ABSTRACT

Anti-aging therapy is the latest frontier in the world of medical science, especially for widespread diseases such as chronic kidney disease (CKD). Both renal aging and CKD are characterized by increased cellular senescence, inflammation and oxidative stress. A variety of cellular signalling mechanisms are involved in these processes, which provide new potential targets for therapeutic strategies aimed at counteracting the onset and progression of CKD. At the same time, sodium–glucose co-transporter 2 inhibitors (SGLT2is) continuously demonstrate large beneficial effects at all stages of the cardiorenal metabolic continuum. The broad-spectrum benefits of SGLT2is have led to changes in several treatment guidelines and to growing scientific interest in the underlying working principles. Multiple mechanisms have been studied to explain these great renal benefits, but many things remain to be solved. With this in mind, we provide an overview of the experimental evidence for the effects of SGLT2is on the molecular pathway’s ability to modulate senescence, aging and parenchymal damage, especially at the kidney level. We propose to shed some light on the role of SGLT2is in kidney care by focusing on their potential to reduce the progression of kidney disease across the spectrum of aging and dysregulation of senescence.

INTRODUCTION

Aging and chronic diseases have been regarded as critical issues due to the increase in the elderly population and its impact on global health. Physiologic kidney aging and several kidney diseases share common biologic processes and molecular pathways [1], including cellular senescence, inflammation, fibrosis, vascular rarefaction, loss of glomeruli and tubular dysfunction, which lead to glomerulosclerosis and tubulointerstitial fibrosis [2, 3]. Nevertheless, the processes driving the progressive decline of kidney function performance during aging and the interrelation between aging and disease are yet not fully understood. In fact, correctly defining the cellular and molecular processes of aging is truly a challenge [4]. Aging-associated response mechanisms such as metabolic adaptation, senescence or apoptosis are mainly programmed. Therefore, aging can be defined as failure to counteract damage, and this failure causes functional decline, pathology and death. Factors causative in the initiation of kidney aging might be stochastic, consisting of acute kidney injury (AKI) or chronic diseases such as glomerulonephritis and diabetic kidney disease. The timely accumulation of different entities of cellular insults causes increased storage of senescent cells driving detrimental effects, thereby leading to accelerated nephron loss [5].

Certainly the balance between tissue dysfunction and repair progressively diminishes during aging. In response to a variety of stressors, cells can either undergo apoptosis or enter a secretory phenotype featuring changes in morphology and transcriptional profile and resistance to apoptosis, namely senescence. Cellular senescence was initially considered with regard to its physiological and homeostatic effects, particularly during embryonic development and wound healing, but it is now seen as a pathological process that contributes to aging as well as to various diseases and metabolic disorders. The characteristic triad of senescence includes arrested cell growth, resistance to apoptosis and senescence-associated secretory phenotype (SASP), which typically lead to macromolecular damage and altered metabolism [6]. In fact, senescent cells are characterized by morphological alterations including large, flat bodies and organelle abnormalities and chromatin organization, as well as loss of physiological functions [7]. In particular, cellular senescence is characterized by altered transcriptome and secretome, sharing characteristic features with kidney aging and leading to chronic kidney disease (CKD) progression (Fig. 1). Interestingly, although aging at the organismal level might be irreversible, the kinetics can be decelerated.

Structural and functional changes in the kidney across the senescence–aging spectrum. AKI, glomerulonephritis and diabetic kidney disease lead to microscopic and macroscopic changes cutting across the senescence–aging spectrum. These changes imply kidney damage that is expressed in proteinuria and kidney function decline.
Figure 1:

Structural and functional changes in the kidney across the senescence–aging spectrum. AKI, glomerulonephritis and diabetic kidney disease lead to microscopic and macroscopic changes cutting across the senescence–aging spectrum. These changes imply kidney damage that is expressed in proteinuria and kidney function decline.

In in vitro or in vivo models, sodium–glucose co-transporter 2 inhibitors (SGLT2is) have been shown to correct or improve many of the pathological processes involved in the development and progression of kidney aging, including inflammation, endothelial dysfunction, mitochondrial injury, fibrosis and cellular senescence [8]. These protective effects are likely to underlie the ability of these agents to slow progression of established CKD as demonstrated in large prospective clinical trials [9]. We believe detailed analysis of their potentiality might stimulate further experimental studies to outline new therapeutic targets in the context of kidney aging prevention and treatment. For these reasons, in this review we attempt to describe the aging-related changes in the kidney cells and the prospects of SGLT2is in the therapeutic management of kidney aging, focusing on its associated molecular pathways.

KIDNEY AGING AND SENESCENCE

The kidney in healthy aging

In clinical studies, aging-associated changes are related to a decrease in renal function [both glomerular filtration rate (GFR) and renal plasma flow (RPF)] [10], with an average yearly decline of GFR of 0.7–0.9 ml/min/1.73 m2 in otherwise healthy individuals [11]. The fundamental origins of the decrease in GFR with healthy aging are not fully understood, but it is a phenomenon related to a slowly progressive loss of nephrons from age 30 years onward. From a histological point of view, kidney aging is mainly characterized by nephrosclerosis, i.e. global glomerulosclerosis, interstitial fibrosis/tubule atrophy (IF/TA) and arteriolosclerosis [12], and the rate of GFR decrease is the result of several mechanisms leading to reduced glomerular density [13]. As a matter of fact, nephron hyperfiltration occurring in residual nephron and haemodynamic stress on podocytes contributes to their degeneration, altering hydraulic permeability and the surface area available for filtration [14].

While healthy aging is associated with structural changes in the kidney and a decrease in GFR, this does not always involve a substantive increase in the risk of end-stage kidney disease (ESKD) or mortality. For this reason, some authors have proposed an age-based modification to the definition of CKD. According to this vision, the definition of CKD when estimated GFR (eGFR) is <75 ml/min/1.73 m2 in younger adults (age <40 years) [15] and when eGFR is <45 ml/min/1.73 m2 in older adults (age >65 years) [16] appears to be properly balanced.

However, due to the pivotal role of the kidney in the development of hypertension and regulation of body volume and metabolic homeostasis, GFR decline and the age-related changes in the structure and function of kidney cells actually increase the overall risk class. Animal and human studies report that age is related to changes in the permeability of the capillary wall in glomeruli, increased susceptibility to podocyte injury, changes in tubular reabsorption and secretory capacities, changes in urinary concentration and production of kidney-derived hormones and bioactive molecules [17], increased apoptosis, oxidative stress and inflammation [18, 19]. The subsequent deterioration of the integrity of the slit pore membrane in glomeruli defines both GFR decline and albumin permeability alteration. In other words, healthy kidney aging could be viewed as a compound of physiological changes with pathological implications. For these reasons, specific management strategies that are of actual benefit to older patients with decreased GFR warrant attention.

Aging in kidney disease

In the context of contemporary nephrology research, the effect of age on the fate of CKD patients is a critical issue. The kidneys of elderly patients are more vulnerable to the detrimental effects of proteinuria due to the greater degree of renal fibrosis and ischaemia, significantly increasing the risk of ESKD in older CKD patients [20].

Research attention has been recently drawn to the knowledge gaps of age-related differences in the mechanisms and pathways that contribute to progression to ESKD. In contrast with healthy aging, the global glomerulosclerosis related to GFR decline in kidney diseases [21] is a result of visceral glomerular podocyte degeneration, inadequate repair and autophagy impairment. Autophagy, a conserved lysosomal pathway for the degradation of cytoplasmic components, is essential to the maintenance of kidney homeostasis, and its reduction has been shown to be detrimental to kidney structure and function. Autophagy defects in kidney cells of both tubular and glomerular compartments have been shown to contribute to the development of diabetic kidney disease, focal segmental glomerulosclerosis and polycystic kidney disease [22–24].

The mechanisms underlying the aging-related reduction of autophagy in podocytes have largely been investigated in mice. Mitochondrial damage, endothelial reticulum stress and accumulation of oxidized and ubiquitylated protein aggregates and lipofuscin were found in a mouse model of autophagy-deficient podocytes. Proteasome activity has been shown to be increased in aged mice, probably for the purpose of clearing protein aggregates and compensating for the loss of autophagy [25]. Another mouse model of podocyte-specific deletion of a lysosomal protease further demonstrated the importance of lysosomal activity in podocyte maintenance during aging. Defective autolysosome degradation in podocytes triggered the accumulation of toxic lipofuscins and protein aggregates, leading to apoptotic podocyte death. As a result, lysosomal-defective mice developed proteinuria and kidney failure [23].

One more longevity factor is sirtuins (SIRT), a family of NAD+-dependent class III histone deacetylases, which are involved in metabolic regulation and are activated by increased NAD+ levels. SIRT1 cross-talks with other pathways regulating different mechanisms involved in energy metabolism and cell survival, including autophagy [26]. Interestingly, it was reported that podocyte-specific knockdown of SIRT1 aggravated age-associated kidney injury in aging mice [27]. Sirtuins role in anti-aging interventions has been confirmed in several experimental studies testing the role of caloric restriction (CR) on aging. CR is well known to enhance lifespans and health spans, preventing many age-related diseases. The biochemical mechanisms by which it increases kidney health is linked, at least in part, to enhanced mitochondrial Ca2+ uptake rates, regulating several aspects of mitochondrial function, including oxidative phosphorylation and redox balance [28, 29].

Accelerated cellular senescence in the kidney

Cellular senescence not only contributes to aging, but also plays a causal role in numerous age-related diseases; in particular, it has been proven to be involved in the pathogenesis of AKI, AKI to CKD transition and many types of CKD [30]. Cell senescence may develop during an acute response after injury as a mechanism of tissue repair [30]. This kind of ‘acute senescence’ is a tightly controlled process that participates in repair mechanisms and limits fibrosis. In contrast, in chronic diseases, senescent cells accumulate in the kidney in response to a variety of stressors, including metabolic stress, telomere shortening [31], oncogenic mutations, inflammation and mitochondrial dysfunction [32]. These stressors promote cell-cycle arrest via pathways either dependent or independent of the DNA damage response. Differently from the ‘acute senescence’ setting, in chronic diseases, senescent cells are scarcely removed by apoptosis or immune clearance and are increasingly considered to be mediators of disease progression [33, 34]. Data from animal models suggest that senescent interstitial and tubular epithelial cells contribute to ischaemia–reperfusion injury and AKI, as well as to AKI to CKD progression. Endothelial cell, podocyte and mesangial cell senescence might contribute to diabetic kidney disease [35]. Tubular epithelial cell senescence has also been detected in many forms of CKD, including obesity-related nephropathy, membranous nephropathy, lupus nephritis, minimal change disease, unilateral ureteral obstruction and immunoglobulin A nephropathy. In contrast, cellular senescence occurs as part of sequential and tightly orchestrated stress-induced effector programs involving metabolic changes associated with obesity and diabetes. High glucose drives in vitro senescence in kidney podocyte, mesangial and tubular cells [36]. Furthermore, hyperglycaemia causes cellular senescence via an SGLT2-dependent pathway in proximal tubules in the early stage of diabetic nephropathy [37].

Regardless of the kidney disease, the major changes in GFR with aging can be attributed primarily to nephron loss. The last evidence shows that the drawbacks of accelerated cell senescence in kidney diseases are 2-fold. First, senescence causes a cell cycle arrest, with a consequent loss of tissue repair capacity; this is especially relevant for cells with low replication rates, like podocytes. Second, senescent cells produce pro-inflammatory and matrix-degrading molecules in what is known as the SASP, leading to development and progression of glomerulosclerosis and IF/TA [38] (Fig. 2). Accordingly, the commonly used biomarkers of in vitro senescent cells are enzymatic activity of the lysosomal hydrolase senescence-associated β-galactosidase (SA-β-GAL), reflecting an increase in lysosomal mass [39, 40], the development of an SASP [41] and arrested cell growth [5].

Overview of various mechanisms involved in cellular senescence. The cell cycle becomes dysfunctional as telomere shortening and mitochondrial dysfunction cause DNA damage, the extracellular matrix is expanded and there is organelle injury and senescent cells develop a SASP that leads to chronic inflammation. All the kidney cells are involved—podocytes, tubules and endothelial cells.
Figure 2:

Overview of various mechanisms involved in cellular senescence. The cell cycle becomes dysfunctional as telomere shortening and mitochondrial dysfunction cause DNA damage, the extracellular matrix is expanded and there is organelle injury and senescent cells develop a SASP that leads to chronic inflammation. All the kidney cells are involved—podocytes, tubules and endothelial cells.

Many of the cell senescence inducers, namely metabolic stress, telomere shortening [31], inflammation and mitochondrial dysfunction [32], cause overexpression of the cyclin-dependent kinase inhibitors p16INKA and/or p21cip1. Their binding to CDK4/6 inhibits cyclin D-CDK4/6 complex and induces cell cycle arrest at the G1/S cell cycle checkpoint. Activation of p16INKA/Rb and p53/p21 tumour suppressor networks implements this growth arrest in the nucleus [38], composing a key signalling cascade implicated in kidney cell senescence. Experimental models of CKD showed that accumulation of senescent cells later the G2/M phase triggers a secretory phenotype, resulting in fibrosis [42]. Interestingly, SIRT1 has been shown to inhibit cellular senescence through regulation of the p53/p21 pathway. Podocyte-specific knockdown of SIRT1 accelerated age-related glomerulosclerosis and podocyte loss in mice kidneys [27].

In contrast, a number of signalling molecules have been identified to exacerbate renal cell senescence and renal aging. For instance, Wnt9a/β-catenin signalling seems to promote renal tubular senescence and renal fibrosis in diseased kidneys, as evidenced by the upregulated expression of p16INK4a, p53 and p21, and increased SA‑β‑GAL activity in renal tubules [43].

Reactive oxygen species (ROS) can induce cellular senescence by regulating the p16INK4a/prb and p53/p21 pathways. In turn, senescent cells produce ROS [44], perpetuating a vicious cycle of cellular damage. Accumulation of ROS results in lipid peroxidation, which leads to elevated advanced glycation end-products and advanced lipoxidation end-products, both of which regulate cellular senescence [45]. One of the shared links between oxidative stress and cellular senescence might be inflammation, which drives and results from both processes and contributes to CKD.

ROS signalling also mediates autophagy-delaying cell senescence and leads to SASP, a distinctive secretome consisting of a various pro-inflammatory molecules, metalloproteases and growth factors [39]. SASP components include interleukin (IL)-1β, IL-6 and IL-8 and transforming growth factor (TGF)-β1, and these can act in a paracrine and autocrine fashion, leading to persistence and propagation of senescence within tissues [46]. Direct evidence in the kidney is lacking and specific studies are needed. Detection of cells displaying the SASP in the kidney would be a major step in determining the choice of treatment.

EFFECT OF SGLT2 INHIBITION IN KIDNEY AGING AND SENESCENCE

Geroscience represents a novel paradigm whereby biological aging is recognized as the major modifiable driver of age-related diseases and other late-life conditions. Some authors have proposed a standardized process for evaluating US Food and Drug Administration (FDA)-approved medications for their geroscience potential, and SGLT2i demonstrated the most robust clinical evidence for a reduction of death from any cause [47].

SGLT2 inhibition has pleiotropic effects on multiple organ systems. As for the heart, there have been several demonstrations of the mechanisms underlying the cardioprotective effect conferred by inhibition of SGLT2 activity, including a reduction of myocardial fibrosis and remodelling, ischaemia–reperfusion injury, oxidative stress and inflammation and improved myocardial contractility [48, 49], which lead to a reduction in heart failure and cardiovascular events [50, 51]. It has also been shown that mitochondrial function is improved and amyloid plaque deposition in the brain is reduced, ameliorating age-related and neurodegeneration disease [52, 53].

Studies on the effect of defective senescent cell formation have provided a major step forward in our understanding of the effects of treating cell senescence. Baker et al. [54] identified a causal link between cellular senescence and aging phenotypes by showing that the clearance of senescent cells in a mouse progeroid model in vivo can delay age-related tissue dysfunction. More recently, it was demonstrated that the removal of senescent cells delays aging, increases lifespan and restores organ function, including the blood vessels [55] and kidney [56], without increasing the risk of cancer. As changes at the molecular level due to SGLT2i involve inflammation, oxidative stress and DNA damage (Fig. 3), we aim to gain a clearer picture of the current scientific understanding of the potential of SGLT2is related to targeting aging and senescence in the kidney (Table 1).

Molecular effects of SGLT2is on kidney aging. SGLT2is act on different metabolic pathways in order to protect kidney function and prevent damage: SIRT1 is able to deacetylate and activate NRF2 that regulates mitochondrial fitness; inhibition of mTOR mediates renoprotection; autophagy is affected by an elevated HIF-2α:HIF-1α ratio; and upregulation of AMPK, adiponectin and PPAR-α results in inhibition of chronic inflammation through energy deprivation. Furthermore, SGLT2is decrease oxidative stress in different ways: they inhibit NADPH-oxidase 4 and reduce the influx of glucose into RPTECs in order to increase ketone activation of the NRF2 pathway and mTOR inhibition; the decreased mitochondrial calcium overload contributes to upregulation of SIRT1 and SOD1 and 2. PPAR-α: peroxisome proliferator-activated receptor α.
Figure 3:

Molecular effects of SGLT2is on kidney aging. SGLT2is act on different metabolic pathways in order to protect kidney function and prevent damage: SIRT1 is able to deacetylate and activate NRF2 that regulates mitochondrial fitness; inhibition of mTOR mediates renoprotection; autophagy is affected by an elevated HIF-2α:HIF-1α ratio; and upregulation of AMPK, adiponectin and PPAR-α results in inhibition of chronic inflammation through energy deprivation. Furthermore, SGLT2is decrease oxidative stress in different ways: they inhibit NADPH-oxidase 4 and reduce the influx of glucose into RPTECs in order to increase ketone activation of the NRF2 pathway and mTOR inhibition; the decreased mitochondrial calcium overload contributes to upregulation of SIRT1 and SOD1 and 2. PPAR-α: peroxisome proliferator-activated receptor α.

Table 1:

Experimental models proving mechanisms involved in kidney protection from aging and/or senescence with SGLT2i treatment

Organs/cells/tissuesExperimental modelsSGLT2i testedMechanisms involvedReferences
Renal proximal tubular cells (mice)Mice with or without diabetes in which HHIP was present or specifically knocked out in renal tubulesCanagliflozinInhibited β-galactosidase activity[70]
Proximal tubules (mice)Damaged proximal tubules of ApoE knockout mice fed a high-fat diet (model of non-proteinuric diabetic kidney disease)EmpagliflozinElevation of ketone body that corrects mTORC1 hyperactivation[53]
Kidney cortex (rats)Male hereditary hypertriglyceridaemic rats fed a standard diet with or without SGLT2iEmpagliflozinIncreased activity of GSH-dependent enzyme GSH-Px and SOD decreased levels of lipoperoxidation products[64]
Kidney tissue sections (mice)Mice aged by subcutaneous injection of D-galactoseEmpagliflozinUpregulated SIRT1 levels[65]
Endothelial cells (ECs) (pigs and male rats)ECs were isolated from porcine coronary arteries and arterial segments from rats, incubated in the absence or presence of either NADPH oxidase inhibitor, an AT1R antagonist, SGLT1 and/or SGLT2i, then Ang II or a nitric oxide synthase inhibitorEmpagliflozinAbolished pro-oxidant responses, SA-β-GAL activity, expression of senescence markers p53 and p21 and p16 protein[63]
Aorta tree and aortic valve (mice)Spontaneously atherosclerotic miceEmpagliflozinImproved protein expression level of p-AMPK affected by ox-LDL in cell; reduced gene expression level of inflammatory factors and protein expression level of NF-κB[69]
Human renal proximal tubular epithelial cells (hRPTECs)hRPTECs were incubated under high-glucose conditionsDapagliflozinReduced p16, inhibited ROS production with blunted DNA damage, ataxia telangiectasia mutated kinase (ATM) and p53 phosphorylation; decreased high-glucose-induced IL-1 and IL-8 production; normalized TGF-1 level[60]
hRPTECshRPTECs were incubated under high-glucose conditionsTofogliflozinReduced MCP-1 gene expression and apoptotic cell death[61]
Human renal tubular epithelial cell line (HK-2 cells)HK-2 cells pretreated with hydrogen peroxide to induce cellular senescenceDapagliflozinInhibited cellular senescence and oxidative stress via ketone-induced NRF2 activation[66]
Human endothelial cells (hECs)hECs were plated on glass-bottom dishes and treated with different concentrations of glucoseEmpagliflozinReduced mitochondrial Ca2+ overload and ROS production attenuated cellular permeability and improved cell viability in response to oxidative stress[62]
Human umbilical vein endothelial cells (HUVEC)HUVEC were incubated under high-glucose conditionsDapagliflozinAbrogated high-glucose-induced endothelial cell dysfunction by AMPK/SIRT1 pathway up-regulation[54]
Organs/cells/tissuesExperimental modelsSGLT2i testedMechanisms involvedReferences
Renal proximal tubular cells (mice)Mice with or without diabetes in which HHIP was present or specifically knocked out in renal tubulesCanagliflozinInhibited β-galactosidase activity[70]
Proximal tubules (mice)Damaged proximal tubules of ApoE knockout mice fed a high-fat diet (model of non-proteinuric diabetic kidney disease)EmpagliflozinElevation of ketone body that corrects mTORC1 hyperactivation[53]
Kidney cortex (rats)Male hereditary hypertriglyceridaemic rats fed a standard diet with or without SGLT2iEmpagliflozinIncreased activity of GSH-dependent enzyme GSH-Px and SOD decreased levels of lipoperoxidation products[64]
Kidney tissue sections (mice)Mice aged by subcutaneous injection of D-galactoseEmpagliflozinUpregulated SIRT1 levels[65]
Endothelial cells (ECs) (pigs and male rats)ECs were isolated from porcine coronary arteries and arterial segments from rats, incubated in the absence or presence of either NADPH oxidase inhibitor, an AT1R antagonist, SGLT1 and/or SGLT2i, then Ang II or a nitric oxide synthase inhibitorEmpagliflozinAbolished pro-oxidant responses, SA-β-GAL activity, expression of senescence markers p53 and p21 and p16 protein[63]
Aorta tree and aortic valve (mice)Spontaneously atherosclerotic miceEmpagliflozinImproved protein expression level of p-AMPK affected by ox-LDL in cell; reduced gene expression level of inflammatory factors and protein expression level of NF-κB[69]
Human renal proximal tubular epithelial cells (hRPTECs)hRPTECs were incubated under high-glucose conditionsDapagliflozinReduced p16, inhibited ROS production with blunted DNA damage, ataxia telangiectasia mutated kinase (ATM) and p53 phosphorylation; decreased high-glucose-induced IL-1 and IL-8 production; normalized TGF-1 level[60]
hRPTECshRPTECs were incubated under high-glucose conditionsTofogliflozinReduced MCP-1 gene expression and apoptotic cell death[61]
Human renal tubular epithelial cell line (HK-2 cells)HK-2 cells pretreated with hydrogen peroxide to induce cellular senescenceDapagliflozinInhibited cellular senescence and oxidative stress via ketone-induced NRF2 activation[66]
Human endothelial cells (hECs)hECs were plated on glass-bottom dishes and treated with different concentrations of glucoseEmpagliflozinReduced mitochondrial Ca2+ overload and ROS production attenuated cellular permeability and improved cell viability in response to oxidative stress[62]
Human umbilical vein endothelial cells (HUVEC)HUVEC were incubated under high-glucose conditionsDapagliflozinAbrogated high-glucose-induced endothelial cell dysfunction by AMPK/SIRT1 pathway up-regulation[54]
Table 1:

Experimental models proving mechanisms involved in kidney protection from aging and/or senescence with SGLT2i treatment

Organs/cells/tissuesExperimental modelsSGLT2i testedMechanisms involvedReferences
Renal proximal tubular cells (mice)Mice with or without diabetes in which HHIP was present or specifically knocked out in renal tubulesCanagliflozinInhibited β-galactosidase activity[70]
Proximal tubules (mice)Damaged proximal tubules of ApoE knockout mice fed a high-fat diet (model of non-proteinuric diabetic kidney disease)EmpagliflozinElevation of ketone body that corrects mTORC1 hyperactivation[53]
Kidney cortex (rats)Male hereditary hypertriglyceridaemic rats fed a standard diet with or without SGLT2iEmpagliflozinIncreased activity of GSH-dependent enzyme GSH-Px and SOD decreased levels of lipoperoxidation products[64]
Kidney tissue sections (mice)Mice aged by subcutaneous injection of D-galactoseEmpagliflozinUpregulated SIRT1 levels[65]
Endothelial cells (ECs) (pigs and male rats)ECs were isolated from porcine coronary arteries and arterial segments from rats, incubated in the absence or presence of either NADPH oxidase inhibitor, an AT1R antagonist, SGLT1 and/or SGLT2i, then Ang II or a nitric oxide synthase inhibitorEmpagliflozinAbolished pro-oxidant responses, SA-β-GAL activity, expression of senescence markers p53 and p21 and p16 protein[63]
Aorta tree and aortic valve (mice)Spontaneously atherosclerotic miceEmpagliflozinImproved protein expression level of p-AMPK affected by ox-LDL in cell; reduced gene expression level of inflammatory factors and protein expression level of NF-κB[69]
Human renal proximal tubular epithelial cells (hRPTECs)hRPTECs were incubated under high-glucose conditionsDapagliflozinReduced p16, inhibited ROS production with blunted DNA damage, ataxia telangiectasia mutated kinase (ATM) and p53 phosphorylation; decreased high-glucose-induced IL-1 and IL-8 production; normalized TGF-1 level[60]
hRPTECshRPTECs were incubated under high-glucose conditionsTofogliflozinReduced MCP-1 gene expression and apoptotic cell death[61]
Human renal tubular epithelial cell line (HK-2 cells)HK-2 cells pretreated with hydrogen peroxide to induce cellular senescenceDapagliflozinInhibited cellular senescence and oxidative stress via ketone-induced NRF2 activation[66]
Human endothelial cells (hECs)hECs were plated on glass-bottom dishes and treated with different concentrations of glucoseEmpagliflozinReduced mitochondrial Ca2+ overload and ROS production attenuated cellular permeability and improved cell viability in response to oxidative stress[62]
Human umbilical vein endothelial cells (HUVEC)HUVEC were incubated under high-glucose conditionsDapagliflozinAbrogated high-glucose-induced endothelial cell dysfunction by AMPK/SIRT1 pathway up-regulation[54]
Organs/cells/tissuesExperimental modelsSGLT2i testedMechanisms involvedReferences
Renal proximal tubular cells (mice)Mice with or without diabetes in which HHIP was present or specifically knocked out in renal tubulesCanagliflozinInhibited β-galactosidase activity[70]
Proximal tubules (mice)Damaged proximal tubules of ApoE knockout mice fed a high-fat diet (model of non-proteinuric diabetic kidney disease)EmpagliflozinElevation of ketone body that corrects mTORC1 hyperactivation[53]
Kidney cortex (rats)Male hereditary hypertriglyceridaemic rats fed a standard diet with or without SGLT2iEmpagliflozinIncreased activity of GSH-dependent enzyme GSH-Px and SOD decreased levels of lipoperoxidation products[64]
Kidney tissue sections (mice)Mice aged by subcutaneous injection of D-galactoseEmpagliflozinUpregulated SIRT1 levels[65]
Endothelial cells (ECs) (pigs and male rats)ECs were isolated from porcine coronary arteries and arterial segments from rats, incubated in the absence or presence of either NADPH oxidase inhibitor, an AT1R antagonist, SGLT1 and/or SGLT2i, then Ang II or a nitric oxide synthase inhibitorEmpagliflozinAbolished pro-oxidant responses, SA-β-GAL activity, expression of senescence markers p53 and p21 and p16 protein[63]
Aorta tree and aortic valve (mice)Spontaneously atherosclerotic miceEmpagliflozinImproved protein expression level of p-AMPK affected by ox-LDL in cell; reduced gene expression level of inflammatory factors and protein expression level of NF-κB[69]
Human renal proximal tubular epithelial cells (hRPTECs)hRPTECs were incubated under high-glucose conditionsDapagliflozinReduced p16, inhibited ROS production with blunted DNA damage, ataxia telangiectasia mutated kinase (ATM) and p53 phosphorylation; decreased high-glucose-induced IL-1 and IL-8 production; normalized TGF-1 level[60]
hRPTECshRPTECs were incubated under high-glucose conditionsTofogliflozinReduced MCP-1 gene expression and apoptotic cell death[61]
Human renal tubular epithelial cell line (HK-2 cells)HK-2 cells pretreated with hydrogen peroxide to induce cellular senescenceDapagliflozinInhibited cellular senescence and oxidative stress via ketone-induced NRF2 activation[66]
Human endothelial cells (hECs)hECs were plated on glass-bottom dishes and treated with different concentrations of glucoseEmpagliflozinReduced mitochondrial Ca2+ overload and ROS production attenuated cellular permeability and improved cell viability in response to oxidative stress[62]
Human umbilical vein endothelial cells (HUVEC)HUVEC were incubated under high-glucose conditionsDapagliflozinAbrogated high-glucose-induced endothelial cell dysfunction by AMPK/SIRT1 pathway up-regulation[54]

SGLT2is effect on aging: the role of caloric restriction

Several experimental models have contributed to the conviction that SGLT2is may exhibit anti-aging effects by providing protection against ROS-induced cellular senescence, DNA damage and attenuated inflammation, reducing end-organ fibrosis and improving endothelial function [57]. Moreover, SGLT2is have been shown to modulate the nutrient-sensing pathways, known drivers of aging. By urinary caloric loss, SGLT2is activate a fasting-like metabolic state and reduce insulin signalling in response to a decrease in the glucose level. These events reduce the insulin:glucagon ratio and increase hepatic and kidney glucose production from glycogenolysis and gluconeogenesis [58]. The compensating formation of ketone bodies, in addition to be a direct energy source for damaged proximal tubular epithelial cells (PTECs), was shown to prevent PTEC and podocyte damage by inhibiting mammalian target of rapamycin (mTOR) signalling in mice models of proteinuric and non-proteinuric diabetic kidney disease [59]. By promoting a nutrient deprivation state, SGLT2is upregulate the energy deprivation sensors adenosine 5′-monophosphate-activated protein kinase (AMPK) and SIRT1, inhibit the nutrient sensors mTOR and insulin/insulin-like growth factor 1 and modulate the closely linked hypoxia-inducible factor (HIF)-2α/HIF-1α pathways [57]. Activation of the AMPK/SIRT1 axis has recently been suggested as a novel target for the treatment of high-glucose-induced endothelial cell injury and dapagliflozin abrogated high-glucose-induced endothelial cell dysfunction by AMPK/SIRT1 pathway up-regulation in human umbilical vein endothelial cells [60].

Several studies have been conducted on the effects of CR on longevity across various animal species, demonstrating an increase in lifespan and improved health outcomes [61]. Among involved molecular processes, DNA methylation has been shown to play a critical role: CR has resulted in attenuation of age-related methylation drift in mice and rhesus monkeys [62, 63]. The Comprehensive Assessment of Long-Term Effects of Reducing Intake of Energy (CALERIE) randomized controlled trial found that CR modified DunedinPACE, a DNA methylation biomarker of the pace of aging. This finding supports the geroscience hypothesis that proposed this strategy to slow or reverse molecular changes that occur with aging, delaying or preventing multiple chronic diseases and extending healthy lifespan [64]. SGLT2is belong to the category of CR mimetics, specifically the downstream type, regulating intracell signalling proteins [65]. Therefore, they represent a fascinating and promising class of drugs that modify cellular mechanisms to mimic the effects of caloric restriction. Their complexity and potential to modulate multiple pathways make them an exciting area of research in the quest to promote healthy aging [66].

SGLT2i: redox control of senescence (oxidative stress and mitochondrial dynamics)

Excess ROS can directly or indirectly damage DNA, proteins and lipids, thereby inducing transgenation, conformational changes in proteins and lipid peroxidation [67, 68]. There is increasing evidence that oxidative stress causes oxidative tissue damage and disrupts cellular metabolism [69, 70], especially in the kidney, which has fast metabolic processes. The increased influx of glucose into the renal proximal tubular epithelial cells (RPTECs) under high-glucose conditions leads to a burst of ROS production, both by the mitochondria and by nicotinamide adenine dinucleotide phosphate (NADPH) oxidase or xanthine oxidase [71]. By decreasing the influx of glucose into RPTECs, dapagliflozin and tofagliflozin significantly ameliorate the high-glucose-induced oxidative stress and the consequent DNA damage response, especially in high-glucose conditions [72, 73].

Oxidative stress is known to be a common pathogenic substrate in chronic disease and may occur in the diabetic kidney at an early time. Specifically, dysregulated mitochondrial fitness has been proposed as a potential root of age-related frailty. Empagliflozin has been shown to significantly reduce mitochondrial Ca2+ overload and ROS production triggered by high glucose in human endothelial cells, attenuate cellular permeability and improve cell viability in response to oxidative stress [74]. These findings were associated with decreased frailty in diabetic and hypertensive patients after 3 months of treatment with empagliflozin as compared with the control group.

This protective role of SGLT2is on endothelial damage has been confirmed by the demonstration that pro-oxidant responses, SA-β-GAL activity, the expression of senescence markers p53 and p21 at both the messenger RNA and protein levels and p16 protein were upregulated by angiotensin II (Ang-II) and abolished by SGLT2i in endothelial cells isolated from porcine coronary arteries [75]. Ang-II up-regulates SGLT1 and 2 protein expression in endothelial cells and arterial segments, promoting sustained oxidative stress, senescence and dysfunction. Such a sequence contributes to coronary artery disease microparticle–induced endothelial dysfunction. Since AT1R/NADPH oxidase/SGLT1 and 2 pathways promote endothelial dysfunction, inhibition of SGLT1 and/or 2 appears as an attractive strategy to promote endothelial health.

Trnovska et al. [76] tested the effect of empagliflozin on metabolic parameters and insulin resistance using non-obese hereditary hypertriglyceridaemic rats, a strain characterized by elevated concentrations of triglycerides and insulin resistance. They observed that empagliflozin reduces oxidative stress in the kidney tissue. In fact, in the kidney cortex, activity of the γ-glutamyl-cysteinyl-glycine (GSH)-dependent enzyme GSH-Px and superoxide dismutase (SOD), an antioxidant enzyme, was increased in the empagliflozin-treated group while levels of lipoperoxidation products were decreased. Empagliflozin can also exert antioxidant effects by upregulating SIRT1 levels through upregulation of SOD1 and 2 in a mice model of senescence [77].

Moreover, dapagliflozin prevented the progression of diabetic kidney disease by inhibiting cellular senescence and oxidative stress via ketone-induced nuclear factor erythroid 2–related factor 2 (NRF2) activation. In particular, expression levels of ageing marker genes (p21, p16 and p53) and the expression levels of SASP in the kidney were reduced in a type 2 model of db/db mice, primarily focusing on cellular senescence and oxidative stress, when treated with dapagliflozin [78]. The fact that such improvement was not observed in the subgroup of mice treated with sulfonylurea suggests that dapagliflozin has antisenescent and antioxidant properties, which is independent of its glucose-lowering effect.

SGLT2is effect on kidney cellular senescence: inflammation pathways

Since SGLT2is promote ketogenesis from a shift toward fatty acid oxidation, one may postulate that they have anti-inflammatory effects similar to CR, with promising effects on longevity and the advantage of not being difficult to adhere to.

Results suggest that SGLT2is possess a tangible activity against low-grade inflammation, an effect possibly mediated by their ability to lower uric acid and insulin concentrations [79]. Moreover, reducing the glucose influx in senescent cells is likely to result in attenuation of their inflammatory affects. In fact, in vitro experiments detected that dapagliflozin decreased high-glucose-induced IL-1 and IL-8 production and normalized the level of TGF-1 in renal tubular epithelial cell culture supernatants [60]. Moreover, both monocyte chemoattractant protein-1 (MCP-1) gene expression and apoptotic cell death were reduced by SGLT2is [80].

The interaction between SGLT2is and the AMPK signal pathway was studied in both spontaneously atherosclerotic mice in vivo and an oxidized low-density lipoprotein (ox-LDL)-induced macrophage inflammation model in vitro. Empagliflozin can improve the protein expression level of phosphorylated AMPK (p-AMPK) affected by ox-LDL in cells and reduce the gene expression level of inflammatory factors and protein expression level of nuclear factor (NF)-κB, thus removing the effect of diabetes in terms of inflammation-mediated damage [81].

Hedgehog interacting protein (HHIP) encodes a protein of 700 amino acids attached to the cell membrane, regulates cellular function and is essential in organ development. HHIP is detectable in renal endothelial and epithelial cells in the normal, mature kidney and is believed to promote SASP through the release of a variety of inflammatory cytokines in remodelled kidney cells, thereby exacerbating the progression of diabetic kidney disease. In human renal proximal tubule HK2 cells, high-glucose-induced HHIP overexpression promoted increased SGLT2 protein expression and cellular senescence [82]. Moreover, their increased β-GAL activity was inhibited by canagliflozin, reaffirming a potential role of SGLT2is in the modulation of inflammaging.

CONCLUDING REMARKS AND FUTURE PROSPECTS

Targeting aging and senescence pathways offers myriad beneficial effects, particularly in the context of chronic disease. We depict aging and senescence as being interacting or intercalating, but different processes are becoming increasingly recognized as possible causes of disease pathophysiology throughout the cardiorenal system.

SGLT2is are antihyperglycaemic drugs that have been shown to protect the kidneys and heart of patients with or without type 2 diabetes and to preserve kidney function or reduce kidney failure. Recent research has provided further insights into the aging treatment paradigm. Many in vivo and in vitro studies are aimed at revealing the pathways underlying the pleiotropic effects of SGLT2is, both blood glucose-dependent and -independent mechanisms [83].

Understanding how aging and senescence of kidney cell types contributes to specific disease phenotypes and how the pathways involved can be regulated by SGLT2is will allow researchers to develop improved, targeted therapies to delay or prevent aging-associated diseases in the kidney.

While other strategies related to CR or the use of molecules such as metformin, rapamycin or resveratrol that interfere with SASP signalling factors are promising in order to delay the onset of age-associated diseases [84, 85], further basic and clinical research is needed to define the potential effects and complex mechanisms of SGLT2is in aging and senescence processes.

FUNDING

This research received no external funding.

AUTHORS’ CONTRIBUTIONS

All authors contributed to the study conception and design, data collection, and draft manuscript preparation. All authors reviewed the results and approved the final version of the manuscript.

DATA AVAILABILITY STATEMENT

No new data were generated or analysed in support of this research. All data supporting the findings of this study are available within the paper.

CONFLICT OF INTEREST STATEMENT

All authors and co-authors declare no conflicts of interest.

REFERENCES

1.

Schmitt
 
R
,
Melk
 
A
.

Molecular mechanisms of renal aging
.
Kidney Int
 
2017
;
92
:
569
79
.
2.

Denic
 
A
,
Glassock
 
RJ
,
Rule
 
AD
.

The kidney in normal aging: a comparison with chronic kidney disease
.
Clin J Am Soc Nephrol
 
2022
;
17
:
137
9
.
3.

Esposito
 
P
,
Verzola
 
D
,
Picciotto
 
D
 et al.   

Myostatin/activin-A signaling in the vessel wall and vascular calcification
.
Cells
 
2021
;
10
:
2070
.
4.

Schmeer
 
C
,
Kretz
 
A
,
Wengerodt
 
D
 et al.   

Dissecting aging and senescence—current concepts and open lessons
.
Cells
 
2019
;
8
:
1446
.
5.

Gorgoulis
 
V
,
Adams
 
PD
,
Alimonti
 
A
 et al.   

Cellular senescence: defining a path forward
.
Cell
 
2019
;
179
:
813
27
.
6.

Kang
 
C
.

Senolytics and senostatics: a two-pronged approach to target cellular senescence for delaying aging and age-related diseases
.
Mol Cells
 
2019
;
42
:
821
7
.
7.

Docherty
 
MH
,
O'Sullivan
 
ED
,
Bonventre
 
JV
 et al.   

Cellular senescence in the kidney
.
J Am Soc Nephrol
 
2019
;
30
:
726
36
.
8.

DeFronzo
 
RA
,
Reeves
 
WB
,
Awad
 
AS
.

Pathophysiology of diabetic kidney disease: impact of SGLT2 inhibitors
.
Nat Rev Nephrol
 
2021
;
17
:
319
34
.
9.

Patel
 
T
.

SGLT2 inhibitors reduce adverse renal and CV outcomes in patients with or without diabetes
.
Ann Intern Med
 
2023
;
176
:
JC27
.
10.

Vlassara
 
H
,
Torreggiani
 
M
,
Post
 
JB
 et al.   

Role of oxidants/inflammation in declining renal function in chronic kidney disease and normal aging
.
Kidney Int Suppl
 
2009
;
76
:
S3
11
.
11.

Glassock
 
RJ
,
Rule
 
AD
.

Aging and the kidneys: anatomy, physiology and consequences for defining chronic kidney disease
.
Nephron
 
2016
;
134
:
25
9
.
12.

Kremers
 
WK
,
Denic
 
A
,
Lieske
 
JC
 et al.   

Distinguishing age-related from disease-related glomerulosclerosis on kidney biopsy: the aging kidney anatomy study
.
Nephrol Dial Transplant
 
2015
;
30
:
2034
9
.
13.

Rule
 
AD
,
Amer
 
H
,
Cornell
 
LD
 et al.   

The association between age and nephrosclerosis on renal biopsy among healthy adults
.
Ann Intern Med
 
2010
;
152
:
561
7
.
14.

Roeder
 
SS
,
Stefanska
 
A
,
Eng
 
DG
 et al.   

Changes in glomerular parietal epithelial cells in mouse kidneys with advanced age
.
Am J Physiol Renal Physiol
 
2015
;
309
:
F164
78
.
15.

Hottel
 
H
,
Hoste
 
L
,
Delanaye
 
P
.

Abnormal glomerular filtration rate in children, adolescents and young adults starts below 75 mL/min/1.73 m2
.
Pediatr Nephrol
 
2015
;
30
:
821
8
.
16.

Glassock
 
R
,
Delanaye
 
P
,
El Nahas
 
M
.

An age-calibrated classification of chronic kidney disease
.
JAMA
 
2015
;
314
:
559
60
.
17.

Wiggins
 
JE
,
Goyal
 
M
,
Sanden
 
SK
 et al.   

Podocyte hypertrophy, “adaptation,” and “decompensation” associated with glomerular enlargement and glomerulosclerosis in the aging rat: prevention by calorie restriction
.
J Am Soc Nephrol
 
2005
;
16
:
2953
66
.
18.

Cai
 
W
,
He
 
JC
,
Zhu
 
L
 et al.   

Reduced oxidant stress and extended lifespan in mice exposed to a low glycotoxin diet: association with increased AGER1 expression
.
Am J Pathol
 
2007
;
170
:
1893
902
.
19.

Betjes
 
MG
.

Immune cell dysfunction and inflammation in end-stage renal disease
.
Nat Rev Nephrol
 
2013
;
9
:
255
65
.
20.

De Nicola
 
L
,
Minutolo
 
R
,
Chiodini
 
P
 et al.   

The effect of increasing age on the prognosis of non-dialysis patients with chronic kidney disease receiving stable nephrology care
.
Kidney Int
 
2012
;
82
:
482
8
.
21.

Elsherbiny
 
HE
,
Alexander
 
MP
,
Kremers
 
WK
 et al.   

Nephron hypertrophy and glomerulosclerosis and their association with kidney function and risk factors among living kidney donors
.
Clin J Am Soc Nephrol
 
2014
;
9
:
1892
902
.
22.

Chen
 
J
,
Chen
 
M
,
Fogo
 
A
 et al.   

mVps34 deletion in podocytes causes glomerulosclerosis by disrupting intracellular vesicle trafficking
.
J Am Soc Nephrol
 
2013
;
24
:
198
207
.
23.

Yamamoto-Nonaka
 
K
,
Koike
 
M
,
Asanuma
 
K
 et al.   

Cathepsin D in podocytes is important in the pathogenesis of proteinuria and CKD
.
J Am Soc Nephrol
 
2016
;
27
:
2685
700
.
24.

Verzola
 
D
,
Saio
 
M
,
Picciotto
 
D
 et al.   

Cellular senescence is associated with faster progression of focal segmental glomerulosclerosis
.
Am J Nephrol
 
2020
;
51
:
950
8
.
25.

Hartleben
 
B
,
Godel
 
M
,
Meye-Schwesinger
 
C
 et al.   

Autophagy influences glomerular disease susceptibility and maintains podocyte homeostasis in aging mice
.
J Clin Invest
 
2010
;
120
:
1084
96
.
26.

Lan
 
F
,
Cacicedo
 
JM
,
Ruderman
 
N
 et al.   

SIRT1 modulation of the acetylation status, cytosolic localization, and activity of LKB1. Possible role in AMP-activated protein kinase activation
.
J Biol Chem
 
2008
;
283
:
27628
35
.
27.

Chuang
 
PY
,
Cai
 
W
,
Li
 
X
 et al.   

Reduction in podocyte SIRT1 accelerates kidney injury in aging mice
.
Am J Physiol Renal Physiol
 
2017
;
313
:
F621
8
.
28.

Serna
 
JDC
,
Amaral
 
AG
,
Caldeira da Silva
 
CC
 et al.   

Regulation of kidney mitochondrial function by caloric restriction
.
Am J Physiol Renal Physiol
 
2022
;
323
:
F92
106
.
29.

Zullo
 
A
,
Simone
 
E
,
Grimaldi
 
M
 et al.   

Sirtuins as mediator of the anti-ageing effects of calorie restriction in skeletal and cardiac muscle
.
Int J Mol Sci
 
2018
;
19
:
928
.
30.

Munoz-Espin
 
D
,
Serrano
 
M
.

Cellular senescence: from physiology to pathology
.
Nat Rev Mol Cell Biol
 
2014
;
15
:
482
96
.
31.

D'Adda di Fagagna
 
F
,
Reaper
 
PM
,
Clay-Farrace
 
L
 et al.   

A DNA damage checkpoint response in telomere-initiated senescence
.
Nature
 
2003
;
426
:
194
.
32.

Tchkonia
 
T
,
Zhu
 
Y
,
Van Deursen
 et al.   

Cellular senescence and the senescent secretory phenotype: therapeutic opportunities
.
J Clin Invest
 
2013
;
123
:
966
72
.
33.

Gandolfo
 
MT
,
Verzola
 
D
,
Salvatore
 
F
 et al.   

Gender and the progression of chronic renal diseases: does apoptosis make the difference?
 
Minerva Urol Nefrol
 
2004
;
56
:
1
14
.
34.

Esposito
 
P
,
Verzola
 
D
,
Saio
 
M
 et al.   

The contribution of muscle innate immunity to uremic cachexia
.
Nutrients
 
2023
;
15
:
2832
.
35.

Huang
 
W
,
Hickson
 
LJ
,
Eirin
 
A
 et al.   

Cellular senescence: the good, the bad and the unknown
.
Nat Rev Nephrol
 
2022
;
18
:
611
27
.
36.

Verzola
 
D
,
Gandolfo
 
MT
,
Gaetani
 
G
 et al.   

Accelerated senescence in the kidneys of patients with type 2 diabetic nephropathy
.
Am J Physiol Renal Physiol
 
2008
;
295
:
F1563
73
.
37.

Kitada
 
K
,
Nakano
 
D
,
Ohsaki
 
H
 et al.   

Hyperglycemia causes cellular senescence via a SGLT2- and p21-dependent pathway in proximal tubules in the early stage of diabetic nephropathy
.
J Diabetes Complications
 
2014
;
28
:
604
11
.
38.

Childs
 
BG
,
Durik
 
M
,
Baker
 
DJ
 et al.   

Cellular senescence in aging and age-related disease: from mechanisms to therapy
.
Nat Med
 
2015
;
21
:
1424
35
.
39.

Lee
 
BY
,
Han
 
JA
,
Im
 
JS
 et al.   

Senescence-associated β-galactosidase is lysosomal β-galactosidase
.
Aging Cell
 
2006
;
5
:
187
95
.
40.

Dimri
 
GP
,
Lee
 
X
,
Basile
 
G
 et al.   

A biomarker that identifies senescent human cells in culture and in aging skin in vivo
.
Proc Natl Acad Sci USA
 
1995
;
92
:
9363
7
.
41.

Coppé
 
JP
,
Desprez
 
PY
,
Krtolica
 
A
 et al.   

The senescence-associated secretory phenotype: the dark side of tumor suppression
.
Annu Rev Pathol
 
2010
;
5
:
99
118
.
42.

Yang
 
L
,
Besschetnova
 
TY
,
Brookk
 
CR
 et al.   

Epithelial cell cycle arrest in G2/M mediates kidney fibrosis after injury
.
Nat Med
 
2010
;
16
:
535
43
.
43.

Luo
 
C
,
Zhou
 
S
,
Zhou
 
Z
 et al.   

Wnt9a promotes renal fibrosis by accelerating cellular senescence in tubular epithelial cells
.
J Am Soc Nephrol
 
2018
;
29
:
1238
56
.
44.

Passos
 
JF
,
Nelson
 
G
,
Wang
 
C
 et al.   

Feedback between p21 and reactive oxygen production is necessary for cell senescence
.
Mol Syst Biol
 
2010
;
6
:
347
.
45.

Moldogazieva
 
NT
,
Mokhosoev
 
IM
,
Mel'nikova
 
TI
 et al.   

Oxidative stress and advanced lipoxidation and glycation end products (ALEs and AGEs) in aging and age-related diseases
.
Oxid Med Cell Longev
 
2019
;
2019
:
3085756
.
46.

Freund
 
A
,
Orjalo
 
AV
,
Desprez
 
PY
 et al.   

Inflammatory networks during cellular senescence: causes and consequences
.
Trends Mol Med
 
2010
;
16
:
238
46
.
47.

Kulkarni
 
AS
,
Aleksic
 
S
,
Berger
 
DM
 et al.   

Geroscience-guided repurposing of FDA-approved drugs to target aging: a proposed process and prioritization
.
Aging Cell
 
2022
;
21
:
e13596
.
48.

Uthman
 
L
,
Baartscheer
 
A
,
Bleijlevens
 
B
 et al.   

Class effects of SGLT2 inhibitors in mouse cardiomyocytes and hearts: inhibition of Na/H exchanger, lowering of cytosolic Na and vasodilatation
.
Diabetologia
 
2018
;
61
:
722
6
.
49.

Packer
 
M
.

Activation and inhibition of sodium-hydrogen exchanger is a mechanism that links the pathophysiology and treatment of diabetes mellitus with that of heart failure
.
Circulation
 
2017
;
136
:
1548
59
.
50.

Verma
 
S
,
McMurray
 
JJV
,
Cherney
 
DZI
.

The metabolodiuretic promise of sodium-dependent glucose cotransporter 2 inhibition: the search for the sweet spot in heart failure
.
JAMA Cardiol
 
2017
;
2
:
939
40
.
51.

Santos-Gallego
 
CG
,
Requena-Ibanez
 
JA
,
San Antonio
 
R
 et al.   

Empagliflozin ameliorates adverse left ventricular remodeling in nondiabetic heart failure by enhancing myocardial energetics
.
J Am Coll Cardiol
 
2019
;
73
:
1931
44
.
52.

Li
 
L
,
Wang
 
Z
,
Zuo
 
Z
.

Chronic intermittent fasting improves cognitive functions and brain structures in mice
.
PLoS One
 
2013
;
8
:
e66069
.
53.

Ma
 
D
,
Wang
 
AC
,
Parikh
 
I
 et al.   

Ketogenic diet enhances neurovascular function with altered gut microbiome in young healthy mice
.
Sci Rep
 
2018
;
8
:
6670
.
54.

Baker
 
DJ
,
Wijshake
 
T
,
Tchkonia
 
T
 et al.   

Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders
.
Nature
 
2011
;
479
:
232
6
.
55.

Roos
 
CM
,
Zhang
 
B
,
Palmer
 
AK
 et al.   

Chronic senolytic treatment alleviates established vasomotor dysfunction in aged or atherosclerotic mice
.
Aging Cell
 
2016
;
15
:
973
7
.
56.

Baar
 
MP
,
Brandt
 
R
,
Putavet
 
D
 et al.   

Targeted apoptosis of senescent cells restores tissue homeostasis in response to chemotoxicity and aging
.
Cell
 
2017
;
169
:
132
147.e16
.
57.

Hoong
 
C
,
Chua
 
M
.

SGLT2 inhibitors as calorie restriction mimetics: insights on longevity pathways and age-related diseases
.
Endocrinology
 
2021
;
162
:
bqab079
.
58.

Brown
 
E
,
Rajeev
 
SP
,
Cuthbertson
 
DJ
 et al.   

A review of the mechanism of action, metabolic profile and haemodynamic effects of sodium-glucose co-transporter-2 inhibitors
.
Diabetes Obes Metab
 
2019
;
21
:
9
18
.
59.

Tomita
 
I
,
Kume
 
S
,
Sugahara
 
S
 et al.   

SGLT2 inhibition mediates protection from diabetic kidney disease by promoting ketone body-induced mTORC1 inhibition
.
Cell Metab
 
2020
;
32
:
404
19.e6
.
60.

Faridvand
 
Y
,
Kazemzadeh
 
H
,
Vahedian
 
V
 et al.   

Dapagliflozin attenuates high glucose-induced endothelial cell apoptosis and inflammation through AMPK/SIRT1 activation
.
Clin Exp Pharmacol Physiol
 
2022
;
49
:
643
51
.
61.

Mattison
 
JA
,
Roth
 
GS
,
Beasley
 
TM
 et al.   

Impact of caloric restriction on health and survival in rhesus monkeys from the NIA study
.
Nature
 
2012
;
489
:
318
21
.
62.

Maegawa
 
S
,
Lu
 
Y
,
Tahara
 
T
 et al.   

Caloric restriction delays age-related methylation drift
.
Nat Commun
 
2017
;
8
:
539
.
63.

Hahn
 
O
,
Grönke
 
S
,
Stubbs
 
TM
 et al.   

Dietary restriction protects from age-associated DNA methylation and induces epigenetic reprogramming of lipid metabolism
.
Genome Biol
 
2017
;
18
:
56
.
64.

Waziry
 
R
,
Ryan
 
CP
,
Corcoran
 
DL
 et al.   

Effect of long-term caloric restriction on DNA methylation measures of biological aging in healthy adults from the CALERIE trial
.
Nat Aging
 
2023
;
3
:
248
57
.
65.

Shintani
 
H
,
Shintani
 
T
,
Ashida
 
H
 et al.   

Calorie restriction mimetics: upstream-type compounds for modulating glucose metabolism
.
Nutrients
 
2018
;
10
:
1821
.
66.

Fonseca-Correa
 
JI
,
Correa-Rotter
 
R
.

Sodium-glucose cotransporter 2 inhibitors mechanisms of action: a review
.
Front Med (Lausanne)
 
2021
;
8
:
777861
.
67.

Hipkiss
 
AR
.

Mitochondrial dysfunction, proteotoxicity, and aging: causes or effects, and the possible impact of NAD+-controlled protein glycation
.
Adv Clin Chem
 
2010
;
50
:
123
50
.
68.

Marchio
 
P
,
Guerra-Ojeda
 
S
,
Vila
 
JM
 et al.   

Targeting early atherosclerosis: a focus on oxidative stress and inflammation
.
Oxid Med Cell Longev
 
2019
;
2019
:
8563845
.
69.

Jha
 
J
,
Banal
 
C
,
Chow
 
B
 et al.   

Diabetes and kidney disease: role of oxidative stress
.
Antioxid Redox Signal
 
2016
;
25
:
657
84
.
70.

Sun
 
Y
,
Ge
 
X
,
Li
 
X
 et al.   

High-fat diet promotes renal injury by inducing oxidative stress and mitochondrial dysfunction
.
Cell Death Dis
 
2020
;
11
:
914
.
71.

Østergaard
 
J
,
Cooper
 
M
,
Jandeleit-Dahm
 
K
 

Targeting oxidative stress and anti-oxidant defence in diabetic kidney disease
.
J Nephrol
 
2020
;
33
:
917
29
.
72.

Eleftheriadis
 
T
,
Pissas
 
G
,
Filippidis
 
G
 et al.   

Dapagliflozin prevents high-glucose-induced cellular senescence in renal tubular epithelial cells
.
Int J Mol Sci
 
2022
;
23
:
16107
.
73.

Ishibashi
 
Y
,
Matsui
 
T
,
Yamagishi
 
S
 et al.   

A highly selective inhibitor of SGLT2 blocks proinflammatory and proapoptotic effects of glucose overload on proximal tubular cells partly by suppressing oxidative stress generation
.
Horm Metab Res
 
2016
;
48
:
191
5
.
74.

Mone
 
P
,
Varzideh
 
F
,
Jankauskas
 
SS
 et al.   

SGLT2 inhibition via empagliflozin improves endothelial function and reduces mitochondrial oxidative stress: insights from frail hypertensive and diabetic patients
.
Hypertension
 
2022
;
79
:
1633
43
.
75.

Park
 
SH
,
Belcastro
 
E
,
Hasan
 
H
 et al.   

Angiotensin II-induced upregulation of SGLT1 and 2 contributes to human microparticle-stimulated endothelial senescence and dysfunction: protective effect of gliflozins
.
Cardiovasc Diabetol
 
2021
;
20
:
65
.
76.

Trnovska
 
J
,
Svoboda
 
P
,
Pelantova
 
H
 et al.   

Complex positive effects of SGLT-2 inhibitor empagliflozin in the liver, kidney and adipose tissue of hereditary hypertriglyceridemic rats: possible contribution of attenuation of cell senescence and oxidative stress
.
Int J Mol Sci
 
2021
;
22
:
10606
.
77.

Fang
 
R
,
Chen
 
J
,
Long
 
J
 et al.   

Empagliflozin improves kidney senescence induced by D-galactose by reducing sirt1-mediated oxidative stress
.
Biogerontology
 
2023
;
4
:
771
82
.
78.

Kim
 
MN
,
Moon
 
JH
,
Cho
 
YM
.

Sodium-glucose cotransporter-2 inhibition reduces cellular senescence in the diabetic kidney by promoting ketone body-induced NRF2 activation
.
Diabetes Obes Metab
 
2021
;
23
:
2561
71
.
79.

La Grotta
 
R
,
de Candia
 
P
,
Olivieri
 
F
 et al.   

Anti-inflammatory effect of SGLT-2 inhibitors via uric acid and insulin
.
Cell Mol Life Sci
 
2022
;
79
:
273
.
80.

Jasleen
 
B
,
Vishal
 
G
,
Sameera
 
M
 et al.   

Sodium-glucose cotransporter 2 (SGLT2) inhibitors: benefits versus risk
.
Cureus
 
2023
;
15
:
e33939
.
81.

Fu
 
J
,
Xu
 
H
,
Wu
 
F
 et al.   

Empagliflozin inhibits macrophage inflammation through AMPK signaling pathway and plays an anti-atherosclerosis role
.
Int J Cardiol
 
2022
;
367
:
56
62
.
82.

Zhao
 
XP
,
Chang
 
SY
,
Pang
 
Y
 et al.   

Hedgehog interacting protein activates sodium-glucose cotransporter 2 expression and promotes renal tubular epithelial cell senescence in a mouse model of type 1 diabetes
.
Diabetologia
 
2023
;
66
:
223
40
.
83.

La Grotta
 
R
,
Frigè
 
C
,
Matacchione
 
G
 et al.   

Repurposing SGLT-2 inhibitors to target aging: available evidence and molecular mechanisms
.
Int J Mol Sci
 
2022
;
23
:
12325
.
84.

Dai
 
L
,
Qureshi
 
AR
,
Witasp
 
A
 et al.   

Early vascular ageing and cellular senescence in chronic kidney disease
.
Comput Struct Biotechnol J
 
2019
;
17
:
721
9
.
85.

Schroth
 
J
,
Thiemermann
 
C
,
Henson
 
SM
.

Senescence and the aging immune system as major drivers of chronic kidney disease
.
Front Cell Dev Biol
 
2020
;
8
:
564461
.
This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial License (https://creativecommons.org/licenses/by-nc/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited. For commercial re-use, please contact journals.permissions@oup.com

Comments

0 Comments
Submit a comment
You have entered an invalid code
Thank you for submitting a comment on this article. Your comment will be reviewed and published at the journal's discretion. Please check for further notifications by email.