  • SGLT2 inhibitors may slow the progression of diabetic kidney disease by inhibiting the NLRP3 inflammasome

Chronic Kidney Disease: Definition and Epidemiology

Chronic kidney disease (CKD) is a public health problem with global impact, characterized by a progressive and irreversible loss of kidney function11. Go AS, Chertow GM, Fan D, McCulloch CE, Hsu CY. Chronic kidney disease and the risks of death, cardiovascular events, and hospitalization. N Engl J Med. 2004;351(13):1296–305. doi: http://doi.org/10.1056/NEJMoa041031. PubMed PMID: 15385656.
https://doi.org/10.1056/NEJMoa041031...
. In recent years, an increase in the incidence of CKD has been estimated as a result of demographic change, with CKD affecting approximately 9% of the world’s population22. GBD Chronic Kidney Disease Collaboration. Global, regional, and national burden of chronic kidney disease, 1990–2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet. 2020;395(10225):709–33. doi: http://doi.org/10.1016/S0140-6736(20)30045-3. PubMed PMID: 32061315.
https://doi.org/10.1016/S0140-6736(20)30...
. Crucial to its characterization is the time requirement, defined by the presence of damage (albuminuria ≥ 30 mg/day) and/or a glomerular filtration rate < 60 mL/min/1.73 m2 for a period greater than or equal to 3 months with progressive, irreversible, and usually asymptomatic loss of kidney function33. Webster AC, Nagler EV, Morton RL, Masson P. Chronic Kidney Disease. Lancet. 2017;389(10075):1238–52. doi: http://doi.org/10.1016/S0140-6736(16)32064-5. PubMed PMID: 27887750.
https://doi.org/10.1016/S0140-6736(16)32...
.

CKD has a multifactorial etiology and its main causes are diabetes mellitus (DM), systemic arterial hypertension (SAH), obesity and a family history22. GBD Chronic Kidney Disease Collaboration. Global, regional, and national burden of chronic kidney disease, 1990–2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet. 2020;395(10225):709–33. doi: http://doi.org/10.1016/S0140-6736(20)30045-3. PubMed PMID: 32061315.
https://doi.org/10.1016/S0140-6736(20)30...
,33. Webster AC, Nagler EV, Morton RL, Masson P. Chronic Kidney Disease. Lancet. 2017;389(10075):1238–52. doi: http://doi.org/10.1016/S0140-6736(16)32064-5. PubMed PMID: 27887750.
https://doi.org/10.1016/S0140-6736(16)32...
. DM is an intermediate metabolic disorder characterized by persistent elevations of plasma glucose levels. The prevalence of DM has increased significantly in recent years, mainly due to sedentary lifestyles, obesity, and increased life expectancy. In 1985, about 30 million cases were reported worldwide, rising to 415 million in 2015 and an estimated 642 million cases by 204044. Ogurtsova K, da Rocha Fernandes JD, Huang Y, Linnenkamp U, Guariguata L, Cho NH, et al. IDF Diabetes Atlas: global estimates for the prevalence of diabetes for 2015 and 2040. Diabetes Res Clin Pract. 2017;128:40–50. doi: http://doi.org/10.1016/j.diabres.2017.03.024. PubMed PMID: 28437734.
https://doi.org/10.1016/j.diabres.2017.0...
.

The development of diabetic kidney disease (DKD) is mainly related to diabetes lifespan and the strictness of glycemic control, with several macrovascular and microvascular associated complications, including peripheral arterial occlusive disease, diabetic retinopathy and DKD or diabetic nephropathy (DN) – the most common chronic complications of DM. It is estimated that approximately 40% of individuals with DM develop CKD, which manifests as DKD. This makes it the leading cause of CKD worldwide3 and a major cause of death in these patients55. Bentata Y, Haddiya I, Latrech H, Serraj K, Abouqal R. Progression of diabetic nephropathy, risk of end-stage kidney disease and mortality in patients with type-1 diabetes. Saudi J Kidney Dis Transpl. 2013;24(2):392–402. doi: http://doi.org/10.4103/1319-2442.109617. PubMed PMID: 23538374.
https://doi.org/10.4103/1319-2442.109617...
.

The pathogenesis of DKD is influenced by functional, histopathological, and immune mechanisms. From a functional point of view, chronic hyperglycemia causes hyperfiltration and an increase in glomerular pressure, which is clinically characterized by an imbalance in the muscle tone of the afferent and efferent arterioles, resulting in an increase in glomerular filtration rate (GFR), glomerular hypertension, and increased excretion of albumin in the urine to levels greater than 30 mg/g33. Webster AC, Nagler EV, Morton RL, Masson P. Chronic Kidney Disease. Lancet. 2017;389(10075):1238–52. doi: http://doi.org/10.1016/S0140-6736(16)32064-5. PubMed PMID: 27887750.
https://doi.org/10.1016/S0140-6736(16)32...
. Histologically, this is manifested by nodular and/or diffuse glomerulosclerosis, tubulointerstitial fibrosis, mesangial dilatation and basement membrane thickening, all changes that are considered characteristic of DKD66. Reidy K, Kang HM, Hostetter T, Susztak K. Molecular mechanisms of diabetic kidney disease. J Clin Invest. 2014;124(6):2333–40. doi: http://doi.org/10.1172/JCI72271. PubMed PMID: 24892707.
https://doi.org/10.1172/JCI72271...
.

Over time, this sustained increase in GFR associated with glomerular hypertension leads to mechanical stress on the filtration barrier, resulting in loss of nephron function, which in turn leads to proteinuria and the development of CKD77. Denic A, Mathew J, Lerman LO, Lieske JC, Larson JJ, Alexander MP, et al. Single-nephron glomerular filtration rate in healthy adults. N Engl J Med. 2017;376(24):2349–57. doi: http://doi.org/10.1056/NEJMoa1614329. PubMed PMID: 28614683.
https://doi.org/10.1056/NEJMoa1614329...
. One cohort (RIACE) has shown that glomerular hyperfiltration is an independent factor for death from any cause in patients with type 2 DM (T2DM)88. Penno G, Orsi E, Solini A, Bonora E, Fondelli C, Trevisan R, et al. Kidney hyperfiltration is independently associated with increased all-cause mortality in individuals with type 2 diabetes: a prospective cohort study. BMJ Open Diabetes Res Care. 2020;8(1):e001481. doi: http://doi.org/10.1136/bmjdrc-2020-001481. PubMed PMID: 32665314.
https://doi.org/10.1136/bmjdrc-2020-0014...
. Minutolo et al.99. Minutolo R, Gabbai FB, Provenzano M, Chiodini P, Borrelli S, Garofalo C, et al. Cardiokidney prognosis by residual proteinuria level in diabetic chronic kidney disease: pooled analysis of four cohort studies. Nephrol Dial Transplant. 2018;33(11):1942–9. doi: http://doi.org/10.1093/ndt/gfy032. PubMed PMID: 29509925.
https://doi.org/10.1093/ndt/gfy032...
showed that the presence of albuminuria is associated with increased mortality and cardiovascular events in patients with DKD compared with patients with CKD of non-diabetic etiology, with the risk directly related to the severity of albuminuria.

Pharmacological Treatment

Pharmacological treatment is an important pillar in the treatment of DN, acting mainly on glycemic control. In the early stages of both type 1 DM (T1DM) and T2DM, microvascular and macrovascular complications can be reduced by strict glycemic control1010. Guedes M, Pecoits-Filho R. Can we cure diabetic kidney disease? Present and future perspectives from a nephrologist’s point of view. J Intern Med. 2022;291(2):165–80. doi: http://doi.org/10.1111/joim.13424. PubMed PMID: 34914852.
https://doi.org/10.1111/joim.13424...
.

Oral diabetes medications can work in a variety of ways, including lowering hepatic glucose production (biguanides/metformin), increasing insulin secretion (glinides and sulphonylureas), lowering intestinal glucose absorption (alpha-glucosidase inhibitors), increasing peripheral insulin sensitivity (thioglitazones), lowering gastric emptying (GLP1 agonists), increasing GLP1 half-life (DPP4 inhibitors) and, more recently, stimulating of urinary glucose excretion (SGLT2 inhibitors)1111. ElSayed NA, Aleppo G, Aroda VR, Bannuru RR, Brown FM, Bruemmer D, et al. Pharmacologic approaches to glycemic treatment: standards of care in diabetes-2023. Diabetes Care. 2023;46(Suppl 1):S140–57. doi: http://doi.org/10.2337/dc23-S009. PubMed PMID: 36507650.
https://doi.org/10.2337/dc23-S009...
.

Angiotensin II converting enzyme (ACE) inhibitors or aldosterone receptor blockers (ARBs) are also key drugs in the treatment of DKD, as they can reduce hyperfiltration and glomerular hypertension by vasoconstriction of the afferent arteriole and vasodilation of the efferent arteriole. Important clinical studies have shown that the use of ACE inhibitors in patients with albuminuria reduces the progression of CKD. These drugs are responsible for the neurohormonal control of the kidneys and their use allows the optimization of kidney hemodynamics due to the pro-inflammatory state present in kidney patients1212. Lewis EJ, Hunsicker LG, Bain RP, Rohde RD, and the The Collaborative Study Group. The effect of angiotensin-converting-enzyme inhibition on diabetic nephropathy. N Engl J Med. 1993;329(20):1456–62. doi: http://doi.org/10.1056/NEJM199311113292004. PubMed PMID: 8413456.
https://doi.org/10.1056/NEJM199311113292...
,1313. Brenner BM, Cooper ME, de Zeeuw D, Keane WF, Mitch WE, Parving HH, et al. Effects of losartan on kidney and cardiovascular outcomes in patients with type 2 diabetes and nephropathy. N Engl J Med. 2001;345(12):861–9. doi: http://doi.org/10.1056/NEJMoa011161. PubMed PMID: 11565518.
https://doi.org/10.1056/NEJMoa011161...
. Despite this improvement, these drugs are not able to eliminate the long-term increased risk of mortality in these patients1414. Provenzano M, Coppolino G, De Nicola L, Serra R, Garofalo C, Andreucci M, et al. Unraveling cardiovascular risk in kidney patients: a new take on old tale. Front Cell Dev Biol. 2019;7:314. doi: http://doi.org/10.3389/fcell.2019.00314. PubMed PMID: 31850348.
https://doi.org/10.3389/fcell.2019.00314...
,1515. Provenzano M, De Nicola L, Pena MJ, Capitoli G, Garofalo C, Borrelli S, et al. Precision nephrology is a non-negligible state of mind in clinical research: remember the past to face the future. Nephron. 2020;144(10):463–78. doi: http://doi.org/10.1159/000508983. PubMed PMID: 32810859.
https://doi.org/10.1159/000508983...
.

SGLT2 Inhibitors

Sodium-glucose cotransporter 2 inhibitors (SGLT2i), a new class of oral antidiabetic agents launched in 2012 to slow disease progression by lowering blood glucose levels via the urine1616. Idris I, Donnelly R. Sodium-glucose co-transporter-2 inhibitors: an emerging new class of oral antidiabetic drug. Diabetes Obes Metab. 2009;11(2):79–88. doi: http://doi.org/10.1111/j.1463-1326.2008.00982.x. PubMed PMID: 19125776.
https://doi.org/10.1111/j.1463-1326.2008...
, have shown metabolic benefits and the ability to slow DKD progression in several clinical studies in recent years.

SGLT2i act on the proximal kidney tubules and inhibit glucose reabsorption independently of insulin or pancreatic beta cell function. In this sense, SGLT2i has been the subject of large studies due to their proven benefit on cardiovascular and kidney outcomes1717. Perkovic V, Jardine MJ, Neal B, Bompoint S, Heerspink HJL, Charytan DM, et al. Canagliflozin and kidney outcomes in type 2 diabetes and nephropathy. N Engl J Med. 2019;380(24):2295–306. doi: http://doi.org/10.1056/NEJMoa1811744. PubMed PMID: 30990260.
https://doi.org/10.1056/NEJMoa1811744...
, mainly because they provide improved glycemic control by regulating glycosylated hemoglobin, fasting glucose and postprandial glycemia, blood pressure reduction due to urinary sodium loss1818. Musso G, Gambino R, Cassader M, Pagano G. A novel approach to control hyperglycemia in type 2 diabetes: sodium glucose co-transport (SGLT) inhibitors: systematic review and meta-analysis of randomized trials. Ann Med. 2012;44(4):375–93. doi: http://doi.org/10.3109/07853890.2011.560181. PubMed PMID: 21495788.
https://doi.org/10.3109/07853890.2011.56...
, and weight loss1919. Cefalu WT. Paradoxical insights into whole body metabolic adaptations following SGLT2 inhibition. J Clin Invest. 2014;124(2):485–7. doi: http://doi.org/10.1172/JCI74297. PubMed PMID: 24463446.
https://doi.org/10.1172/JCI74297...
.

In the DAPA-CKD trial, dapagliflozin use significantly reduced the risk of kidney function loss (estimated by GFR), CKD progression and kidney-related death in patients with CKD, regardless of blood glucose status2020. 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(15):1436–46. doi: http://doi.org/10.1056/NEJMoa2024816. PubMed PMID: 32970396.
https://doi.org/10.1056/NEJMoa2024816...
. The study also showed that non-diabetic patients with CKD who received dapagliflozin had improved kidney function (also estimated by GFR) and a lower risk of kidney-related death. Another landmark study in the medical literature is the EMPA Kidney trial, which showed that empagliflozin reduced the risk of kidney disease progression or cardiovascular death in patients with CKD by 28% compared to placebo2121. Herrington WG, Staplin N, Wanner C, Green JB, Hauske SJ, Emberson JR, et al. Empagliflozin in Patients with Chronic Kidney Disease. N Engl J Med. 2023;388(2):117–27. doi: http://doi.org/10.1056/NEJMoa2204233. PubMed PMID: 36331190.
https://doi.org/10.1056/NEJMoa2204233...
.

SGLTs are ATP-dependent transmembrane transporters that act as carriers of glucose, ions and amino acids, with the most studied subtypes being SGLT1 and SGLT22222. Ehrenkranz JR, Lewis NG, Kahn CR, Roth J. Phlorizin: a review. Diabetes Metab Res Rev. 2005;21(1):31–8. doi: http://doi.org/10.1002/dmrr.532. PubMed PMID: 15624123.
https://doi.org/10.1002/dmrr.532...
. The SGLT1 subtype is less specific as it is found in various organs such as the small intestine, brain, heart and distal segments of the proximal kidney tubules (segment S3) and is responsible for the reabsorption of approximately 10% of the glucose filtered by the kidney1717. Perkovic V, Jardine MJ, Neal B, Bompoint S, Heerspink HJL, Charytan DM, et al. Canagliflozin and kidney outcomes in type 2 diabetes and nephropathy. N Engl J Med. 2019;380(24):2295–306. doi: http://doi.org/10.1056/NEJMoa1811744. PubMed PMID: 30990260.
https://doi.org/10.1056/NEJMoa1811744...
. Because they are located in different parts of the body, SGLT1 subtype inhibitors can cause various adverse effects, such as gastrointestinal disorders, which led to the discontinuation of studies that initially involved this subtype2222. Ehrenkranz JR, Lewis NG, Kahn CR, Roth J. Phlorizin: a review. Diabetes Metab Res Rev. 2005;21(1):31–8. doi: http://doi.org/10.1002/dmrr.532. PubMed PMID: 15624123.
https://doi.org/10.1002/dmrr.532...
. On the other hand, the SGLT2 subtype is a specific transporter of kidney tissue with low affinity and high glucose reabsorption capacity, which is found in the apical membrane of the proximal cells of the kidney tubules (segment S1) and is responsible for the reabsorption of approximately 90% of the glucose filtered by the kidney1616. Idris I, Donnelly R. Sodium-glucose co-transporter-2 inhibitors: an emerging new class of oral antidiabetic drug. Diabetes Obes Metab. 2009;11(2):79–88. doi: http://doi.org/10.1111/j.1463-1326.2008.00982.x. PubMed PMID: 19125776.
https://doi.org/10.1111/j.1463-1326.2008...
.

The first drug of the SGLT2i class to be discovered was phlorizin, a molecule found in the leaves and buds of apple trees2323. Provenzano M, Pelle MC, Zaffina I, Tassone B, Pujia R, Ricchio M, et al. Sodium-glucose co-transporter-2 inhibitors and nephroprotection in diabetic patients: more than a challenge. Front Med (Lausanne). 2021;8:654557. doi: http://doi.org/10.3389/fmed.2021.654557. PubMed PMID: 34150796.
https://doi.org/10.3389/fmed.2021.654557...
. However, due to its low selectivity for SGLT2 and various side effects, mainly gastrointestinal, the drug ceased to be used2424. Giugliano D, Esposito K. Class effect for SGLT-2 inhibitors: a tale of 9 drugs. Cardiovasc Diabetol. 2019;18(1):94. doi: http://doi.org/10.1186/s12933-019-0899-9. PubMed PMID: 31337395.
https://doi.org/10.1186/s12933-019-0899-...
. Nevertheless, other drugs have been developed based on its molecular structure that increase selectivity for SGLT2, thereby reducing side effects and increasing bioavailability.

To date, there are approximately 9 molecules that can inhibit the SGLT2 transporter, but only 4 have been approved by the Food and Drug Administration (FDA) for the treatment of DM2, namely: empagliflozin, dapagliflozin, canagliflozin and ertagliflozin2525. Garcia-Ropero A, Badimon JJ, Santos-Gallego CG. The pharmacokinetics and pharmacodynamics of SGLT2 inhibitors for type 2 diabetes mellitus: the latest developments. Expert Opin Drug Metab Toxicol. 2018;14(12):1287–302. doi: http://doi.org/10.1080/17425255.2018.1551877. PubMed PMID: 30463454.
https://doi.org/10.1080/17425255.2018.15...
. Dapagliflozin was the first drug in this class to be approved in 2012, while empagliflozin was approved in 20142626. Shubrook JH, Bokaie BB, Adkins SE. Empagliflozin in the treatment of type 2 diabetes: evidence to date. Drug Des Devel Ther. 2015;9:5793–803. doi: http://doi.org/10.2147/DDDT.S69926. PubMed PMID: 26586935.
https://doi.org/10.2147/DDDT.S69926...
. In the class, empagliflozin has the highest affinity for SGLT2 compared to SGLT12727. 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(22):2117–28. doi: http://doi.org/10.1056/NEJMoa1504720. PubMed PMID: 26378978.
https://doi.org/10.1056/NEJMoa1504720...
,2828. Ghezzi C, Loo DDF, Wright EM. Physiology of kidney glucose handling via SGLT1, SGLT2 and GLUT2. Diabetologia. 2018;61(10):2087–97. doi: http://doi.org/10.1007/s00125-018-4656-5. PubMed PMID: 30132032.
https://doi.org/10.1007/s00125-018-4656-...
,2929. Liu JJ, Lee T, DeFronzo RA. Why Do SGLT2 inhibitors inhibit only 30-50% of kidney glucose reabsorption in humans? Diabetes. 2012;61(9):2199–204. doi: http://doi.org/10.2337/db12-0052. PubMed PMID: 22923645.
https://doi.org/10.2337/db12-0052...
.

In the proximal tubules, the sodium-to-glucose ratio is 1:1 for SGLT2 (S1 segment) and 2:1 for SGLT1 (S3 segment)3030. Gallo LA, Wright EM, Vallon V. Probing SGLT2 as a therapeutic target for diabetes: basic physiology and consequences. Diab Vasc Dis Res. 2015;12(2):78–89. doi: http://doi.org/10.1177/1479164114561992. PubMed PMID: 25616707.
https://doi.org/10.1177/1479164114561992...
. The reabsorbed glucose is then transferred to the interstitium by facilitated diffusion through glucose transporters (GLUTs) present in the basolateral membrane of the cell3131. Thomas MC. Kidney effects of dapagliflozin in patients with type 2 diabetes. Ther Adv Endocrinol Metab. 2014;5(3):53–61. doi: http://doi.org/10.1177/2042018814544153. PubMed PMID: 25126408.
https://doi.org/10.1177/2042018814544153...
.

It is estimated that a healthy adult can filter approximately 180 g of glucose per day3232. 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(1):R75–83. doi: http://doi.org/10.1152/ajpregu.00357.2011. PubMed PMID: 21940401.
https://doi.org/10.1152/ajpregu.00357.20...
. At higher levels, the ability of the kidney to reabsorb glucose is impaired, so that elevated glucose levels are detected in urine tests, causing symptoms of non-glycemic control such as polyuria and polydipsia. Paradoxically, this loss of glucose in diabetics leads to hypertrophy of the cells of the proximal kidney tubules, where the SGLT2 transporter is located, with an increase in its expression in the apical membrane, increasing the capacity for reabsorption of kidney glucose by this transporter to preserve energy in the body.

In this way, there is an increase in the reabsorption of glucose and sodium, which leads to a decrease in the amount of NaCl in the lumen of the distal tubules near the dense macula. This initiates tubuloglomerular feedback, which relaxes the tone of the kidney afferent arteriole, increasing blood flow and GFR, causing glomerular hypertension3232. 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(1):R75–83. doi: http://doi.org/10.1152/ajpregu.00357.2011. PubMed PMID: 21940401.
https://doi.org/10.1152/ajpregu.00357.20...
. This process further increases the already high glycemic levels, leading to increased insulin secretion and body weight gain3333. 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. 2021;320(5):F761–71. doi: http://doi.org/10.1152/ajprenal.00552.2020. PubMed PMID: 33645318.
https://doi.org/10.1152/ajprenal.00552.2...
.

By blocking the SGLT2 transporter, this class of drugs promotes a reduction in sodium absorption, which has important effects on the dynamics of kidney filtration. The first effect is related to the excretion of sodium in the urine, resulting in a decrease in blood pressure and effective circulating volume3434. van Bommel EJM, Muskiet MHA, van Baar MJB, Tonneijck L, Smits MM, Emanuel AL, et al. The kidney 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(1):202–12. doi: http://doi.org/10.1016/j.kint.2019.09.013. PubMed PMID: 31791665.
https://doi.org/10.1016/j.kint.2019.09.0...
. The second effect is related to tubuloglomerular feedback, in which the increased presence of sodium in the macula densa causes vasoconstriction of the afferent arteriole, reducing glomerular hyperfiltration3434. van Bommel EJM, Muskiet MHA, van Baar MJB, Tonneijck L, Smits MM, Emanuel AL, et al. The kidney 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(1):202–12. doi: http://doi.org/10.1016/j.kint.2019.09.013. PubMed PMID: 31791665.
https://doi.org/10.1016/j.kint.2019.09.0...
.

Experimental studies in rats have shown that the use of SGLT2i can reduce hyperfiltration by about 25% through the decrease in the reabsorption of NaCl and glucose in the proximal tubules, thus reducing intraglomerular pressure (5–8 mmHg)3535. Thomas MC, Cherney DZI. The actions of SGLT2 inhibitors on metabolism, kidney function and blood pressure. Diabetologia. 2018;61(10):2098–107. doi: http://doi.org/10.1007/s00125-018-4669-0. PubMed PMID: 30132034.
https://doi.org/10.1007/s00125-018-4669-...
. The reduction of intraglomerular pressure is an important factor in the reduction of albuminuria caused by SGLT2i3535. Thomas MC, Cherney DZI. The actions of SGLT2 inhibitors on metabolism, kidney function and blood pressure. Diabetologia. 2018;61(10):2098–107. doi: http://doi.org/10.1007/s00125-018-4669-0. PubMed PMID: 30132034.
https://doi.org/10.1007/s00125-018-4669-...
. Another hypothesis about the mechanism of action of SGLT2i is related to changes in lipid metabolism and increased production of ketone bodies3636. Szekeres Z, Toth K, Szabados E. The Effects of SGLT2 Inhibitors on Lipid Metabolism. Metabolites. 2021;11(2):87. doi: http://doi.org/10.3390/metabo11020087. PubMed PMID: 33535652.
https://doi.org/10.3390/metabo11020087...
as well as hemodynamic changes3535. Thomas MC, Cherney DZI. The actions of SGLT2 inhibitors on metabolism, kidney function and blood pressure. Diabetologia. 2018;61(10):2098–107. doi: http://doi.org/10.1007/s00125-018-4669-0. PubMed PMID: 30132034.
https://doi.org/10.1007/s00125-018-4669-...
.

Inflammatory Responses and Kidney Oxidative Stress

Recent studies have shown that the inflammatory response and oxidative stress are important in the development and progression of DKD. This is mainly due to the innate immune system, the increase in reactive oxygen species (ROS) and the increase in pro-inflammatory cells and cytokines such as interleukin-1ß, interleukin-18, tumor necrosis factor alpha (TNF-α) and nuclear factor kB (NF-kβ)3737. Hickey FB, Martin F. Role of the immune system in diabetic kidney disease. Curr Diab Rep. 2018;18(4):20. doi: http://doi.org/10.1007/s11892-018-0984-6. PubMed PMID: 29532281.
https://doi.org/10.1007/s11892-018-0984-...
,3838. Kawanami D, Matoba K, Utsunomiya K. Signaling pathways in diabetic nephropathy. Histol Histopathol. 2016;31(10):1059–67. PubMed PMID: 27094540.,3939. Matoba K, Takeda Y, Nagai Y, Kawanami D, Utsunomiya K, Nishimura R. Unraveling the role of inflammation in the pathogenesis of diabetic kidney disease. Int J Mol Sci. 2019;20(14):3393. doi: http://doi.org/10.3390/ijms20143393. PubMed PMID: 31295940.
https://doi.org/10.3390/ijms20143393...
,4040. Komada T, Muruve DA. The role of inflammasomes in kidney disease. Nat Rev Nephrol. 2019;15(8):501–20. doi: http://doi.org/10.1038/s41581-019-0158-z. PubMed PMID: 31164720.
https://doi.org/10.1038/s41581-019-0158-...
.

Historically, DKD was believed to be a metabolic disease characterized only by hyperglycemia associated with cardio-metabolic risk factors such as high blood pressure, obesity and dyslipidemia3838. Kawanami D, Matoba K, Utsunomiya K. Signaling pathways in diabetic nephropathy. Histol Histopathol. 2016;31(10):1059–67. PubMed PMID: 27094540.. Despite this, a number of studies have highlighted the complexity of the disease and described the importance of innate immunity, inflammatory response and oxidative stress in its development and progression3939. Matoba K, Takeda Y, Nagai Y, Kawanami D, Utsunomiya K, Nishimura R. Unraveling the role of inflammation in the pathogenesis of diabetic kidney disease. Int J Mol Sci. 2019;20(14):3393. doi: http://doi.org/10.3390/ijms20143393. PubMed PMID: 31295940.
https://doi.org/10.3390/ijms20143393...
,4040. Komada T, Muruve DA. The role of inflammasomes in kidney disease. Nat Rev Nephrol. 2019;15(8):501–20. doi: http://doi.org/10.1038/s41581-019-0158-z. PubMed PMID: 31164720.
https://doi.org/10.1038/s41581-019-0158-...
.

The inflammasome NLRP3 complex, an intracytoplasmic receptor found in cells of the innate immune system, is a key regulator of this inflammatory mechanism. It is known to play a role in the pathophysiology of neurodegenerative and cardiovascular diseases such as Alzheimer’s and heart failure, and has recently been studied in relation to metabolic diseases such as gout and T2DM4040. Komada T, Muruve DA. The role of inflammasomes in kidney disease. Nat Rev Nephrol. 2019;15(8):501–20. doi: http://doi.org/10.1038/s41581-019-0158-z. PubMed PMID: 31164720.
https://doi.org/10.1038/s41581-019-0158-...
.

Additionally, oxidative stress is a crucial factor in the pathogenesis of DKD. In DKD, oxidative damage to the kidneys is caused by an increase in reactive oxygen species (ROS) production and a decrease in antioxidant defense mechanisms. In DKD, hyperglycemia, dyslipidemia and activation of the renin-angiotensin-aldosterone system contribute to oxidative stress4141. Jha JC, Banal C, Chow BS, Cooper ME, Jandeleit-Dahm K. Diabetes and kidney disease: role of oxidative stress. Antioxid Redox Signal. 2016;25(12):657–84. doi: http://doi.org/10.1089/ars.2016.6664. PubMed PMID: 26906673.
https://doi.org/10.1089/ars.2016.6664...
,4242. Volpe CMO, Villar-Delfino PH, Dos Anjos PMF, Nogueira-Machado JA. Cellular death, reactive oxygen species (ROS) and diabetic complications. Cell Death Dis. 2018;9(2):119. doi: http://doi.org/10.1038/s41419-017-0135-z. PubMed PMID: 29371661.
https://doi.org/10.1038/s41419-017-0135-...
.

ROS are generated by mitochondrial respiratory chain enzymes and can directly damage lipids, proteins and DNA as well as activate multiple signaling pathways that contribute to inflammation and fibrosis. For example, hyperglycemia activates a specific metabolic pathway involving diacylglycerol (DAG), protein kinase C (PKC), and NADPH oxidase, resulting in the production of ROS4242. Volpe CMO, Villar-Delfino PH, Dos Anjos PMF, Nogueira-Machado JA. Cellular death, reactive oxygen species (ROS) and diabetic complications. Cell Death Dis. 2018;9(2):119. doi: http://doi.org/10.1038/s41419-017-0135-z. PubMed PMID: 29371661.
https://doi.org/10.1038/s41419-017-0135-...
.

In DKD, chronic hyperglycemia increases ROS production and induces cell apoptosis, contributing to the progression of diabetic complications. Thus, reducing oxidative stress through various mechanisms (lifestyle modifications, medications and targeted antioxidant therapies) can prevent or delay the progression of DKD4343. Singh DK, Winocour P, Farrington K. Oxidative stress in early diabetic nephropathy: fueling the fire. Nat Rev Endocrinol. 2011;7(3):176–84. doi: http://doi.org/10.1038/nrendo.2010.212. PubMed PMID: 21151200.
https://doi.org/10.1038/nrendo.2010.212...
.

The Innate Immunity

Innate immunity is the body’s first line of defense against infectious agents. It consists primarily of natural killer cells, the complement system and the phagocytic system. The phagocytic system, especially represented by macrophages, is important for the production of pro-inflammatory cytokines, ROS and metalloproteinases, which are responsible for local inflammation, atherogenesis, and tissue damage4444. Galkina E, Ley K. Leukocyte recruitment and vascular injury in diabetic nephropathy. J Am Soc Nephrol. 2006;17(2):368–77. doi: http://doi.org/10.1681/ASN.2005080859. PubMed PMID: 16394109.
https://doi.org/10.1681/ASN.2005080859...
-46.

Several studies have already demonstrated that the magnitude of infiltration by these cells in the kidney parenchyma is directly related to the degree of functional dysfunction of the kidneys, suggesting a possible causal relationship4747. Awad AS, Kinsey GR, Khutsishvili K, Gao T, Bolton WK, Okusa MD. Monocyte/macrophage chemokine receptor CCR2 mediates diabetic kidney injury. Am J Physiol Renal Physiol. 2011;301(6):F1358–66. doi: http://doi.org/10.1152/ajprenal.00332.2011. PubMed PMID: 21880831.
https://doi.org/10.1152/ajprenal.00332.2...
,4848. Lim AK, Ma FY, Nikolic-Paterson DJ, Kitching AR, Thomas MC, Tesch GH. Lymphocytes promote albuminuria, but not kidney dysfunction or histological damage in a mouse model of diabetic kidney injury. Diabetologia. 2010;53(8):1772–82. doi: http://doi.org/10.1007/s00125-010-1757-1. PubMed PMID: 20422398.
https://doi.org/10.1007/s00125-010-1757-...
. Furuta et al. were among the first to observe this phenomenon in 1993 when studying macrophage infiltration in biopsies from DM patients4949. Furuta T, Saito T, Ootaka T, Soma J, Obara K, Abe K, et al. The role of macrophages in diabetic glomerulosclerosis. Am J Kidney Dis. 1993;21(5):480–5. doi: http://doi.org/10.1016/S0272-6386(12)80393-3. PubMed PMID: 8488815.
https://doi.org/10.1016/S0272-6386(12)80...
and suggested that the phagocytic system may be related to the irreversible damage to the glomerular structure. In 2006, Nguyen et al. demonstrated an accumulation of macrophages and immune cells in the kidney tubules in biopsy tissues from patients with DKD, linking the intensity of interstitial accumulation to the decline in kidney function, demonstrating the importance of these cells in the pathogenesis of the disease5050. Nguyen D, Ping F, Mu W, Hill P, Atkins RC, Chadban SJ. Macrophage accumulation in human progressive diabetic nephropathy. Nephrology (Carlton). 2006;11(3):226–31. doi: http://doi.org/10.1111/j.1440-1797.2006.00576.x. PubMed PMID: 16756636.
https://doi.org/10.1111/j.1440-1797.2006...
.

Ninichuk et al. showed the following year that blocking CCR-1, a chemokine that attracts macrophages to kidney tissue, was able to reduce tubular fibrosis and interstitial inflammation in a rat model of DKD5151. Ninichuk V, Khandoga AG, Segerer S, Loetscher P, Schlapbach A, Revesz L, et al. The role of interstitial macrophages in nephropathy of type 2 diabetic db/db mice. Am J Pathol. 2007;170(4):1267–76. doi: http://doi.org/10.2353/ajpath.2007.060937. PubMed PMID: 17392166.
https://doi.org/10.2353/ajpath.2007.0609...
. In 2017, Klessens et al. used a similar method to demonstrate accumulation of macrophages in the glomeruli from biopsy of 88 patients with kidney disease from diabetes and linked phagocytic accumulation as an important factor in disease progression5252. Klessens CQF, Zandbergen M, Wolterbeek R, Bruijn JA, Rabelink TJ, Bajema IM, et al. Macrophages in diabetic nephropathy in patients with type 2 diabetes. Nephrol Dial Transplant. 2017;32(8):1322–9. PubMed PMID: 27416772.. The increase in expression of chemokines and adhesion molecules (CCR-1, CCR-2) has already been observed in kidney biopsies from patients with diabetic kidney disease5353. Tang SC, Chan LY, Leung JC, Cheng AS, Chan KW, Lan HY, et al. Bradykinin and high glucose promote kidney tubular inflammation. Nephrol Dial Transplant. 2010;25(3):698–710. doi: http://doi.org/10.1093/ndt/gfp599. PubMed PMID: 19923143.
https://doi.org/10.1093/ndt/gfp599...
,5454. Kang YS, Lee MH, Song HK, Ko GJ, Kwon OS, Lim TK, et al. CCR2 antagonism improves insulin resistance, lipid metabolism, and diabetic nephropathy in type 2 diabetic mice. Kidney Int. 2010;78(9):883–94. doi: http://doi.org/10.1038/ki.2010.263. PubMed PMID: 20686445.
https://doi.org/10.1038/ki.2010.263...
. Studies evaluating the use of antagonists of these receptors have already demonstrated benefits in animal models, resulting in a reduction in macrophage infiltration in the kidney parenchyma and a decrease in albumin excretion5454. Kang YS, Lee MH, Song HK, Ko GJ, Kwon OS, Lim TK, et al. CCR2 antagonism improves insulin resistance, lipid metabolism, and diabetic nephropathy in type 2 diabetic mice. Kidney Int. 2010;78(9):883–94. doi: http://doi.org/10.1038/ki.2010.263. PubMed PMID: 20686445.
https://doi.org/10.1038/ki.2010.263...
.

Immune System Recognition Receptors

Cells of the immune system have specific receptors known as pattern recognition receptors (PRR), which are responsible for detecting structural molecules of microorganisms known as pathogen-associated molecular patterns (PAMPs), and toxins or fragments of DNA and RNA resulting from cell damage, such as nucleic acids, intracellular proteins such as HMGB1 (High-mobility group B1), HSPs (heat shock proteins) and messenger RNA, known as tissue damage-associated molecules (DAMPs)5555. Kumar H, Kawai T, Akira S. Pathogen recognition by the innate immune system. Int Rev Immunol. 2011;30(1):16–34. doi: http://doi.org/10.3109/08830185.2010.529976. PubMed PMID: 21235323.
https://doi.org/10.3109/08830185.2010.52...
.

In the context of DKD, the chronic hyperglycemic state can cause endothelial and cellular lesions that release DAMPs and PAMPs into the interstitium, which are recognized by PRRs. PRRs, which may be found in the plasma membrane or cell cytoplasm, work together to recognize stress signals produced by cells during infection or cell injury5656. Tesch GH. Diabetic nephropathy - is this an immune disorder? Clin Sci (Lond). 2017;131(16):2183–99. doi: http://doi.org/10.1042/CS20160636. PubMed PMID: 28760771.
https://doi.org/10.1042/CS20160636...
. When these receptors are present on the membrane, they are referred to as Toll Like Receptors (TLRs). When expressed on the surface of kidney cells, they are largely responsible for inducing the immune response by recognizing DAMPs and PAMPs5757. Wada J, Makino H. Inflammation and the pathogenesis of diabetic nephropathy. Clin Sci (Lond). 2013;124(3):139–52. doi: http://doi.org/10.1042/CS20120198. PubMed PMID: 23075333.
https://doi.org/10.1042/CS20120198...
. These DAMPS and PAMPS lead to activation of the innate immune response5858. Ram C, Jha AK, Ghosh A, Gairola S, Syed AM, Murty US, et al. Targeting NLRP3 inflammasome as a promising approach for treatment of diabetic nephropathy: preclinical evidences with therapeutic approaches. Eur J Pharmacol. 2020;885:173503. doi: http://doi.org/10.1016/j.ejphar.2020.173503. PubMed PMID: 32858047.
https://doi.org/10.1016/j.ejphar.2020.17...
, leading to kidney inflammation and tissue damage mainly through activation of the NF-kB pathway5959. Brubaker SW, Bonham KS, Zanoni I, Kagan JC. Innate immune pattern recognition: a cell biological perspective. Annu Rev Immunol. 2015;33(1):257–90. doi: http://doi.org/10.1146/annurev-immunol-032414-112240. PubMed PMID: 25581309.
https://doi.org/10.1146/annurev-immunol-...
.

C-type lectin receptors (CLRs), retinoic acid-inducible gene-like receptors (RLRs), absence-of-melanoma-like receptors (ALRs), and nucleotide-binding and oligomerization domain (NOD)-like receptors (NLRs) are all examples of PRRs6060. Takeuchi O, Akira S. Pattern recognition receptors and inflammation. Cell. 2010;140(6):805-20. doi: http://doi.org/10.1016/j.cell.2010.01.022. PubMed PMID: 20303872.
https://doi.org/10.1016/j.cell.2010.01.0...
,6161. Kanneganti TD, Lamkanfi M, Nunez G. Intracellular NOD-like receptors in host defense and disease. Immunity. 2007;27(4):549–59. doi: http://doi.org/10.1016/j.immuni.2007.10.002. PubMed PMID: 17967410.
https://doi.org/10.1016/j.immuni.2007.10...
. The NLRs are responsible for the host’s second line of defense and are further subdivided into subtypes such as NLRP1, NLRP3, NAIP, and NLRC46262. Barbe F, Douglas T, Saleh M. Advances in Nod-like receptors (NLR) biology. Cytokine Growth Factor Rev. 2014;25(6):681–97. doi: http://doi.org/10.1016/j.cytogfr.2014.07.001. PubMed PMID: 25070125.
https://doi.org/10.1016/j.cytogfr.2014.0...
. Each has the ability to be activated specifically by certain groups of PAMPs and/or DAMPs, and their activation leads to the formation of cytoplasmic protein complexes known as inflammasomes.

The NLRP3 Inflammasome

NLRs are cytoplasmic receptors that act as the host’s second line of defense and have three functional structures: a central oligomerization domain (NATCH), a C-terminal domain rich in leucine repeats (LRR), and an N-terminal domain responsible for stimulus transduction with the subdomains CARD (Caspase activation and recruitment domain) or PYD (pyrin domain)6363. Kim SM, Lee SH, Kim YG, Kim SY, Seo JW, Choi YW, et al. Hyperuricemia-induced NLRP3 activation of macrophages contributes to the progression of diabetic nephropathy. Am J Physiol Renal Physiol. 2015;308(9):F993–1003. doi: http://doi.org/10.1152/ajprenal.00637.2014. PubMed PMID: 25651569.
https://doi.org/10.1152/ajprenal.00637.2...
. These subdomains are responsible for triggering specific immune responses, causing the NLRs being further subdivided into other subtypes, such as NLRP1, NLRP3, NAIP and NLRC46262. Barbe F, Douglas T, Saleh M. Advances in Nod-like receptors (NLR) biology. Cytokine Growth Factor Rev. 2014;25(6):681–97. doi: http://doi.org/10.1016/j.cytogfr.2014.07.001. PubMed PMID: 25070125.
https://doi.org/10.1016/j.cytogfr.2014.0...
. The receptor subtype whose mechanism of function is best understood in the context of DKD is NLRP36464. Yang Y, Wang H, Kouadir M, Song H, Shi F. Recent advances in the mechanisms of NLRP3 inflammasome activation and its inhibitors. Cell Death Dis. 2019;10(2):128. doi: http://doi.org/10.1038/s41419-019-1413-8. PubMed PMID: 30755589.
https://doi.org/10.1038/s41419-019-1413-...
,6565. Shi J, Zhao Y, Wang K, Shi X, Wang Y, Huang H, et al. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature. 2015;526(7575):660–5. doi: http://doi.org/10.1038/nature15514. PubMed PMID: 26375003.
https://doi.org/10.1038/nature15514...
.

In the NLRP3 complex, the N-terminal domain, responsible for stimulus transduction has a PYD subdomain. On the other hand, the pro-caspase-1 enzyme has a CARD subdomain. In order for the interaction between the NLRP3 protein and the pro-caspase-1 enzyme to occur, an adaptation protein (ASC) is therefore required that has the PYD and CARD domains6565. Shi J, Zhao Y, Wang K, Shi X, Wang Y, Huang H, et al. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature. 2015;526(7575):660–5. doi: http://doi.org/10.1038/nature15514. PubMed PMID: 26375003.
https://doi.org/10.1038/nature15514...
. Through its PYD domain, this protein binds to NLRP3, and on the opposite side, the presence of its CARD subdomain allows the recruitment of caspase-1 (via CARD-CARD interaction). After this interaction, oligomerization of the NLRP3 complex occurs with autocleavage of the caspase-1 enzyme and subsequent activation of IL-1β and IL-18. Furthermore, cell death occurs mediated by gasdermin-D (GSDMD) activation, a process known as pyroptosis6565. Shi J, Zhao Y, Wang K, Shi X, Wang Y, Huang H, et al. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature. 2015;526(7575):660–5. doi: http://doi.org/10.1038/nature15514. PubMed PMID: 26375003.
https://doi.org/10.1038/nature15514...
,6666. Shahzad K, Bock F, Dong W, Wang H, Kopf S, Kohli S, et al. Nlrp3-inflammasome activation in non-myeloid-derived cells aggravates diabetic nephropathy. Kidney Int. 2015;87(1):74–84. doi: http://doi.org/10.1038/ki.2014.271. PubMed PMID: 25075770.
https://doi.org/10.1038/ki.2014.271...
.

The activation of the NLRP3 inflammasome requires two complementary mechanisms. The first mechanism is related to the stimulation of PRRs located in the cell membrane through PAMPs or DAMPs, which triggers an intracellular signaling cascade that culminates in the activation and translocation of the transcription factor NF-kB to the cell nucleus6767. Schroder K, Zhou R, Tschopp J. The NLRP3 inflammasome: a sensor for metabolic danger? Science. 2010;327(5963):296–300. doi: http://doi.org/10.1126/science.1184003. PubMed PMID: 20075245.
https://doi.org/10.1126/science.1184003...
. When NF-kB enters the nucleus, it stimulates the transcription of pro-IL-1β, pro-IL-18 and the expression of the NLRP3 enzyme6464. Yang Y, Wang H, Kouadir M, Song H, Shi F. Recent advances in the mechanisms of NLRP3 inflammasome activation and its inhibitors. Cell Death Dis. 2019;10(2):128. doi: http://doi.org/10.1038/s41419-019-1413-8. PubMed PMID: 30755589.
https://doi.org/10.1038/s41419-019-1413-...
.

This is followed by the second activation signal, which is the cleavage of precursors previously activated by NF-kB in the cell nucleus6868. Cookson BT, Brennan MA. Pro-inflammatory programmed cell death. Trends Microbiol. 2001;9(3):113–4. doi: http://doi.org/10.1016/S0966-842X(00)01936-3. PubMed PMID: 11303500.
https://doi.org/10.1016/S0966-842X(00)01...
,6969. Zhang C, Boini KM, Xia M, Abais JM, Li X, Liu Q, et al. Activation of Nod-like receptor protein 3 inflammasomes turns on podocyte injury and glomerular sclerosis in hyperhomocysteinemia. Hypertension. 2012;60(1):154–62. doi: http://doi.org/10.1161/HYPERTENSIONAHA.111.189688. PubMed PMID: 22647887.
https://doi.org/10.1161/HYPERTENSIONAHA....
. This is triggered by the presence of DAMPs or other stimuli, such as an increase in the concentration of ROS, extracellular ATP, potassium efflux, uric acid crystals and nuclear proteins such as HMGB16969. Zhang C, Boini KM, Xia M, Abais JM, Li X, Liu Q, et al. Activation of Nod-like receptor protein 3 inflammasomes turns on podocyte injury and glomerular sclerosis in hyperhomocysteinemia. Hypertension. 2012;60(1):154–62. doi: http://doi.org/10.1161/HYPERTENSIONAHA.111.189688. PubMed PMID: 22647887.
https://doi.org/10.1161/HYPERTENSIONAHA....
,7070. Zhou R, Tardivel A, Thorens B, Choi I, Tschopp J. Thioredoxin-interacting protein links oxidative stress to inflammasome activation. Nat Immunol. 2010;11(2):136–40. doi: http://doi.org/10.1038/ni.1831. PubMed PMID: 20023662.
https://doi.org/10.1038/ni.1831...
. These molecules can activate and oligomerize the NLRP3 complex, involving the recruitment and maturation of caspase enzymes, which, when performing a self-cleavage process, activate IL-1β and IL-18, which are released into the extracellular environment via channels created in the cytoplasmic membrane7171. Turner CM, Arulkumaran N, Singer M, Unwin RJ, Tam FW. Is the inflammasome a potential therapeutic target in kidney disease? BMC Nephrol. 2014;15(1):21. doi: http://doi.org/10.1186/1471-2369-15-21. PubMed PMID: 24450291.
https://doi.org/10.1186/1471-2369-15-21...
,7272. El-Horany HE, Abd-Ellatif RN, Watany M, Hafez YM, Okda HI. NLRP3 expression and urinary HSP72 in relation to biomarkers of inflammation and oxidative stress in diabetic nephropathy patients. IUBMB Life. 2017;69(8):623–30. doi: http://doi.org/10.1002/iub.1645. PubMed PMID: 28631886.
https://doi.org/10.1002/iub.1645...
to exert their pro-inflammatory action. In addition, GSDMD is cleaved and its N-terminal subdomain is released, which penetrates the plasma membrane of the cell forming pores. This process, known as pyroptosis, is a form of programmed cell death that generates an intense inflammatory response by allowing cell osmolysis, DNA lysis and the release of cell components and inflammatory mediators such as IL-1β and IL-186767. Schroder K, Zhou R, Tschopp J. The NLRP3 inflammasome: a sensor for metabolic danger? Science. 2010;327(5963):296–300. doi: http://doi.org/10.1126/science.1184003. PubMed PMID: 20075245.
https://doi.org/10.1126/science.1184003...
,6868. Cookson BT, Brennan MA. Pro-inflammatory programmed cell death. Trends Microbiol. 2001;9(3):113–4. doi: http://doi.org/10.1016/S0966-842X(00)01936-3. PubMed PMID: 11303500.
https://doi.org/10.1016/S0966-842X(00)01...
, 6969. Zhang C, Boini KM, Xia M, Abais JM, Li X, Liu Q, et al. Activation of Nod-like receptor protein 3 inflammasomes turns on podocyte injury and glomerular sclerosis in hyperhomocysteinemia. Hypertension. 2012;60(1):154–62. doi: http://doi.org/10.1161/HYPERTENSIONAHA.111.189688. PubMed PMID: 22647887.
https://doi.org/10.1161/HYPERTENSIONAHA....
.

The Role of the NLRP3 Complex in Diabetic Kidney Disease

Despite being more commonly found in cells of the immune system, reports have already shown that the constituent molecules of inflammasomes are also present in other cells of the body, e.g. podocytes and mesangial cells in the kidney system7272. El-Horany HE, Abd-Ellatif RN, Watany M, Hafez YM, Okda HI. NLRP3 expression and urinary HSP72 in relation to biomarkers of inflammation and oxidative stress in diabetic nephropathy patients. IUBMB Life. 2017;69(8):623–30. doi: http://doi.org/10.1002/iub.1645. PubMed PMID: 28631886.
https://doi.org/10.1002/iub.1645...
,7373. Ozaki E, Campbell M, Doyle SL. Targeting the NLRP3 inflammasome in chronic inflammatory diseases: current perspectives. J Inflamm Res. 2015;8:15–27. PubMed PMID: 25653548.. The NLRP3 subtype is most abundant in the kidneys, and activation of this complex is involved in the development and progression of DKD7272. El-Horany HE, Abd-Ellatif RN, Watany M, Hafez YM, Okda HI. NLRP3 expression and urinary HSP72 in relation to biomarkers of inflammation and oxidative stress in diabetic nephropathy patients. IUBMB Life. 2017;69(8):623–30. doi: http://doi.org/10.1002/iub.1645. PubMed PMID: 28631886.
https://doi.org/10.1002/iub.1645...
,7474. Fang L, Xie D, Wu X, Cao H, Su W, Yang J. Involvement of endoplasmic reticulum stress in albuminuria induced inflammasome activation in kidney proximal tubular cells. PLoS One. 2013;8(8):e72344. doi: http://doi.org/10.1371/journal.pone.0072344. PubMed PMID: 23977286.
https://doi.org/10.1371/journal.pone.007...
.

The discovery of the NLRP3 complex as an inflammatory component involved in the pathogenesis of DKD has led to the development of therapies aimed at inhibiting its activity to attenuate kidney injury7373. Ozaki E, Campbell M, Doyle SL. Targeting the NLRP3 inflammasome in chronic inflammatory diseases: current perspectives. J Inflamm Res. 2015;8:15–27. PubMed PMID: 25653548.,7474. Fang L, Xie D, Wu X, Cao H, Su W, Yang J. Involvement of endoplasmic reticulum stress in albuminuria induced inflammasome activation in kidney proximal tubular cells. PLoS One. 2013;8(8):e72344. doi: http://doi.org/10.1371/journal.pone.0072344. PubMed PMID: 23977286.
https://doi.org/10.1371/journal.pone.007...
,7575. Wu M, Han W, Song S, Du Y, Liu C, Chen N, et al. NLRP3 deficiency ameliorates kidney inflammation and fibrosis in diabetic mice. Mol Cell Endocrinol. 2018;478:115–25. doi: http://doi.org/10.1016/j.mce.2018.08.002. PubMed PMID: 30098377.
https://doi.org/10.1016/j.mce.2018.08.00...
. In the context of DKD, modulation of the NLRP3 complex has been shown to prevent inflammation and slow the progression of fibrosis6767. Schroder K, Zhou R, Tschopp J. The NLRP3 inflammasome: a sensor for metabolic danger? Science. 2010;327(5963):296–300. doi: http://doi.org/10.1126/science.1184003. PubMed PMID: 20075245.
https://doi.org/10.1126/science.1184003...
. For example, knockout of the NLRP3 enzyme reduced inflammation and kidney fibrosis in diabetic mice7575. Wu M, Han W, Song S, Du Y, Liu C, Chen N, et al. NLRP3 deficiency ameliorates kidney inflammation and fibrosis in diabetic mice. Mol Cell Endocrinol. 2018;478:115–25. doi: http://doi.org/10.1016/j.mce.2018.08.002. PubMed PMID: 30098377.
https://doi.org/10.1016/j.mce.2018.08.00...
and knockout of the ASC enzyme attenuated proteinuria and glomerular damage in mice on a high-fat diet7676. Boini KM, Xia M, Abais JM, Li G, Pitzer AL, Gehr TW, et al. Activation of inflammasomes in podocyte injury of mice on the high fat diet: effects of ASC gene deletion and silencing. Biochim Biophys Acta. 2014;1843(5):836–45. doi: http://doi.org/10.1016/j.bbamcr.2014.01.033. PubMed PMID: 24508291.
https://doi.org/10.1016/j.bbamcr.2014.01...
. Thus, attenuating the inflammatory response by modulating the NLRP3 complex has become a new therapeutic strategy in the context of kidney diseases, especially those related to DM, aiming to reduce disease progression by reducing the inflammatory response mediated by the NLRP3 complex.

One of the most important studies involving the relationship between the NLRP3 complex and DKD was conducted in 2015 by Shahzad et al. The study showed that knockout of the NLPR3 enzyme or caspase-1 in diabetic mice was able to reduce albuminuria and extracellular matrix accumulation in these animals, but without changing glycemic levels or body weight6666. Shahzad K, Bock F, Dong W, Wang H, Kopf S, Kohli S, et al. Nlrp3-inflammasome activation in non-myeloid-derived cells aggravates diabetic nephropathy. Kidney Int. 2015;87(1):74–84. doi: http://doi.org/10.1038/ki.2014.271. PubMed PMID: 25075770.
https://doi.org/10.1038/ki.2014.271...
. The same study also showed that circulating levels of IL-1β and IL-18 and kidney expression of NALP3 were elevated in DKD mice and that this increase preceded the process of albuminuria and mesangial expansion, implying that NLRP3 activation could be a trigger for DKD. Additionally, pharmacological inhibition of the IL-1 receptor was able to reduce albuminuria and mesangial expansion in diabetic mice.

Wu et al. demonstrated in 2018 that knocking out the NLRP3 enzyme in diabetic mice also protected against the progression of diabetic kidney disease. This resulted in improved creatinine clearance and urinary albumin/creatinine ratio, as well as histopathological consequences such as improved glomerular hypertrophy and expanded mesangial and interstitial fibrosis compared to the control group7575. Wu M, Han W, Song S, Du Y, Liu C, Chen N, et al. NLRP3 deficiency ameliorates kidney inflammation and fibrosis in diabetic mice. Mol Cell Endocrinol. 2018;478:115–25. doi: http://doi.org/10.1016/j.mce.2018.08.002. PubMed PMID: 30098377.
https://doi.org/10.1016/j.mce.2018.08.00...
.

In the same line, several drugs from the oral antidiabetic class have demonstrated potential to inhibit the inflammatory response and modulate the NLRP3 complex pathway in diabetic patients and in animal models. Examples include sulfonylureas, biguanides, glitazones, the glucagon-like peptide 1 (RA-GLP-1) receptor agonists (albiglutide, dulaglutide and exenatide), DPP4 inhibitors and, more recently, SGTL2 inhibitors7777. Yaribeygi H, Katsiki N, Butler AE, Sahebkar A. Effects of antidiabetic drugs on NLRP3 inflammasome activity, with a focus on diabetic kidneys. Drug Discov Today. 2019;24(1):256–62. doi: http://doi.org/10.1016/j.drudis.2018.08.005. PubMed PMID: 30086405.
https://doi.org/10.1016/j.drudis.2018.08...
,7878. Panchapakesan U, Pegg K, Gross S, Komala MG, Mudaliar H, Forbes J, et al. Effects of SGLT2 inhibition in human kidney proximal tubular cells--renoprotection in diabetic nephropathy? PLoS One. 2013;8(2):e54442. doi: http://doi.org/10.1371/journal.pone.0054442. PubMed PMID: 23390498.
https://doi.org/10.1371/journal.pone.005...
.

Chronic glycemic exposure of kidney tubule cells causes changes in cellular metabolism, including increased expression of pro-inflammatory cytokines, growth and pro-fibrotic factors and reactive oxygen species. Thus, inhibition of glucose reabsorption by proximal tubule cells may be one mechanism of action by which SGLT2i reduces these adverse effects7979. De Nicola L, Gabbai FB, Liberti ME, Sagliocca A, Conte G, Minutolo R. Sodium/glucose cotransporter 2 inhibitors and prevention of diabetic nephropathy: targeting the kidney tubule in diabetes. Am J Kidney Dis. 2014;64(1):16–24. doi: http://doi.org/10.1053/j.ajkd.2014.02.010. PubMed PMID: 24673844.
https://doi.org/10.1053/j.ajkd.2014.02.0...
.

SGLT2 Inhibitors and the Inflammatory Response

Recent studies have shown that SGLT2 inhibitors may inhibit activation of the NLRP3 inflammasome in several animal models, including obesity, lung injury, myocardial infarction, DKD, depression and atherosclerosis. This inhibition is thought to occur through a number of mechanisms, including reduction of glucose uptake and oxidative stress, and modulation of the gut microbiome. The main articles investigating the possible relationship between SGLTi and the NLRP3 complex and their findings, are shown in Table 1.

Table 1
Studies evaluating the possible relationship between SGLT2I and NLRP3 complex

In the context of DKD, modulation of the inflammatory response has been observed in different animal models using SGLT2i, which attenuated the progression of kidney disease8080. Gembardt F, Bartaun C, Jarzebska N, Mayoux E, Todorov VT, Hohenstein B, et al. 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. 2014;307(3):F317–25. doi: http://doi.org/10.1152/ajprenal.00145.2014. PubMed PMID: 24944269.
https://doi.org/10.1152/ajprenal.00145.2...
,8181. Vallon V, Gerasimova M, Rose MA, Masuda T, Satriano J, Mayoux E, et al. SGLT2 inhibitor empagliflozin reduces kidney growth and albuminuria in proportion to hyperglycemia and prevents glomerular hyperfiltration in diabetic Akita mice. Am J Physiol Renal Physiol. 2014;306(2):F194–204. doi: http://doi.org/10.1152/ajprenal.00520.2013. PubMed PMID: 24226524.
https://doi.org/10.1152/ajprenal.00520.2...
,8282. Tahara A, Kurosaki E, Yokono M, Yamajuku D, Kihara R, Hayashizaki Y, et al. Effects of sodium-glucose cotransporter 2 selective inhibitor ipragliflozin on hyperglycaemia, oxidative stress, inflammation and liver injury in streptozotocin-induced type 1 diabetic rats. J Pharm Pharmacol. 2014;66(7):975–87. doi: http://doi.org/10.1111/jphp.12223. PubMed PMID: 24533859.
https://doi.org/10.1111/jphp.12223...
. The use of SGLT2i has already demonstrated glycemic benefits and a reduction of pro-inflammatory markers8282. Tahara A, Kurosaki E, Yokono M, Yamajuku D, Kihara R, Hayashizaki Y, et al. Effects of sodium-glucose cotransporter 2 selective inhibitor ipragliflozin on hyperglycaemia, oxidative stress, inflammation and liver injury in streptozotocin-induced type 1 diabetic rats. J Pharm Pharmacol. 2014;66(7):975–87. doi: http://doi.org/10.1111/jphp.12223. PubMed PMID: 24533859.
https://doi.org/10.1111/jphp.12223...
through inhibition of macrophage activity8383. Xu L, Nagata N, Nagashimada M, Zhuge F, Ni Y, Chen G, et al. SGLT2 inhibition by empagliflozin promotes fat utilization and browning and attenuates inflammation and insulin resistance by polarizing m2 macrophages in diet-induced obese mice. EBioMedicine. 2017;20:137–49. doi: http://doi.org/10.1016/j.ebiom.2017.05.028. PubMed PMID: 28579299.
https://doi.org/10.1016/j.ebiom.2017.05....
, suppression of molecular pathways involving PPRs7878. Panchapakesan U, Pegg K, Gross S, Komala MG, Mudaliar H, Forbes J, et al. Effects of SGLT2 inhibition in human kidney proximal tubular cells--renoprotection in diabetic nephropathy? PLoS One. 2013;8(2):e54442. doi: http://doi.org/10.1371/journal.pone.0054442. PubMed PMID: 23390498.
https://doi.org/10.1371/journal.pone.005...
,8181. Vallon V, Gerasimova M, Rose MA, Masuda T, Satriano J, Mayoux E, et al. SGLT2 inhibitor empagliflozin reduces kidney growth and albuminuria in proportion to hyperglycemia and prevents glomerular hyperfiltration in diabetic Akita mice. Am J Physiol Renal Physiol. 2014;306(2):F194–204. doi: http://doi.org/10.1152/ajprenal.00520.2013. PubMed PMID: 24226524.
https://doi.org/10.1152/ajprenal.00520.2...
,8484. Chen L, Klein T, Leung PS. Effects of combining linagliptin treatment with BI-38335, a novel SGLT2 inhibitor, on pancreatic islet function and inflammation in db/db mice. Curr Mol Med. 2012;12(8):995–1004. doi: http://doi.org/10.2174/156652412802480970. PubMed PMID: 22804249.
https://doi.org/10.2174/1566524128024809...
and reduction of oxidative stress8585. Tang L, Wu Y, Tian M, Sjostrom CD, Johansson U, Peng XR, et al. Dapagliflozin slows the progression of the kidney and liver fibrosis associated with type 2 diabetes. Am J Physiol Endocrinol Metab. 2017;313(5):E563–76. doi: http://doi.org/10.1152/ajpendo.00086.2017. PubMed PMID: 28811292.
https://doi.org/10.1152/ajpendo.00086.20...
.

Concerning the modulation of the NLRP3 complex in the context of DKD, the few studies that have evaluated this relationship have already demonstrated a reduction in the activity of the complex8686. Birnbaum Y, Bajaj M, Yang HC, Ye Y. Combined SGLT2 and DPP4 inhibition reduces the activation of the Nlrp3/ASC inflammasome and attenuates the development of diabetic nephropathy in mice with type 2 diabetes. Cardiovasc Drugs Ther. 2018;32(2):135–45. doi: http://doi.org/10.1007/s10557-018-6778-x. PubMed PMID: 29508169.
https://doi.org/10.1007/s10557-018-6778-...
, which has led to an improvement in glycemic levels and a reduction in the progression of kidney disease, with functional and histological advances5858. Ram C, Jha AK, Ghosh A, Gairola S, Syed AM, Murty US, et al. Targeting NLRP3 inflammasome as a promising approach for treatment of diabetic nephropathy: preclinical evidences with therapeutic approaches. Eur J Pharmacol. 2020;885:173503. doi: http://doi.org/10.1016/j.ejphar.2020.173503. PubMed PMID: 32858047.
https://doi.org/10.1016/j.ejphar.2020.17...
,8787. Benetti E, Mastrocola R, Vitarelli G, Cutrin JC, Nigro D, Chiazza F, et al. Empagliflozin protects against diet-induced NLRP-3 inflammasome activation and lipid accumulation. J Pharmacol Exp Ther. 2016;359(1):45–53. doi: http://doi.org/10.1124/jpet.116.235069. PubMed PMID: 27440421.
https://doi.org/10.1124/jpet.116.235069...
,8888. Ye Y, Bajaj M, Yang HC, Perez-Polo JR, Birnbaum Y. SGLT-2 inhibition with dapagliflozin reduces the activation of the Nlrp3/ASC inflammasome and attenuates the development of diabetic cardiomyopathy in mice with Type 2 Diabetes. Further Augmentation of the Effects with Saxagliptin, a DPP4 Inhibitor. Cardiovasc Drugs Ther. 2017;31(2):119–32. doi: http://doi.org/10.1007/s10557-017-6725-2. PubMed PMID: 28447181.
https://doi.org/10.1007/s10557-017-6725-...
. The full activation process of the NLRP3 inflammasome and the possible relationship with SGLT2i is shown in Figure 1.

Figure 1
NLRP3 inflammasome activation and the possible mechanisms of action of SGLT2i. The NLRP3 inflammasome activation relies on two key mechanisms. Firstly, NF-kB activation in the nucleus prompts the transcription of pro-IL-1β, pro-IL-18, and NLRP3 enzyme. Subsequently, a secondary signal triggers NLRP3 complex activation through cleavage of pre-activated precursors by stimuli like DAMPs or increased ROS concentration, leading to cytokine release and pyroptosis. SGLT2i potentially inhibits NF-kB, NLRP3, ASC, and Pro-caspase-1 activities, Gasdermin-D activation, and reduces IL-1β levels and mitochondrial reactive oxygen species (mROS).

In 2014, Tahara et al. demonstrated a positive impact on glycemic levels and a reduction of pro-inflammatory markers (IL-6 and TNF-α), chemokines, and oxidative stress with the use of ipragliflozin in a model of T1DM induced by streptozotocin in rats8282. Tahara A, Kurosaki E, Yokono M, Yamajuku D, Kihara R, Hayashizaki Y, et al. Effects of sodium-glucose cotransporter 2 selective inhibitor ipragliflozin on hyperglycaemia, oxidative stress, inflammation and liver injury in streptozotocin-induced type 1 diabetic rats. J Pharm Pharmacol. 2014;66(7):975–87. doi: http://doi.org/10.1111/jphp.12223. PubMed PMID: 24533859.
https://doi.org/10.1111/jphp.12223...
, and these benefits were also replicated in a model of T2DM induced by streptozomicin and a hypercaloric diet in mice8989. Tahara A, Kurosaki E, Yokono M, Yamajuku D, Kihara R, Hayashizaki Y, et al. Effects of SGLT2 selective inhibitor ipragliflozin on hyperglycemia, hyperlipidemia, hepatic steatosis, oxidative stress, inflammation, and obesity in type 2 diabetic mice. Eur J Pharmacol. 2013;715(1-3):246–55. doi: http://doi.org/10.1016/j.ejphar.2013.05.014. PubMed PMID: 23707905.
https://doi.org/10.1016/j.ejphar.2013.05...
.

In 2016, Benetti et al. evaluated for the first time the effects of an SGLT2 inhibitor on the NLRP3 complex in the context of metabolic syndrome in an animal model of diet-induced obesity and insulin resistance8787. Benetti E, Mastrocola R, Vitarelli G, Cutrin JC, Nigro D, Chiazza F, et al. Empagliflozin protects against diet-induced NLRP-3 inflammasome activation and lipid accumulation. J Pharmacol Exp Ther. 2016;359(1):45–53. doi: http://doi.org/10.1124/jpet.116.235069. PubMed PMID: 27440421.
https://doi.org/10.1124/jpet.116.235069...
. In this study, the authors demonstrated for the first time that empaglifozin treatment was able to improve glycemic levels and pathophysiological changes in metabolic syndrome, in addition to reducing NLRP3 complex activation, with IL-1β inhibition in a dose-dependent manner. Also, the authors found that animals treated with iSGTL2 showed fewer pathological changes in their histology, especially less tubular vacuolation, which is one of the first signs of kidney tubule degeneration.

In 2017, Ye et al. assessed the use of dapagliflozin in the interaction with the NLRP3 complex and the progression of diabetic cardiomyopathy in mice with type 2 diabetes8888. Ye Y, Bajaj M, Yang HC, Perez-Polo JR, Birnbaum Y. SGLT-2 inhibition with dapagliflozin reduces the activation of the Nlrp3/ASC inflammasome and attenuates the development of diabetic cardiomyopathy in mice with Type 2 Diabetes. Further Augmentation of the Effects with Saxagliptin, a DPP4 Inhibitor. Cardiovasc Drugs Ther. 2017;31(2):119–32. doi: http://doi.org/10.1007/s10557-017-6725-2. PubMed PMID: 28447181.
https://doi.org/10.1007/s10557-017-6725-...
. The findings revealed a reduction in pathological cardiac remodeling, a decrease in glycemic levels in glucose tolerance tests and a decrease in mRNA levels of NALP3, ASC, IL-1β, IL-6, and caspase-1. In 2018, Birnbaum et al. demonstrated that the use of dapagliflozin could attenuate the inflammatory response and thus kidney injury and glomerulosclerosis in diabetic rats by reducing the expression levels of ASC mRNA, caspase-1, IL-6, IL-1β and TNF-α8686. Birnbaum Y, Bajaj M, Yang HC, Ye Y. Combined SGLT2 and DPP4 inhibition reduces the activation of the Nlrp3/ASC inflammasome and attenuates the development of diabetic nephropathy in mice with type 2 diabetes. Cardiovasc Drugs Ther. 2018;32(2):135–45. doi: http://doi.org/10.1007/s10557-018-6778-x. PubMed PMID: 29508169.
https://doi.org/10.1007/s10557-018-6778-...
.

Future Perspectives

Studies involving the influence of SGLT2 inhibitors on the inflammasome complex have opened up new avenues for the treatment of DKD. However, there is still much inconclusive information. Therefore, future perspectives for studies on the influence of SGLT2 inhibitors on the inflammasome complex in DKD should include:

  1. Elucidation of the molecular mechanisms involved in the regulation of the inflammasome complex by SGLT2 inhibitors. Although studies have shown that SGLT2 inhibitors reduce NLRP3 inflammasome activation, the precise molecular mechanisms involved are not fully understood.

  2. The use of SGLT2 inhibitors in combination with other therapies. Currently available SGLT2 inhibitors have been shown to be effective in the treatment of DM. However, there is still room for improvement in terms of effectiveness and safety. Future research could focus on the clinical and molecular responses of the new SGLT2 inhibitors when combined with physical activity, specific diets, and nutritional supplements, among others.

  3. Clinical trials to evaluate the long-term effects of SGLT2 inhibitors on the progression of DKD. Although several studies have shown that SGLT2 inhibitors improve the inflammasome complex and kidney function in people with DKD, the long-term effects and timeframe of these medications in the progression of the disease are still not fully understood.

  4. Identification of biomarkers that predict response to SGLT2 inhibitors. Not all people with diabetic kidney disease respond to SGLT2 inhibitors. Biomarkers that predict response to these drugs can help identify individuals most likely to benefit from treatment.

In conclusion, the studies on the influence of SGLT2 inhibitors on the inflammasome complex have increased knowledge of the potential therapeutic effect in the treatment of DKD. Future studies could focus on elucidating the molecular mechanisms involved, evaluating the use of complementary therapies to achieve better efficacy and safety profiles, assessing the long-term effects of these drugs, and identifying biomarkers to predict response to such treatment.

Acknowledgments

This work was supported by Fundação Araucária-PR.

Data Availability

All datasets generated and analyzed during the current study are available from the corresponding author on reasonable request.

References

  • 1.
    Go AS, Chertow GM, Fan D, McCulloch CE, Hsu CY. Chronic kidney disease and the risks of death, cardiovascular events, and hospitalization. N Engl J Med. 2004;351(13):1296–305. doi: http://doi.org/10.1056/NEJMoa041031. PubMed PMID: 15385656.
    » https://doi.org/10.1056/NEJMoa041031
  • 2.
    GBD Chronic Kidney Disease Collaboration. Global, regional, and national burden of chronic kidney disease, 1990–2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet. 2020;395(10225):709–33. doi: http://doi.org/10.1016/S0140-6736(20)30045-3. PubMed PMID: 32061315.
    » https://doi.org/10.1016/S0140-6736(20)30045-3
  • 3.
    Webster AC, Nagler EV, Morton RL, Masson P. Chronic Kidney Disease. Lancet. 2017;389(10075):1238–52. doi: http://doi.org/10.1016/S0140-6736(16)32064-5. PubMed PMID: 27887750.
    » https://doi.org/10.1016/S0140-6736(16)32064-5
  • 4.
    Ogurtsova K, da Rocha Fernandes JD, Huang Y, Linnenkamp U, Guariguata L, Cho NH, et al. IDF Diabetes Atlas: global estimates for the prevalence of diabetes for 2015 and 2040. Diabetes Res Clin Pract. 2017;128:40–50. doi: http://doi.org/10.1016/j.diabres.2017.03.024. PubMed PMID: 28437734.
    » https://doi.org/10.1016/j.diabres.2017.03.024
  • 5.
    Bentata Y, Haddiya I, Latrech H, Serraj K, Abouqal R. Progression of diabetic nephropathy, risk of end-stage kidney disease and mortality in patients with type-1 diabetes. Saudi J Kidney Dis Transpl. 2013;24(2):392–402. doi: http://doi.org/10.4103/1319-2442.109617. PubMed PMID: 23538374.
    » https://doi.org/10.4103/1319-2442.109617
  • 6.
    Reidy K, Kang HM, Hostetter T, Susztak K. Molecular mechanisms of diabetic kidney disease. J Clin Invest. 2014;124(6):2333–40. doi: http://doi.org/10.1172/JCI72271. PubMed PMID: 24892707.
    » https://doi.org/10.1172/JCI72271
  • 7.
    Denic A, Mathew J, Lerman LO, Lieske JC, Larson JJ, Alexander MP, et al. Single-nephron glomerular filtration rate in healthy adults. N Engl J Med. 2017;376(24):2349–57. doi: http://doi.org/10.1056/NEJMoa1614329. PubMed PMID: 28614683.
    » https://doi.org/10.1056/NEJMoa1614329
  • 8.
    Penno G, Orsi E, Solini A, Bonora E, Fondelli C, Trevisan R, et al. Kidney hyperfiltration is independently associated with increased all-cause mortality in individuals with type 2 diabetes: a prospective cohort study. BMJ Open Diabetes Res Care. 2020;8(1):e001481. doi: http://doi.org/10.1136/bmjdrc-2020-001481. PubMed PMID: 32665314.
    » https://doi.org/10.1136/bmjdrc-2020-001481
  • 9.
    Minutolo R, Gabbai FB, Provenzano M, Chiodini P, Borrelli S, Garofalo C, et al. Cardiokidney prognosis by residual proteinuria level in diabetic chronic kidney disease: pooled analysis of four cohort studies. Nephrol Dial Transplant. 2018;33(11):1942–9. doi: http://doi.org/10.1093/ndt/gfy032. PubMed PMID: 29509925.
    » https://doi.org/10.1093/ndt/gfy032
  • 10.
    Guedes M, Pecoits-Filho R. Can we cure diabetic kidney disease? Present and future perspectives from a nephrologist’s point of view. J Intern Med. 2022;291(2):165–80. doi: http://doi.org/10.1111/joim.13424. PubMed PMID: 34914852.
    » https://doi.org/10.1111/joim.13424
  • 11.
    ElSayed NA, Aleppo G, Aroda VR, Bannuru RR, Brown FM, Bruemmer D, et al. Pharmacologic approaches to glycemic treatment: standards of care in diabetes-2023. Diabetes Care. 2023;46(Suppl 1):S140–57. doi: http://doi.org/10.2337/dc23-S009. PubMed PMID: 36507650.
    » https://doi.org/10.2337/dc23-S009
  • 12.
    Lewis EJ, Hunsicker LG, Bain RP, Rohde RD, and the The Collaborative Study Group. The effect of angiotensin-converting-enzyme inhibition on diabetic nephropathy. N Engl J Med. 1993;329(20):1456–62. doi: http://doi.org/10.1056/NEJM199311113292004. PubMed PMID: 8413456.
    » https://doi.org/10.1056/NEJM199311113292004
  • 13.
    Brenner BM, Cooper ME, de Zeeuw D, Keane WF, Mitch WE, Parving HH, et al. Effects of losartan on kidney and cardiovascular outcomes in patients with type 2 diabetes and nephropathy. N Engl J Med. 2001;345(12):861–9. doi: http://doi.org/10.1056/NEJMoa011161. PubMed PMID: 11565518.
    » https://doi.org/10.1056/NEJMoa011161
  • 14.
    Provenzano M, Coppolino G, De Nicola L, Serra R, Garofalo C, Andreucci M, et al. Unraveling cardiovascular risk in kidney patients: a new take on old tale. Front Cell Dev Biol. 2019;7:314. doi: http://doi.org/10.3389/fcell.2019.00314. PubMed PMID: 31850348.
    » https://doi.org/10.3389/fcell.2019.00314
  • 15.
    Provenzano M, De Nicola L, Pena MJ, Capitoli G, Garofalo C, Borrelli S, et al. Precision nephrology is a non-negligible state of mind in clinical research: remember the past to face the future. Nephron. 2020;144(10):463–78. doi: http://doi.org/10.1159/000508983. PubMed PMID: 32810859.
    » https://doi.org/10.1159/000508983
  • 16.
    Idris I, Donnelly R. Sodium-glucose co-transporter-2 inhibitors: an emerging new class of oral antidiabetic drug. Diabetes Obes Metab. 2009;11(2):79–88. doi: http://doi.org/10.1111/j.1463-1326.2008.00982.x. PubMed PMID: 19125776.
    » https://doi.org/10.1111/j.1463-1326.2008.00982.x
  • 17.
    Perkovic V, Jardine MJ, Neal B, Bompoint S, Heerspink HJL, Charytan DM, et al. Canagliflozin and kidney outcomes in type 2 diabetes and nephropathy. N Engl J Med. 2019;380(24):2295–306. doi: http://doi.org/10.1056/NEJMoa1811744. PubMed PMID: 30990260.
    » https://doi.org/10.1056/NEJMoa1811744
  • 18.
    Musso G, Gambino R, Cassader M, Pagano G. A novel approach to control hyperglycemia in type 2 diabetes: sodium glucose co-transport (SGLT) inhibitors: systematic review and meta-analysis of randomized trials. Ann Med. 2012;44(4):375–93. doi: http://doi.org/10.3109/07853890.2011.560181. PubMed PMID: 21495788.
    » https://doi.org/10.3109/07853890.2011.560181
  • 19.
    Cefalu WT. Paradoxical insights into whole body metabolic adaptations following SGLT2 inhibition. J Clin Invest. 2014;124(2):485–7. doi: http://doi.org/10.1172/JCI74297. PubMed PMID: 24463446.
    » https://doi.org/10.1172/JCI74297
  • 20.
    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(15):1436–46. doi: http://doi.org/10.1056/NEJMoa2024816. PubMed PMID: 32970396.
    » https://doi.org/10.1056/NEJMoa2024816
  • 21.
    Herrington WG, Staplin N, Wanner C, Green JB, Hauske SJ, Emberson JR, et al. Empagliflozin in Patients with Chronic Kidney Disease. N Engl J Med. 2023;388(2):117–27. doi: http://doi.org/10.1056/NEJMoa2204233. PubMed PMID: 36331190.
    » https://doi.org/10.1056/NEJMoa2204233
  • 22.
    Ehrenkranz JR, Lewis NG, Kahn CR, Roth J. Phlorizin: a review. Diabetes Metab Res Rev. 2005;21(1):31–8. doi: http://doi.org/10.1002/dmrr.532. PubMed PMID: 15624123.
    » https://doi.org/10.1002/dmrr.532
  • 23.
    Provenzano M, Pelle MC, Zaffina I, Tassone B, Pujia R, Ricchio M, et al. Sodium-glucose co-transporter-2 inhibitors and nephroprotection in diabetic patients: more than a challenge. Front Med (Lausanne). 2021;8:654557. doi: http://doi.org/10.3389/fmed.2021.654557. PubMed PMID: 34150796.
    » https://doi.org/10.3389/fmed.2021.654557
  • 24.
    Giugliano D, Esposito K. Class effect for SGLT-2 inhibitors: a tale of 9 drugs. Cardiovasc Diabetol. 2019;18(1):94. doi: http://doi.org/10.1186/s12933-019-0899-9. PubMed PMID: 31337395.
    » https://doi.org/10.1186/s12933-019-0899-9
  • 25.
    Garcia-Ropero A, Badimon JJ, Santos-Gallego CG. The pharmacokinetics and pharmacodynamics of SGLT2 inhibitors for type 2 diabetes mellitus: the latest developments. Expert Opin Drug Metab Toxicol. 2018;14(12):1287–302. doi: http://doi.org/10.1080/17425255.2018.1551877. PubMed PMID: 30463454.
    » https://doi.org/10.1080/17425255.2018.1551877
  • 26.
    Shubrook JH, Bokaie BB, Adkins SE. Empagliflozin in the treatment of type 2 diabetes: evidence to date. Drug Des Devel Ther. 2015;9:5793–803. doi: http://doi.org/10.2147/DDDT.S69926. PubMed PMID: 26586935.
    » https://doi.org/10.2147/DDDT.S69926
  • 27.
    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(22):2117–28. doi: http://doi.org/10.1056/NEJMoa1504720. PubMed PMID: 26378978.
    » https://doi.org/10.1056/NEJMoa1504720
  • 28.
    Ghezzi C, Loo DDF, Wright EM. Physiology of kidney glucose handling via SGLT1, SGLT2 and GLUT2. Diabetologia. 2018;61(10):2087–97. doi: http://doi.org/10.1007/s00125-018-4656-5. PubMed PMID: 30132032.
    » https://doi.org/10.1007/s00125-018-4656-5
  • 29.
    Liu JJ, Lee T, DeFronzo RA. Why Do SGLT2 inhibitors inhibit only 30-50% of kidney glucose reabsorption in humans? Diabetes. 2012;61(9):2199–204. doi: http://doi.org/10.2337/db12-0052. PubMed PMID: 22923645.
    » https://doi.org/10.2337/db12-0052
  • 30.
    Gallo LA, Wright EM, Vallon V. Probing SGLT2 as a therapeutic target for diabetes: basic physiology and consequences. Diab Vasc Dis Res. 2015;12(2):78–89. doi: http://doi.org/10.1177/1479164114561992. PubMed PMID: 25616707.
    » https://doi.org/10.1177/1479164114561992
  • 31.
    Thomas MC. Kidney effects of dapagliflozin in patients with type 2 diabetes. Ther Adv Endocrinol Metab. 2014;5(3):53–61. doi: http://doi.org/10.1177/2042018814544153. PubMed PMID: 25126408.
    » https://doi.org/10.1177/2042018814544153
  • 32.
    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(1):R75–83. doi: http://doi.org/10.1152/ajpregu.00357.2011. PubMed PMID: 21940401.
    » https://doi.org/10.1152/ajpregu.00357.2011
  • 33.
    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. 2021;320(5):F761–71. doi: http://doi.org/10.1152/ajprenal.00552.2020. PubMed PMID: 33645318.
    » https://doi.org/10.1152/ajprenal.00552.2020
  • 34.
    van Bommel EJM, Muskiet MHA, van Baar MJB, Tonneijck L, Smits MM, Emanuel AL, et al. The kidney 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(1):202–12. doi: http://doi.org/10.1016/j.kint.2019.09.013. PubMed PMID: 31791665.
    » https://doi.org/10.1016/j.kint.2019.09.013
  • 35.
    Thomas MC, Cherney DZI. The actions of SGLT2 inhibitors on metabolism, kidney function and blood pressure. Diabetologia. 2018;61(10):2098–107. doi: http://doi.org/10.1007/s00125-018-4669-0. PubMed PMID: 30132034.
    » https://doi.org/10.1007/s00125-018-4669-0
  • 36.
    Szekeres Z, Toth K, Szabados E. The Effects of SGLT2 Inhibitors on Lipid Metabolism. Metabolites. 2021;11(2):87. doi: http://doi.org/10.3390/metabo11020087. PubMed PMID: 33535652.
    » https://doi.org/10.3390/metabo11020087
  • 37.
    Hickey FB, Martin F. Role of the immune system in diabetic kidney disease. Curr Diab Rep. 2018;18(4):20. doi: http://doi.org/10.1007/s11892-018-0984-6. PubMed PMID: 29532281.
    » https://doi.org/10.1007/s11892-018-0984-6
  • 38.
    Kawanami D, Matoba K, Utsunomiya K. Signaling pathways in diabetic nephropathy. Histol Histopathol. 2016;31(10):1059–67. PubMed PMID: 27094540.
  • 39.
    Matoba K, Takeda Y, Nagai Y, Kawanami D, Utsunomiya K, Nishimura R. Unraveling the role of inflammation in the pathogenesis of diabetic kidney disease. Int J Mol Sci. 2019;20(14):3393. doi: http://doi.org/10.3390/ijms20143393. PubMed PMID: 31295940.
    » https://doi.org/10.3390/ijms20143393
  • 40.
    Komada T, Muruve DA. The role of inflammasomes in kidney disease. Nat Rev Nephrol. 2019;15(8):501–20. doi: http://doi.org/10.1038/s41581-019-0158-z. PubMed PMID: 31164720.
    » https://doi.org/10.1038/s41581-019-0158-z
  • 41.
    Jha JC, Banal C, Chow BS, Cooper ME, Jandeleit-Dahm K. Diabetes and kidney disease: role of oxidative stress. Antioxid Redox Signal. 2016;25(12):657–84. doi: http://doi.org/10.1089/ars.2016.6664. PubMed PMID: 26906673.
    » https://doi.org/10.1089/ars.2016.6664
  • 42.
    Volpe CMO, Villar-Delfino PH, Dos Anjos PMF, Nogueira-Machado JA. Cellular death, reactive oxygen species (ROS) and diabetic complications. Cell Death Dis. 2018;9(2):119. doi: http://doi.org/10.1038/s41419-017-0135-z. PubMed PMID: 29371661.
    » https://doi.org/10.1038/s41419-017-0135-z
  • 43.
    Singh DK, Winocour P, Farrington K. Oxidative stress in early diabetic nephropathy: fueling the fire. Nat Rev Endocrinol. 2011;7(3):176–84. doi: http://doi.org/10.1038/nrendo.2010.212. PubMed PMID: 21151200.
    » https://doi.org/10.1038/nrendo.2010.212
  • 44.
    Galkina E, Ley K. Leukocyte recruitment and vascular injury in diabetic nephropathy. J Am Soc Nephrol. 2006;17(2):368–77. doi: http://doi.org/10.1681/ASN.2005080859. PubMed PMID: 16394109.
    » https://doi.org/10.1681/ASN.2005080859
  • 45.
    Sester U, Sester M, Heine G, Kaul H, Girndt M, Kohler H. Strong depletion of CD14(+)CD16(+) monocytes during haemodialysis treatment. Nephrol Dial Transplant. 2001;16(7):1402–8. doi: http://doi.org/10.1093/ndt/16.7.1402. PubMed PMID: 11427632.
    » https://doi.org/10.1093/ndt/16.7.1402
  • 46.
    Alicic RZ, Johnson EJ, Tuttle KR. Inflammatory mechanisms as new biomarkers and therapeutic targets for diabetic kidney disease. Adv Chronic Kidney Dis. 2018;25(2):181–91. doi: http://doi.org/10.1053/j.ackd.2017.12.002. PubMed PMID: 29580582.
    » https://doi.org/10.1053/j.ackd.2017.12.002
  • 47.
    Awad AS, Kinsey GR, Khutsishvili K, Gao T, Bolton WK, Okusa MD. Monocyte/macrophage chemokine receptor CCR2 mediates diabetic kidney injury. Am J Physiol Renal Physiol. 2011;301(6):F1358–66. doi: http://doi.org/10.1152/ajprenal.00332.2011. PubMed PMID: 21880831.
    » https://doi.org/10.1152/ajprenal.00332.2011
  • 48.
    Lim AK, Ma FY, Nikolic-Paterson DJ, Kitching AR, Thomas MC, Tesch GH. Lymphocytes promote albuminuria, but not kidney dysfunction or histological damage in a mouse model of diabetic kidney injury. Diabetologia. 2010;53(8):1772–82. doi: http://doi.org/10.1007/s00125-010-1757-1. PubMed PMID: 20422398.
    » https://doi.org/10.1007/s00125-010-1757-1
  • 49.
    Furuta T, Saito T, Ootaka T, Soma J, Obara K, Abe K, et al. The role of macrophages in diabetic glomerulosclerosis. Am J Kidney Dis. 1993;21(5):480–5. doi: http://doi.org/10.1016/S0272-6386(12)80393-3. PubMed PMID: 8488815.
    » https://doi.org/10.1016/S0272-6386(12)80393-3
  • 50.
    Nguyen D, Ping F, Mu W, Hill P, Atkins RC, Chadban SJ. Macrophage accumulation in human progressive diabetic nephropathy. Nephrology (Carlton). 2006;11(3):226–31. doi: http://doi.org/10.1111/j.1440-1797.2006.00576.x. PubMed PMID: 16756636.
    » https://doi.org/10.1111/j.1440-1797.2006.00576.x
  • 51.
    Ninichuk V, Khandoga AG, Segerer S, Loetscher P, Schlapbach A, Revesz L, et al. The role of interstitial macrophages in nephropathy of type 2 diabetic db/db mice. Am J Pathol. 2007;170(4):1267–76. doi: http://doi.org/10.2353/ajpath.2007.060937. PubMed PMID: 17392166.
    » https://doi.org/10.2353/ajpath.2007.060937
  • 52.
    Klessens CQF, Zandbergen M, Wolterbeek R, Bruijn JA, Rabelink TJ, Bajema IM, et al. Macrophages in diabetic nephropathy in patients with type 2 diabetes. Nephrol Dial Transplant. 2017;32(8):1322–9. PubMed PMID: 27416772.
  • 53.
    Tang SC, Chan LY, Leung JC, Cheng AS, Chan KW, Lan HY, et al. Bradykinin and high glucose promote kidney tubular inflammation. Nephrol Dial Transplant. 2010;25(3):698–710. doi: http://doi.org/10.1093/ndt/gfp599. PubMed PMID: 19923143.
    » https://doi.org/10.1093/ndt/gfp599
  • 54.
    Kang YS, Lee MH, Song HK, Ko GJ, Kwon OS, Lim TK, et al. CCR2 antagonism improves insulin resistance, lipid metabolism, and diabetic nephropathy in type 2 diabetic mice. Kidney Int. 2010;78(9):883–94. doi: http://doi.org/10.1038/ki.2010.263. PubMed PMID: 20686445.
    » https://doi.org/10.1038/ki.2010.263
  • 55.
    Kumar H, Kawai T, Akira S. Pathogen recognition by the innate immune system. Int Rev Immunol. 2011;30(1):16–34. doi: http://doi.org/10.3109/08830185.2010.529976. PubMed PMID: 21235323.
    » https://doi.org/10.3109/08830185.2010.529976
  • 56.
    Tesch GH. Diabetic nephropathy - is this an immune disorder? Clin Sci (Lond). 2017;131(16):2183–99. doi: http://doi.org/10.1042/CS20160636. PubMed PMID: 28760771.
    » https://doi.org/10.1042/CS20160636
  • 57.
    Wada J, Makino H. Inflammation and the pathogenesis of diabetic nephropathy. Clin Sci (Lond). 2013;124(3):139–52. doi: http://doi.org/10.1042/CS20120198. PubMed PMID: 23075333.
    » https://doi.org/10.1042/CS20120198
  • 58.
    Ram C, Jha AK, Ghosh A, Gairola S, Syed AM, Murty US, et al. Targeting NLRP3 inflammasome as a promising approach for treatment of diabetic nephropathy: preclinical evidences with therapeutic approaches. Eur J Pharmacol. 2020;885:173503. doi: http://doi.org/10.1016/j.ejphar.2020.173503. PubMed PMID: 32858047.
    » https://doi.org/10.1016/j.ejphar.2020.173503
  • 59.
    Brubaker SW, Bonham KS, Zanoni I, Kagan JC. Innate immune pattern recognition: a cell biological perspective. Annu Rev Immunol. 2015;33(1):257–90. doi: http://doi.org/10.1146/annurev-immunol-032414-112240. PubMed PMID: 25581309.
    » https://doi.org/10.1146/annurev-immunol-032414-112240
  • 60.
    Takeuchi O, Akira S. Pattern recognition receptors and inflammation. Cell. 2010;140(6):805-20. doi: http://doi.org/10.1016/j.cell.2010.01.022. PubMed PMID: 20303872.
    » https://doi.org/10.1016/j.cell.2010.01.022
  • 61.
    Kanneganti TD, Lamkanfi M, Nunez G. Intracellular NOD-like receptors in host defense and disease. Immunity. 2007;27(4):549–59. doi: http://doi.org/10.1016/j.immuni.2007.10.002. PubMed PMID: 17967410.
    » https://doi.org/10.1016/j.immuni.2007.10.002
  • 62.
    Barbe F, Douglas T, Saleh M. Advances in Nod-like receptors (NLR) biology. Cytokine Growth Factor Rev. 2014;25(6):681–97. doi: http://doi.org/10.1016/j.cytogfr.2014.07.001. PubMed PMID: 25070125.
    » https://doi.org/10.1016/j.cytogfr.2014.07.001
  • 63.
    Kim SM, Lee SH, Kim YG, Kim SY, Seo JW, Choi YW, et al. Hyperuricemia-induced NLRP3 activation of macrophages contributes to the progression of diabetic nephropathy. Am J Physiol Renal Physiol. 2015;308(9):F993–1003. doi: http://doi.org/10.1152/ajprenal.00637.2014. PubMed PMID: 25651569.
    » https://doi.org/10.1152/ajprenal.00637.2014
  • 64.
    Yang Y, Wang H, Kouadir M, Song H, Shi F. Recent advances in the mechanisms of NLRP3 inflammasome activation and its inhibitors. Cell Death Dis. 2019;10(2):128. doi: http://doi.org/10.1038/s41419-019-1413-8. PubMed PMID: 30755589.
    » https://doi.org/10.1038/s41419-019-1413-8
  • 65.
    Shi J, Zhao Y, Wang K, Shi X, Wang Y, Huang H, et al. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature. 2015;526(7575):660–5. doi: http://doi.org/10.1038/nature15514. PubMed PMID: 26375003.
    » https://doi.org/10.1038/nature15514
  • 66.
    Shahzad K, Bock F, Dong W, Wang H, Kopf S, Kohli S, et al. Nlrp3-inflammasome activation in non-myeloid-derived cells aggravates diabetic nephropathy. Kidney Int. 2015;87(1):74–84. doi: http://doi.org/10.1038/ki.2014.271. PubMed PMID: 25075770.
    » https://doi.org/10.1038/ki.2014.271
  • 67.
    Schroder K, Zhou R, Tschopp J. The NLRP3 inflammasome: a sensor for metabolic danger? Science. 2010;327(5963):296–300. doi: http://doi.org/10.1126/science.1184003. PubMed PMID: 20075245.
    » https://doi.org/10.1126/science.1184003
  • 68.
    Cookson BT, Brennan MA. Pro-inflammatory programmed cell death. Trends Microbiol. 2001;9(3):113–4. doi: http://doi.org/10.1016/S0966-842X(00)01936-3. PubMed PMID: 11303500.
    » https://doi.org/10.1016/S0966-842X(00)01936-3
  • 69.
    Zhang C, Boini KM, Xia M, Abais JM, Li X, Liu Q, et al. Activation of Nod-like receptor protein 3 inflammasomes turns on podocyte injury and glomerular sclerosis in hyperhomocysteinemia. Hypertension. 2012;60(1):154–62. doi: http://doi.org/10.1161/HYPERTENSIONAHA.111.189688. PubMed PMID: 22647887.
    » https://doi.org/10.1161/HYPERTENSIONAHA.111.189688
  • 70.
    Zhou R, Tardivel A, Thorens B, Choi I, Tschopp J. Thioredoxin-interacting protein links oxidative stress to inflammasome activation. Nat Immunol. 2010;11(2):136–40. doi: http://doi.org/10.1038/ni.1831. PubMed PMID: 20023662.
    » https://doi.org/10.1038/ni.1831
  • 71.
    Turner CM, Arulkumaran N, Singer M, Unwin RJ, Tam FW. Is the inflammasome a potential therapeutic target in kidney disease? BMC Nephrol. 2014;15(1):21. doi: http://doi.org/10.1186/1471-2369-15-21. PubMed PMID: 24450291.
    » https://doi.org/10.1186/1471-2369-15-21
  • 72.
    El-Horany HE, Abd-Ellatif RN, Watany M, Hafez YM, Okda HI. NLRP3 expression and urinary HSP72 in relation to biomarkers of inflammation and oxidative stress in diabetic nephropathy patients. IUBMB Life. 2017;69(8):623–30. doi: http://doi.org/10.1002/iub.1645. PubMed PMID: 28631886.
    » https://doi.org/10.1002/iub.1645
  • 73.
    Ozaki E, Campbell M, Doyle SL. Targeting the NLRP3 inflammasome in chronic inflammatory diseases: current perspectives. J Inflamm Res. 2015;8:15–27. PubMed PMID: 25653548.
  • 74.
    Fang L, Xie D, Wu X, Cao H, Su W, Yang J. Involvement of endoplasmic reticulum stress in albuminuria induced inflammasome activation in kidney proximal tubular cells. PLoS One. 2013;8(8):e72344. doi: http://doi.org/10.1371/journal.pone.0072344. PubMed PMID: 23977286.
    » https://doi.org/10.1371/journal.pone.0072344
  • 75.
    Wu M, Han W, Song S, Du Y, Liu C, Chen N, et al. NLRP3 deficiency ameliorates kidney inflammation and fibrosis in diabetic mice. Mol Cell Endocrinol. 2018;478:115–25. doi: http://doi.org/10.1016/j.mce.2018.08.002. PubMed PMID: 30098377.
    » https://doi.org/10.1016/j.mce.2018.08.002
  • 76.
    Boini KM, Xia M, Abais JM, Li G, Pitzer AL, Gehr TW, et al. Activation of inflammasomes in podocyte injury of mice on the high fat diet: effects of ASC gene deletion and silencing. Biochim Biophys Acta. 2014;1843(5):836–45. doi: http://doi.org/10.1016/j.bbamcr.2014.01.033. PubMed PMID: 24508291.
    » https://doi.org/10.1016/j.bbamcr.2014.01.033
  • 77.
    Yaribeygi H, Katsiki N, Butler AE, Sahebkar A. Effects of antidiabetic drugs on NLRP3 inflammasome activity, with a focus on diabetic kidneys. Drug Discov Today. 2019;24(1):256–62. doi: http://doi.org/10.1016/j.drudis.2018.08.005. PubMed PMID: 30086405.
    » https://doi.org/10.1016/j.drudis.2018.08.005
  • 78.
    Panchapakesan U, Pegg K, Gross S, Komala MG, Mudaliar H, Forbes J, et al. Effects of SGLT2 inhibition in human kidney proximal tubular cells--renoprotection in diabetic nephropathy? PLoS One. 2013;8(2):e54442. doi: http://doi.org/10.1371/journal.pone.0054442. PubMed PMID: 23390498.
    » https://doi.org/10.1371/journal.pone.0054442
  • 79.
    De Nicola L, Gabbai FB, Liberti ME, Sagliocca A, Conte G, Minutolo R. Sodium/glucose cotransporter 2 inhibitors and prevention of diabetic nephropathy: targeting the kidney tubule in diabetes. Am J Kidney Dis. 2014;64(1):16–24. doi: http://doi.org/10.1053/j.ajkd.2014.02.010. PubMed PMID: 24673844.
    » https://doi.org/10.1053/j.ajkd.2014.02.010
  • 80.
    Gembardt F, Bartaun C, Jarzebska N, Mayoux E, Todorov VT, Hohenstein B, et al. 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. 2014;307(3):F317–25. doi: http://doi.org/10.1152/ajprenal.00145.2014. PubMed PMID: 24944269.
    » https://doi.org/10.1152/ajprenal.00145.2014
  • 81.
    Vallon V, Gerasimova M, Rose MA, Masuda T, Satriano J, Mayoux E, et al. SGLT2 inhibitor empagliflozin reduces kidney growth and albuminuria in proportion to hyperglycemia and prevents glomerular hyperfiltration in diabetic Akita mice. Am J Physiol Renal Physiol. 2014;306(2):F194–204. doi: http://doi.org/10.1152/ajprenal.00520.2013. PubMed PMID: 24226524.
    » https://doi.org/10.1152/ajprenal.00520.2013
  • 82.
    Tahara A, Kurosaki E, Yokono M, Yamajuku D, Kihara R, Hayashizaki Y, et al. Effects of sodium-glucose cotransporter 2 selective inhibitor ipragliflozin on hyperglycaemia, oxidative stress, inflammation and liver injury in streptozotocin-induced type 1 diabetic rats. J Pharm Pharmacol. 2014;66(7):975–87. doi: http://doi.org/10.1111/jphp.12223. PubMed PMID: 24533859.
    » https://doi.org/10.1111/jphp.12223
  • 83.
    Xu L, Nagata N, Nagashimada M, Zhuge F, Ni Y, Chen G, et al. SGLT2 inhibition by empagliflozin promotes fat utilization and browning and attenuates inflammation and insulin resistance by polarizing m2 macrophages in diet-induced obese mice. EBioMedicine. 2017;20:137–49. doi: http://doi.org/10.1016/j.ebiom.2017.05.028. PubMed PMID: 28579299.
    » https://doi.org/10.1016/j.ebiom.2017.05.028
  • 84.
    Chen L, Klein T, Leung PS. Effects of combining linagliptin treatment with BI-38335, a novel SGLT2 inhibitor, on pancreatic islet function and inflammation in db/db mice. Curr Mol Med. 2012;12(8):995–1004. doi: http://doi.org/10.2174/156652412802480970. PubMed PMID: 22804249.
    » https://doi.org/10.2174/156652412802480970
  • 85.
    Tang L, Wu Y, Tian M, Sjostrom CD, Johansson U, Peng XR, et al. Dapagliflozin slows the progression of the kidney and liver fibrosis associated with type 2 diabetes. Am J Physiol Endocrinol Metab. 2017;313(5):E563–76. doi: http://doi.org/10.1152/ajpendo.00086.2017. PubMed PMID: 28811292.
    » https://doi.org/10.1152/ajpendo.00086.2017
  • 86.
    Birnbaum Y, Bajaj M, Yang HC, Ye Y. Combined SGLT2 and DPP4 inhibition reduces the activation of the Nlrp3/ASC inflammasome and attenuates the development of diabetic nephropathy in mice with type 2 diabetes. Cardiovasc Drugs Ther. 2018;32(2):135–45. doi: http://doi.org/10.1007/s10557-018-6778-x. PubMed PMID: 29508169.
    » https://doi.org/10.1007/s10557-018-6778-x
  • 87.
    Benetti E, Mastrocola R, Vitarelli G, Cutrin JC, Nigro D, Chiazza F, et al. Empagliflozin protects against diet-induced NLRP-3 inflammasome activation and lipid accumulation. J Pharmacol Exp Ther. 2016;359(1):45–53. doi: http://doi.org/10.1124/jpet.116.235069. PubMed PMID: 27440421.
    » https://doi.org/10.1124/jpet.116.235069
  • 88.
    Ye Y, Bajaj M, Yang HC, Perez-Polo JR, Birnbaum Y. SGLT-2 inhibition with dapagliflozin reduces the activation of the Nlrp3/ASC inflammasome and attenuates the development of diabetic cardiomyopathy in mice with Type 2 Diabetes. Further Augmentation of the Effects with Saxagliptin, a DPP4 Inhibitor. Cardiovasc Drugs Ther. 2017;31(2):119–32. doi: http://doi.org/10.1007/s10557-017-6725-2. PubMed PMID: 28447181.
    » https://doi.org/10.1007/s10557-017-6725-2
  • 89.
    Tahara A, Kurosaki E, Yokono M, Yamajuku D, Kihara R, Hayashizaki Y, et al. Effects of SGLT2 selective inhibitor ipragliflozin on hyperglycemia, hyperlipidemia, hepatic steatosis, oxidative stress, inflammation, and obesity in type 2 diabetic mice. Eur J Pharmacol. 2013;715(1-3):246–55. doi: http://doi.org/10.1016/j.ejphar.2013.05.014. PubMed PMID: 23707905.
    » https://doi.org/10.1016/j.ejphar.2013.05.014

Publication Dates

  • Publication in this collection
    09 Sept 2024
  • Date of issue
    Oct-Dec 2024

History

  • Received
    11 Dec 2023
  • Accepted
    28 May 2024
Sociedade Brasileira de Nefrologia Rua Machado Bittencourt, 205 - 5ºandar - conj. 53 - Vila Clementino - CEP:04044-000 - São Paulo SP, Telefones: (11) 5579-1242/5579-6937, Fax (11) 5573-6000 - São Paulo - SP - Brazil
E-mail: bjnephrology@gmail.com