Next Article in Journal
Systematic Identification and Functional Analysis of the Hypericum perforatum L. bZIP Gene Family Indicating That Overexpressed HpbZIP69 Enhances Drought Resistance
Next Article in Special Issue
Transfection of Vein Grafts with Early Growth Response Factor-1 Oligodeoxynucleotide Decoy: Effects on Stem-Cell Genes and Toll-like Receptor-Mediated Inflammation
Previous Article in Journal
A Distinct Microglial Cell Population Expressing Both CD86 and CD206 Constitutes a Dominant Type and Executes Phagocytosis in Two Mouse Models of Retinal Degeneration
Previous Article in Special Issue
Glucagon-like Peptide-1 Receptor Activation Reduces Pulmonary Vein Arrhythmogenesis and Regulates Calcium Homeostasis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Impact of SGLT2 Inhibitors in the Heart and Kidneys Regardless of Diabetes Status

by
Jennifer Matthews
1,†,
Lakshini Herat
1,†,
Markus P. Schlaich
2,3 and
Vance Matthews
1,*
1
Royal Perth Hospital Unit, Dobney Hypertension Centre, School of Biomedical Sciences, University of Western Australia, Crawley, WA 6009, Australia
2
Royal Perth Hospital Unit, Dobney Hypertension Centre, School of Medicine, University of Western Australia, Crawley, WA 6009, Australia
3
Department of Cardiology and Department of Nephrology, Royal Perth Hospital, Perth, WA 6000, Australia
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(18), 14243; https://doi.org/10.3390/ijms241814243
Submission received: 17 August 2023 / Revised: 12 September 2023 / Accepted: 15 September 2023 / Published: 18 September 2023

Abstract

:
Chronic Kidney Disease (CKD) and Cardiovascular Disease (CVD) are two devastating diseases that may occur in nondiabetics or individuals with diabetes and, when combined, it is referred to as cardiorenal disease. The impact of cardiorenal disease on society, the economy and the healthcare system is enormous. Although there are numerous therapies for cardiorenal disease, one therapy showing a great deal of promise is sodium-dependent glucose cotransporter 2 (SGLT2) inhibitors. The SGLT family member, SGLT2, is often implicated in the pathogenesis of a range of diseases, and the dysregulation of the activity of SGLT2 markedly effects the transport of glucose and sodium across the luminal membrane of renal cells. Inhibitors of SGLT2 were developed based on the antidiabetic action initiated by inhibiting renal glucose reabsorption, thereby increasing glucosuria. Of great medical significance, large-scale clinical trials utilizing a range of SGLT2 inhibitors have demonstrated both metabolic and biochemical benefits via numerous novel mechanisms, such as sympathoinhibition, which will be discussed in this review. In summary, SGLT2 inhibitors clearly exert cardio-renal protection in people with and without diabetes in both preclinical and clinical settings. This exciting class of inhibitors improve hyperglycemia, high blood pressure, hyperlipidemia and diabetic retinopathy via multiple mechanisms, of which many are yet to be elucidated.

1. Introduction

Both Chronic Kidney Disease (CKD) and Cardiovascular Disease (CVD) are two of the most prevalent diseases globally. Chronic Kidney Disease is a progressive condition that affects over 10% (over 800 million) of the global population [1] and may ultimately lead to the final stage of kidney disease, known as end-stage kidney disease. However, according to the World Health Organization, CVD is the leading cause of death globally.
An even more disturbing phenomenon occurring now is the combination of cardiac and renal dysfunction, known as cardiorenal disease. In fact, it is now known that many individuals dealing with CKD do not actually reach the stage of dialysis, because they die of heart disease [2]. This is the case because CKD is multifactorial and could be related to a variety of reasons, including impaired coronary flow reserve (ratio of the maximal or hyperemic flow down a coronary vessel to the resting flow), reduced aortic compliance (the ability of the arterial wall to distend and increase volume with increasing transmural pressure), increase in the level of angiotensin II and changes in the concentrations of essential vitamins like potassium, calcium and magnesium, as well as fibrosis in the hearts of patients on dialysis [3].
Cardiovascular Diseases (CVD) are the leading cause of disease burden throughout the world, with prevalent cases of CVD doubling from 271 million people in 1990 to 523 million people in 2019 and the number of CVD deaths increasing from 12.1 million in 1990 to 18.6 million in 2019, and it is more fatal in men than women [4]. Treatment of CVD is complex, as there are a multitude of underlying causes of cardiovascular death, such as ischemic heart disease, stroke, hypertensive heart disease, cardiomyopathy/myocarditis, atherosclerosis, aortic aneurysm and peripheral artery disease. Alarmingly, modifiable risk factors such as high systolic blood pressure, high fasting plasma glucose, high LDL cholesterol, high BMI and poor diet choices are also on the rise [4]. In a study including 197 countries, the total costs of heart failure alone in 2012 was USD $108 billion [5].
Diabetes is one of the leading causes of death and disability worldwide, with 529 million people living with this condition as of 2021, resulting in health expenditures of USD $966 billion globally, and they are forecasted to reach more than USD $1054 billion by 2045. Type 2 diabetes makes up 96% of all diabetes cases worldwide, and it is equally prevalent in both males and females [6]. Diabetic Kidney Disease (DKD) develops in approximately 40% of patients with type 2 diabetes (T2D) and 30% of patients with type 1 diabetes (T1D), and it is the leading cause of chronic kidney disease (CKD) and end-stage renal disease [7,8]. The mortality risk associated with DKD has increased by 31.1%, and it increases with worsening disease severity [9]. It is reported that DKD affects males and females equally, and it rarely develops before 10 years of duration of T1D [10]. The humanistic, societal and economic impact of DKD is enormous. It places a significant burden on the health care system and seriously affects the physical health and quality of life of patients [11]. Globally, in 2019, there were 2.6 million cases of CKD due to diabetes mellitus [12].
At present, there are a multitude of therapies that are used for cardio-renal disease. Some of these include pharmacological agents such as diuretics, vasodilators, angiotensin-converting enzyme (ACE) inhibitors or angiotensin receptor blockers (ARBs) [13]. In fact, it has been shown that in an Italian diabetic population affected by both DKD and diabetic retinopathy, multifactorial intervention using a combination of angiotensin-converting enzyme inhibitors and angiotensin II receptor blockers resulted in improved outcomes [14]. Although standard treatments have been found to slow the progression of CKD, they do not halt the disease. Therefore, alternative treatments may be required, such as the non-steroidal mineralocorticoid receptor antagonist finerenone, which is used to treat DKD with albuminuria [15]. Other emerging therapies that are currently undergoing clinical trials include endothelin receptor-A antagonists, complement inhibition, Janus kinase (JAK) inhibition, chemokine inhibition, renal denervation and, of course, the topic of this review, sodium glucose cotransporter 2 (SGLT2) inhibitors.

2. What Are Sodium Glucose Cotransporters?

The entry of glucose into cells is regulated by facilitative glucose transporters (GLUTs) and sodium-dependent glucose cotransporters (SGLTs). Of the SGLT family, SGLT1 and SGLT2 are frequently investigated in a range of disease settings [16], as they play key roles in the transport of glucose and sodium across the brush-border membrane of intestinal and renal cells [17]. Although SGLT1 is less researched than SGLT2, it is more widely expressed throughout the body and is found predominantly in the small intestine [18,19,20] (Figure 1; Table 1), and it only accounts for 5–10% of the glucose reabsorption in the kidneys. Our studies have shown that heightened Sympathetic Nervous System (SNS) activity upregulates this SGLT1 expression, and therefore, inhibition of this protein could also be beneficial in treating cardiometabolic disorders [21]. SGLT2 is a high-capacity, low-affinity glucose cotransporter, mainly found in the S1 and S2 segments of the renal convoluted proximal tubules (Figure 1; Table 1), and it is required for the reabsorption of a majority of the glucose (~90–95%) filtered by the kidneys [22,23].
SGLT2 inhibitors (SGLT2i’s) have been developed based on the antidiabetic action initiated by inhibiting renal glucose reabsorption, thereby increasing urinary glucose excretion [24] (Table 2). As demonstrated by several large-scale clinical trials [25,26,27], SGLT2i’s are now recognized to be capable of altering a range of metabolic and biochemical parameters via novel mechanisms, which are discussed in this review, thereby exerting cardio-renal protection in individuals with and without diabetes [28,29].
One critical requirement before SGLT2i’s can be prescribed for the treatment of CKD is a functional glomerular filtration rate (GFR). The glomerular filtration rate is considered the optimal way to measure kidney function. In a healthy kidney, the glomerular filtration rate is 120 mL/min/1.73 m2, but as CKD progresses, it may decline to 60 mL/min/1.73 m2 or less. Interestingly, studies have shown that SGLT2i’s are safe and beneficial to use in patients with a GFR above 20 mL/min/1.73 m2 and can actually be used in subjects that have a GFR of less than 20 mL/min/1.73 m2, as long as they are tolerating it well and are not on dialysis [30]. However, this topic of discussion is controversial, and a number of other specific aspects need to be addressed. Currently, SGLT2 inhibitors can be used in CKD patients up until starting dialysis due to their nephro- and cardioprotective effects. It is also believed that SGLT2 inhibitors may be used safely in hemodialysis and peritoneal dialysis patients, especially if they have additional heart failure.
Table 1. Areas of expression of SGLT1 and SGLT2 in the human body.
Table 1. Areas of expression of SGLT1 and SGLT2 in the human body.
LocationSGLT1SGLT2
Small IntestineApical membrane, K and L cells [19].Not expressed.
EyeRetina [20].Retina, cornea and lens [20].
KidneySection 3 of the proximal tubule. [21].Section 1 and 2 of the proximal tubules [22].
PancreasPancreatic alpha cells [18].Not expressed.
LiverBiliary duct cells [19].Not expressed.
HeartCapillaries [19].Not expressed.
Table 2. Summary of key findings from the characterization of Canagliflozin (CANA), Dapagliflozin (DAPA), Ipragliflozin, Empagliflozin (EMPA), Tofogliflozin and Luseogliflozin [31,32,33].
Table 2. Summary of key findings from the characterization of Canagliflozin (CANA), Dapagliflozin (DAPA), Ipragliflozin, Empagliflozin (EMPA), Tofogliflozin and Luseogliflozin [31,32,33].
Study ParametersKey Findings
Pharmacokinetic
properties
Longest plasma half-life: Canagliflozin.
Longest half-life in the kidney: Dapagliflozin.
Highest distribution in the kidney: Ipragliflozin.
Drug distribution in the kidney suggested to be dependent on chemical structure.
Pharmacodynamic
properties
All SGLT2i’s increased urinary glucose excretion in a dose-dependent manner.
Long-acting SGLT2i’s exhibited persistent action, even 18 h post dose.
Close correlation between the duration of action, plasma drug concentration, drug distribution and kidney retention.
Pharmacologic
properties
Significant reductions in blood glucose and plasma insulin with all SGLT2i’s.
Significant improvement in glucose tolerance with all SGLT2i’s.
Long-acting SGLT2i’s exert stronger anti-hyperglycaemic effects through persistent urine glucose excretion.
Intermediate-acting SGLT2i’s may provide better glycaemic control when administered twice daily.
Anti-diabetic effectsAll SGLT2i’s significantly improved hyperglycaemia and hyperinsulinemia.
All SGLT2i’s significantly increased pancreatic insulin content by prevention of pancreatic exhaustion.
Long-acting SGLT2i’s exert favourable glycaemic control over 24 h and may have slightly enhanced antidiabetic effects compared with intermediate-acting SGLT2i’s.
Effects on diabetic complicationsAll SGLT2i’s exhibited significant improvements/trends in obesity parameters (e.g., body and visceral fat weights, lipid metabolism markers), proinflammatory cytokines and endothelial dysfunction markers.
All SGLT2i’s significantly decreased or showed a decreasing trend in steatohepatitis parameters (e.g., liver weight, plasma levels of liver enzymes) and renal parameters (e.g., creatinine clearance, renal tubular injury markers).
Long-acting SGLT2i’s (0.3 mg/kg) demonstrated slight superiority in comparison with intermediate-acting SGLT2i’s (3 mg/kg) on several parameters (e.g., daily blood glucose control, visceral fat weight).
Blue colouration is related to pharmacological effects. Green colouration is related to anti-diabetic effects. Pink colouration is related to effects on diabetic complications.

3. Use of Selective SGLT2 Inhibition as an Antidiabetic Therapy

There are many studies that have shown the beneficial cardiovascular and renal effects that SGLT2i’s can have in both T1D and T2D. In patients with T2D, SGLT2i treatments are now being considered as the first line of therapy because of their metabolic and cardio-renal benefits. As just mentioned, SGLT2i’s provide benefits by promoting the excretion of glucose in the urine, therefore assisting with a reduction in hyperglycemia and subsequently a decrease in weight. SGLT2 inhibitors also promote both uric acid reduction [34,35] and stimulation of erythropoiesis, which may have cardiovascular and renal effects [34]. However, aside from the metabolic benefits of SGLT2i’s, there are numerous other beneficial protective mechanisms, as outlined below.

3.1. Preclinical Studies

3.1.1. Blood Pressure Reduction

One common complication of patients with diabetes is hypertension, and although the anti-hypertensive therapies have improved over the years, there is definitely still room for improvement. Although SGLT2i’s are not predominantly prescribed as hypertensive medications, research has now shown that SGLT2i’s have overwhelming beneficial effects in patients with both hypertension and diabetes. Mechanisms that may contribute to their anti-hypertensive action are their mild natriuresis, osmotic diuresis and weight-loss effects in patients with diabetes [36]. In human studies, both Empagliflozin (EMPA) [37] and Dapagliflozin (DAPA) [38] were associated with significant reductions in blood pressure in patients with diabetes compared with placebo. We have also demonstrated in our murine studies that SGLT2i’s may promote sympathoinhibition in the kidneys and heart in diabetic mice, and this may be an underlying mechanism for blood pressure reduction [39].

3.1.2. Improved Digestive Health

The intestinal microbiota is aggravated as diabetes progresses and, during the development of DKD, there is an increased imbalance in the gut microbiota. The SGLT2i DAPA has been beneficial in lowering the level of dysbacteriosis and bile acids [40], altering the microbiota composition [41] and reducing succinate levels (pathogenic factor in diabetic retinopathy) [42], while the inhibitor EMPA reduces the lipopolysaccharide (LPS)-producing bacteria and increases the short-chain fatty acid (SCFA)-producing bacteria [43]. Additional inhibitors such as Luseogliflozin have also been found to be beneficial in altering the microbiota. When mice were given the SGLT2 inhibitor Luseogliflozin, there was a significant increase in the abundance of the species Syntrophothermus lipocalidus, Syntrophomonadaceae and Anaerotignum, which are all involved in the biosynthesis of important SCFAs such as acetic acid, propionic acid and butyric acid [44].

3.1.3. Diabetic Retinopathy

Diabetic retinopathy is a common complication associated with diabetes, and the SGLT2i’s EMPA [45] and DAPA [46] have improved diabetic retinopathy in T2D db/db mice. Empagliflozin has also been shown to mitigate ocular edema and microaneurysms in the retina as well as inhibit the mammalian target of raptomycin activation. One of the characteristics of diabetic retinopathy in db/db mice is an increase in acellular capillary numbers [46], and DAPA has been shown to produce a substantial decrease in the acellular capillary numbers compared with placebo. Aside from this, inflammation is another main factor associated with diabetic retinopathy. According to these studies, both EMPA and DAPA have been shown to downregulate inflammatory and angiogenic factors, such as Tumor Necrosis Factor-α (TNF-α) [45,46], Interleukin 1-β (IL1-β) [46], Interleukin 6 (IL-6) [45], Vascular Cell Adhesion Molecule 1 (VCAM-1) [45] and Vascular Endothelial Growth Factor (VEGF) [45] in the retina.

3.1.4. Kidney Health

There have been numerous preclinical studies conducted which show the renoprotective effects of SGLT2i’s. Dapagliflozin treatment in db/db mice halted the progressive increases in albumineria and glomerulosclerosis [47], while in the Otsuka Long–Evans Tokushima Fatty (OLETF) T2D rat model, DAPA was shown to reverse renal oxidative stress markers as well as attenuate inflammatory cell infiltration, mesangial widening, interstitial fibrosis and total collagen content [48]. Luseogliflozin treatment in a T2D nephropathy rat model has been shown to prevent the fall in GFR and reduce the degree of glomerular injury, renal fibrosis and tubular necrosis compared with those on vehicle or insulin alone [49]. Empagliflozin treatment has also been found to ameliorate albuminuria and glomerular injury in db/db mice [50].

3.1.5. Cardiovascular Benefits

There have been many preclinical animal studies showing the cardioprotective effects of SGLT2i’s in T2D. When EMPA was administered to db/db mice over a 10-week period, it was shown to significantly ameliorate cardiac interstitial fibrosis, pericoronary arterial thickening, cardiac macrophage infiltration and the impairment of vascular dilation [50]. Further discussion of SGLT2i-mediated cardiovascular benefits is presented later in the review.

3.1.6. Improved Cognitive Function in T2D

One of the most underrecognized but life-changing effects of T2D is the decline in cognitive function. Therefore, the question as to whether SGLT2i’s reduce the cognitive impairment associated with T2D has been investigated in numerous animal studies. Empagliflozin has been shown to significantly prevent the impairment of cognitive function in T2D db/db mice due to its ability to attenuate cerebral oxidative stress, as well as increase cerebral Brain-Derived Neurotrophic Factor (BDNF) levels [50].

3.2. Human Clinical Trials

There are a multitude of clinical trials that have been conducted utilizing the SGLT2 inhibitors EMPA, DAPA, Canagliflozin (CANA) and Ertugliflozin, and they are outlined below (Table 3).

3.2.1. Empagliflozin

The groundbreaking clinical trial assessing the effectiveness of the SGLT2 inhibitor EMPA in preventing Cardiovascular Disease in people with diabetes is the Empagliflozin Cardiovascular Outcome Event Trial in type 2 diabetes Mellitus Patients–Removing Excess Glucose (EMPA-REG) [25,51]. This trial demonstrated that when patients who had T2D as well as a high risk of cardiovascular events were treated with EMPA, mortality (3.7% vs. 5.9%) and hospitalizations (2.7% vs. 4.1%) due to heart failure were reduced compared with placebo [25]. Empagliflozin also promoted a slowing down of the progression of kidney disease (12.7% vs. 18.8%), as well as a significantly lower risk of clinically relevant renal events, including renal replacement therapy (0.3% vs. 0.6%) versus placebo controls [51].

3.2.2. Dapagliflozin

When it comes to cardiovascular and kidney disease, there are two main clinical trials evaluating the effectiveness of the SGLT2 inhibitor DAPA in people with diabetes. One major cause of Cardiovascular Disease is atherosclerosis, which may be caused by both traditional and nontraditional risk factors [4] (Figure 2). The Dapagliflozin Effect on Cardiovascular Events–Thrombolysis in Myocardial Infarction 58 (DELCARE-TIMI 58) trial [26] recruited people who had both diabetes and atherosclerotic Cardiovascular Disease, and although Dapagliflozin did not result in a higher or lower rate of Major Adverse Cardiovascular Events (MACE), it did lead to a lower rate of cardiovascular death or hospitalization for heart failure (4.9% vs. 5.8%) compared with placebo. In addition to this trial, the Dapagliflozin and Prevention of Adverse Outcomes in Chronic Kidney Disease (DAPA-CKD) study [57] found that not only did DAPA reduce the risk of a declining GFR level (5.2% vs. 9.3%), it decreased the risk of end-stage kidney disease (5.1% vs. 7.5%) and death from renal causes (<0.1% vs. 0.3%) versus controls.

3.2.3. Canagliflozin

There are also two main clinical trials determining the effectiveness of the SGLT2 inhibitor CANA on Cardiovascular Disease in people with type 2 diabetes. The first is the Canagliflozin Cardiovascular Assessment Study (CANVAS) [27], where it was found that CANA not only reduced the risk of major adverse cardiovascular events in T2D patients at an increased risk of Cardiovascular Disease, it also offered renoprotective benefits, such as the regression of albuminuria levels (8.9% vs. 12.9%) and a decrease in the need for renal-replacement therapy or death (0.55% vs. 0.9%) when compared with placebo. In the second trial, known as the Canagliflozin and Renal Endpoints in Diabetes with Established Nephropathy Clinical Evaluation (CREDENCE) study [52], it was concluded that in patients who had both T2D and kidney disease, the risk of kidney failure and cardiovascular events was lower in the CANA group compared with the placebo group.

3.2.4. Ertugliflozin

The final major SGLT2 inhibitor clinical trial is the Evaluation of Ertugliflozin efficacy and Safety Cardiovascular Outcomes (VERTIS) study [53], where the effect of Ertugliflozin in patients with both T2D and Cardiovascular Disease was investigated. It was found that not only did this inhibitor reduce the risk for first and total hospitalizations for heart failure, it also reduced the risk of death from heart failure/Cardiovascular Disease (8.1% vs. 9.1%) versus controls.

4. Use of Selective SGLT2 Inhibition as a Nondiabetic Therapy

Although SGLT2 inhibitors were originally utilized for the treatment of type 2 diabetes specifically, this drug class is now also being used in people without diabetes to assist in protection against cardiorenal disease (Table 3).

4.1. Preclinical Studies

4.1.1. Modulation of Sympathetic Nervous System Activity

The SNS is a driver of hypertension, hyperglycemia and chronic kidney disease and, remarkably, our team and others have shown that SGLT2i’s may protect the kidneys and the heart through their sympathoinhibitory abilities [22,62]. In Apo E−/− mice, it was discovered that with the use of EMPA, norepinephrine (NE), the marker of heightened sympathetic activity was partially inhibited [63]. Our study with neurogenically hypertensive BPH/2J mice showed that DAPA lowered the NE and tyrosine hydroxylase (TH) levels in heart and kidneys [22].
Interestingly, the SNS performs diverse functions in different tissues. For instance, our team discovered that SGLT2 inhibition in the white adipose tissue (WAT) actually promoted sympathoexcitation and beiging [64]. In a cohort of BPH/2J mice, DAPA was administered via oral gavage, and the mice were found to have an increased level of tyrosine hydroxylase (TH) and norepinephrine expression in the WAT [64]. This is particularly exciting, as beiging is a process that typically occurs during fasting or exercise. During beiging, there is an upregulation of Uncoupling Protein 1 (UCP1), but there is also an increase in the molecules’ α-Aminoisobutyric acid, irisin and fibroblast growth factor 21 (FGF-21) [65].

4.1.2. Reductions in Blood Pressure

Studies have highlighted that SGLT2 inhibition promotes remarkable reductions in blood pressure. In our neurogenically hypertensive BPH/2J mice, treatment with DAPA significantly reduced systolic and diastolic blood pressure, as well as mean arterial pressure (Figure 3). This blood pressure reduction also correlated with a remarkable sympathoinhibitory effect in the kidneys [22].

4.1.3. Inflammation Control

One contributing pathogenic factor when it comes to both Chronic Kidney Disease and heart disease is that of inflammation. In Apo E−/− mice, EMPA, DAPA and CANA were all found to reduce inflammatory markers. Empagliflozin reduced IL-1β and IL-6 [63] and DAPA reduced IL-1β, Interleukin 18 (IL-18) and the inflammasome marker NOD-LRR and Pyrin domain containing protein 3 (NLRP3) [66], while CANA reduced VCAM-1 and the Monocyte chemotactic protein-1 (MCP-1) inflammatory protein while increasing the TIMP metallopeptidase inhibitor 1 (TIMP-1) [24]. In our neurogenically hypertensive BPH/2J mice, we found that DAPA treatment reduced inflammation in the heart by significantly reducing the inflammatory cytokine IL-6, while at the same time increasing the anti-inflammatory cytokine Interleukin 10 (IL-10) [22].

4.1.4. Increases in Ketone Levels

More and more research is now showing that ketone bodies are an efficient substrate for the heart [67] and the kidneys [68]. Empagliflozin has been shown in multiple studies to increase ketone bodies in patients with chronic heart failure [69], as well as in both ZSF1 [70] and ApoE knockout mice [71]. Although ketoacidosis is a topic of concern when it comes to utilization of SGLT2i’s, it is of much greater concern for those with T1D as opposed to T2D. Although ketoacidosis can occur in T2D patients not on insulin, a human study has shown that when T2D patients were admitted to hospital with COVID-19, those who were administered SGLT2i therapy were at no greater risk of ketoacidosis than those not taking the inhibitors [72]. The subject of whether ketones increase or decrease SNS activity is still a controversy. In a preclinical study, it was found that the ketone body β-hydroxybutyrate suppresses SNS activity by antagonizing G-protein-coupled receptor 41 (GPR41) [73]. However, an alternative study involving rats showed that increased ketone body utilization did not suppress SNS activity and may stimulate it similarly to results seen with carbohydrates or fats [74]. In our future studies, we aim to assess whether ketone bodies suppress SNS and, consequently, how they may promote cardiorenal health.

4.1.5. Improved Cardiovascular Health

Cardiomyopathy is a condition caused by inflammation in the cardiomyocytes, often through HFD consumption. In an in vitro study involving cultured rat H9c2 cardiomyocyte cells, treatment with the SGLT2 inhibitor DAPA attenuated hypertrophy, fibrosis and apoptosis. Furthermore, HFD-fed mice were administered DAPA, and this treatment improved the lipid profile as well as alleviated HFD-induced cardiac dysfunction and cardiac inflammation [75].

4.1.6. Steatosis and Insulin Resistance

It is well known that SGLT2i’s lead to blood glucose reductions and weight loss in T2D. However, the influence of SGLT2 inhibition on high-fat diet (HFD)-induced obesity and insulin resistance is less well known. Preclinical studies on C57BL/6J HFD mice found that when given the SGLT2i’s EMPA [76,77] or CANA [78], not only did it increase the urinary excretion of glucose, it also increased weight loss and attenuated hepatic steatosis in the treatment groups compared with vehicle. In addition, EMPA suppressed the HFD-induced weight gain by enhancing fat utilization and browning and attenuated obesity-induced inflammation and insulin resistance [76].

4.2. Human Clinical Trials

4.2.1. Empagliflozin

Empagliflozin is an SGLT2 inhibitor that has been approved for use in people with diabetes, but it is now being increasingly used in people who do not have diabetes but do have cardiovascular and/or renal disease. In the Empagliflozin Outcome Trial in Patients with Chronic Heart Failure and a Reduced Ejection Fraction (EMPEROR-REDUCED) [56], it was concluded that patients on EMPA had a 25% lower risk of cardiovascular death or hospitalization for heart failure (19.4% vs. 24.7%) than subjects on placebo, irrespective of whether the patients had diabetes or not. Another trial, known as the Empagliflozin Outcome Trial in Patients with Chronic Heart Failure with Preserved Ejection Fraction (EMPEROR-PRESERVED) [58], determined that EMPA treatment reduced the risk and severity of a broad range of inpatient and outpatient worsening heart failure events, including a reduction in hospitalization for heart failure (8.6% vs. 11.8%) and death from cardiovascular causes (7.3% vs. 8.2%) versus controls, therefore offering protective benefits. Aside from the EMPEROR trials, another clinical trial, known as Empagliflozin in Acute Myocardial Infarction (EMMY), investigated the effect of EMPA on CVD [59]. This trial assessed the benefits of utilizing EMPA in patients following acute myocardial infarction and found that EMPA significantly improved the echocardiographic functional and structural parameters. In addition, the EMMY clinical trial demonstrated that EMPA promoted a greater reduction in the N-terminal prohormone of brain natriuretic peptide (NT-proBNP), which is a heart failure marker. Finally, in order to ascertain the effect of EMPA on renal health, the Study of Heart and Kidney Protection with Empagliflozin (EMPA-KIDNEY) trial [60] was conducted, which showed a lower risk of progression of kidney disease or death from cardiovascular causes (13.1% vs. 16.9%) compared with placebo.

4.2.2. Dapagliflozin

The second SGLT2 inhibitor that has been found to be beneficial in treating cardiovascular and renal disease in people without diabetes is DAPA. In the Dapagliflozin and Prevention of Adverse Outcomes in Heart Failure (DAPA-HF) [54] and the Dapagliflozin Evaluation to Improve the Lives of Patients with Preserved Ejection Fraction Heart Failure (DELIVER) [61] clinical trials, patients on DAPA had a lowered risk of worsening heart failure (16.3% vs. 21.2% and 11.8% vs. 14.5%, respectively) or death from cardiovascular causes (9.6% vs. 11.5% and 7.4% vs. 8.3%, respectively) compared with placebo. To ascertain the effect DAPA has on renal health, the DAPA-CKD [57] and the Effects of the SGLT2 inhibitor Dapagliflozin on proteinuria in nondiabetic patients with Chronic Kidney Disease (DIAMOND) [55] trials were conducted. The DAPA-CKD trial showed a significant reduction in the risk of death from renal or cardiovascular causes (9.2% vs. 14.5%) compared with placebos. Meanwhile, the DIAMOND trial showed that while DAPA treatment did not affect proteinuria in patients with CKD, it reduced GFR levels compared with placebo. While reduced GFR is a sign of progressive kidney disease, there is a phenomenon that occurs with SGLT2i’s known as the estimated GFR (eGFR) acute dip, which is an acute reversible reduction in GFR. Of significance, the EMPA-REG [51], VERTIS-CV [53] and the CREDENCE [52] clinical trials have all confirmed that the dip in eGFR is not associated with progressive loss of long-term kidney function or acute kidney injury [79].

4.2.3. Ipragliflozin

Although not one of the main SGLT2i’s, Ipragliflozin may also offer clinical potential, as demonstrated by a recent case study [80]. In this study, an 83-year-old man with chronic heart failure and T2D was hospitalized four times over 5 years, but with the use of Ipragliflozin, he displayed reduced cardiac sympathetic nerve activity and was not hospitalized for 2 years afterwards. The improved health of the patient may be due to the reduction in observed cardiac sympathetic nerve hyperactivity, and this finding warrants further investigation of this inhibitor [80].

5. Discussion

5.1. Are Dual SGLT1/2 Inhibitors More Effective Than Sole SGLT2 Inhibitors?

One of the burning questions at present is whether utilizing a dual SGLT1/2 inhibitor like Sotagliflozin (SOTA) may be more beneficial than sole SGLT2i’s such as EMPA, CANA or DAPA when it comes to the treatment of cardiorenal disease in both patients with and without diabetes. As dual SGLT1/2 inhibition is a relatively new pharmacological therapy compared with sole SGLT2 inhibitors, the breadth of research findings is limited.
A study conducted on nondiabetic C57BL/6J mice found that treatment with SOTA attenuated cardiac hypertrophy and histological markers of cardiac fibrosis which were induced by the transverse aortic constriction (TAC) procedure [81].
Two clinical trials utilizing SOTA are the Effect of Sotagliflozin on Cardiovascular Events in Patients with Type 2 Diabetes Post Worsening Heart Failure (SOLOIST-WHF) trial [82] and the Effect of Sotagliflozin on Cardiovascular and Renal Events in Participants with Type 2 Diabetes and Renal Impairment Who Are At Cardiovascular Risk (SCORED) trial [83]. Both the SOLOIST-WHF trial and the SCORED trial showed that in patients with T2D and CKD, the rate of hospitalization and urgent care visits for heart failure was reduced when patients were administered SOTA compared with placebo. A new clinical trial, called the Sotagliflozin in Heart Failure with Preserved Ejection Fraction Patients (SOTA-P-CARDIA) [84], is currently underway to investigate the SOTA-mediated cardiovascular effects and mechanisms of action in patients with heart failure with preserved ejection fraction (HFpEF) but without diabetes.

5.2. Interesting Avenues for SGLT2i Therapy for the Treatment of T1D

To date, SGLT2i’s have not been clinically approved for the sole treatment of T1D due to the concerns surrounding hypoglycemia and ketoacidosis. However, it is a field of research that is gaining momentum, both in the context of preclinical and clinical pilot studies.

5.2.1. Animal Studies Utilizing SGLT2i’s as a Treatment for T1D

Our team has conducted a great deal of research utilizing our T1D Akimba mouse model. We have shown that SGLT2i’s may be a potential therapeutic for not just T2D but also T1D. When our Akimba mice were treated with the SGLT2i’s DAPA [85], CANA [86] and EMPA [86], metabolic parameters such as fasting blood glucose levels, polydipsia (excessive thirst) and weight management were all improved. Aside from the metabolic advantages in our T1D mice, we determined that DAPA and EMPA conferred beneficial effects on (i) digestive health [42] by significantly increasing the beneficial short-chain fatty acid butyric acid and (ii) diabetic retinopathy by reducing microvascular lesions [85,86,87] and reducing the pathogenic factor succinate [42].
Studies have shown that SGLT2i’s promote an upregulation of the family member SGLT1 in the kidneys (Figure 4) and also cause a reduction in kidney size and an improvement in renal histology [88]. The compensatory upregulation of SGLT1 with SGLT2i’s warrants the use of dual SGLT1/2 inhibitors such as SOTA.
While we have studied the effects of SGLT2i’s in our Akita and Akimba mice with regards to kidney health, our future studies aim to determine how these inhibitors may also improve cardiovascular health in our T1D Akita mice, as this strain is known to manifest diabetic cardiomyopathy with ageing [89].
Figure 4. SGLT2 inhibition results in increased luminal SGLT1 expression in renal tissue of diabetic Akimba mice: Representative SGLT1 immunohistochemistry images of renal tissue of mice treated with (A) vehicle or (B) Empagliflozin (EMPA) via drinking water (25 mg/kg/day) for 8 weeks. (C) Quantitation of SGLT1 intensity; n = 3–4 mice/group; mean + SEM. ** p = 0.005. Black arrow = luminal SGLT1 expression. Intensity: 0 = absent–3 = high intensity; FOV, field of view. Figure taken from [90].
Figure 4. SGLT2 inhibition results in increased luminal SGLT1 expression in renal tissue of diabetic Akimba mice: Representative SGLT1 immunohistochemistry images of renal tissue of mice treated with (A) vehicle or (B) Empagliflozin (EMPA) via drinking water (25 mg/kg/day) for 8 weeks. (C) Quantitation of SGLT1 intensity; n = 3–4 mice/group; mean + SEM. ** p = 0.005. Black arrow = luminal SGLT1 expression. Intensity: 0 = absent–3 = high intensity; FOV, field of view. Figure taken from [90].
Ijms 24 14243 g004

5.2.2. Human Pilot Studies/Clinical Trials Utilizing SGLT2i’s as an Add-On to Insulin for Patients with Type 1 Diabetes

Due to the difficulty in managing insulin in T1D patients, there are many clinical trials and pilot studies that have investigated the effect of utilizing the three main SGLT2 inhibitors, CANA [91], DAPA [92,93] or EMPA [94], as adjunctive therapies alongside insulin. While the inhibitors promoted ketoacidosis (KA), it was indicated that it was related to inadequately controlled insulin. Proper monitoring of glucose and ketone levels, as well as titration of the inhibitor/insulin, may be able to control the incidence of ketoacidosis. The Empagliflozin as Adjunctive to Insulin Therapy (EASE) trial determined that a dose of 2.5 mg/day of EMPA (phase 3) as opposed to a dose of 10 mg/day of EMPA (phase 2) helped to reduce the incidence of KA and, therefore, the lower dose may be a viable treatment option.
All clinical trials, including the Study of Effects of Canagliflozin as Add-on Therapy to Insulin in the Treatment of Participants With Type 1 Diabetes Mellitus (T1DM) [91] trial, the Dapagliflozin Evaluation in Patients With Inadequately Controlled Type 1 Diabetes (DEPICT-1) [92] and Efficacy and Safety of Dapagliflozin in Patients With Inadequately Controlled Type 1 Diabetes (DEPICT-2) [93] trials, as well as the EASE-2/3 [94] Empagliflozin trials, showed that SGLT2i’s promoted reductions in HbA1c, body weight and insulin requirements. Additionally, the aforementioned clinical trials most importantly improved glycemic control [95,96] after treatment, without producing any hypoglycemia.

5.3. Use of Dual SGLT 1/2 Inhibitors in T1D

As highlighted above, there are many benefits to utilizing SGLT2i’s for the treatment of T1D. This review highlighted the promising emerging studies that have been conducted to show that SOTA is beneficial for T2D [97]. A question that remains is whether the dual SGLT1/2 inhibitor, SOTA, may be beneficial for T1D and even surpass the benefits conferred by sole SGLT2 inhibitors. In our recent study utilizing our T1D Akimba mice [90], we concluded that SOTA not only significantly decreased fasting blood glucose levels but also promoted healthy weight gain compared with vehicle counterparts. Aside from this, SOTA also improved diabetes-associated polydipsia. As mentioned previously, overactivation of the SNS is strongly associated with diabetes as well as cardiorenal disease and, therefore, understanding the effects that SOTA has on SNS activation is of critical importance. We found that SOTA therapy resulted in a reduction in the main neurotransmitter of the SNS, NE. This phenomenon is otherwise known as sympathoinhibition.

6. Conclusions

We clearly demonstrated that SGLT2 inhibitors have enormous potential in improving renal and cardiovascular outcomes in patients with or without diabetes (Figure 5). Although most research has been conducted on T2D, the use of SGLT2i’s in the treatment of T1D has also been found to be beneficial and definitely warrants further research.

Author Contributions

Conceptualization, J.M. and V.M.; funding acquisition, L.H., V.M., M.P.S. and J.M.; writing—original draft, J.M. and V.M.; writing—review and editing, J.M., L.H., M.P.S. and V.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Diabetes Research WA.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Conflicts of Interest

Jennifer Matthews, Lakshini Y. Herat and Vance B. Matthews declare that they have no conflict of interest. Markus P. Schlaich reports grants from Boehringer Ingelheim while conducting the study.

References

  1. Kovesdy, C. Epidemiology of chronic kidney disease: An update 2022. Kidney Int. Suppl. 2022, 12, 7–11. [Google Scholar] [CrossRef] [PubMed]
  2. Keith, D.; Nichols, G.; Guillion, C.; Brown, J.; Smith, D. Longitudinal follow-up and outcomes among a population with chronic kidney disease in a large managed care organization. Arch. Intern. Med. 2004, 164, 659–663. [Google Scholar] [CrossRef] [PubMed]
  3. Aoki, J.; Nakajima, H.; Mori, M.; Sugimoto, T.; Hatori, M.; Tanimoto, S.; Amiya, E.; Hara, K. Clinical and pathologic characteristics of dilated cardiomyopathy in hemodialysis patients. Kidney Int. 2005, 67, 333–340. [Google Scholar] [CrossRef]
  4. Roth, G.; Mensah, G.; Johnson, C.; Addolorato, G.; Ammirati, E.; Baddour, L.; Barengo, N.; Beaton, A.; Benjamin, E.; Benziger, C.; et al. Global Burden of Cardiovascular Diseases and Risk Factors, 1990–2019. J. Am. Coll. Cardiol. 2020, 76, 2982–3021. [Google Scholar] [CrossRef]
  5. Cook, C.; Cole, G.; Asaria, P.; Jabbour, R.; Francis, D. The annual global economic burden of heart failure. Int. J. Cardiol. 2014, 171, 368–376. [Google Scholar] [CrossRef] [PubMed]
  6. GBD 2021 Diabetes Collaborators. Global, regional, and national burden of diabetes from 1990 to 2021, with projections of prevalence to 2050: A systematic analysis for the Global Burden of Disease Study 2021. Lancet 2023, 402, 203–234. [Google Scholar] [CrossRef] [PubMed]
  7. Collins, A.; Foley, R.; Gilbertson, D.; Chen, S. United States Renal Data System public health surveillance of chronic kidney disease and end-stage renal disease. Kidney Int. Suppl. 2015, 5, 2–7. [Google Scholar] [CrossRef]
  8. Reutens, A. Epidemiology of diabetic kidney disease. Med. Clin. N. Am. 2013, 97, 1–18. [Google Scholar] [CrossRef]
  9. Hussain, S.; Chand Jamali, M.; Habib, A.; Hussain, M.; Akhtar, M.; Najmi, A. Diabetic kidney disease: An overview of prevalence, risk factors, and biomarkers. Clin. Epidemiol. Glob. Health 2021, 9, 2–6. [Google Scholar] [CrossRef]
  10. Pavkov, M.; Bennett, P.; Knowler, W.; Krakoff, J.; Sievers, M.; Nelson, R. Effect of youth-onset type 2 diabetes mellitus on incidence of end-stage renal disease and mortality in young and middle-aged Pima Indians. JAMA 2006, 296, 421–426. [Google Scholar] [CrossRef]
  11. Dasgupta, I. Cost of treating diabetic kidney disease. Indian J. Nephrol. 2014, 24, 139–140. [Google Scholar] [CrossRef] [PubMed]
  12. Deng, Y.; Li, N.; Wu, Y.; Wang, M.; Yang, S.; Zheng, Y.; Deng, X.; Xiang, D.; Zhu, Y.; Xu, P.; et al. Global, Regional and National Burden of Diabetes-Related Chronic Kidney Disease from 1990 to 2019. Front. Endocrinol. 2021, 12, 672350. [Google Scholar] [CrossRef]
  13. Verma, D.; Firoz, A.; Garlapati, S.; Sathi, T.; Haris, M.; Dhungana, B.; Ray, B.; Shah, G.; Kc, B.; Paudel, P. Emerging treatments of Cardiorenal Syndrome: An update on Pathophysiology and Management. Cureus 2021, 13, e17240. [Google Scholar] [CrossRef] [PubMed]
  14. Sasso, F.; Pafundi, P.; Simeon, V.; De Nicola, L.; Chiodini, P.; Galiero, R.; Rinaldi, L.; Nevola, R.; Salvatore, T.; Sardu, C.; et al. Efficacy and durability of multifactorial intervention on mortality and MACEs: A randomized clinical trial in type-2 diabetic kidney disease. Cardiovasc. Diabetol. 2021, 20, 145. [Google Scholar] [CrossRef] [PubMed]
  15. Breyer, M.; Susztak, K. The next generation of therapeutics for chronic kidney disease. Nat. Rev. Drug Discov. 2016, 15, 568–588. [Google Scholar] [CrossRef]
  16. Sano, R.; Shinozaki, Y.; Ohta, T. Sodium-glucose cotransporters: Functional properties and pharmaceutical potential. J. Diabetes Investig. 2020, 11, 770–782. [Google Scholar] [CrossRef]
  17. Wright, E. The Intestinal Na+/Glucose Cotransporter. Annu. Rev. Physiol. 1993, 55, 575–589. [Google Scholar] [CrossRef] [PubMed]
  18. Suga, T.; Kikuchi, O.; Kobayashi, M.; Matsui, S.; Yokota-Hashimoto, H.; Wada, E.; Kohno, D.; Sasaki, T.; Takeuchi, K.; Kakizaki, S.; et al. SGLT1 in pancreatic a cells regulate glucagon secretion in mice, possibly explaining the distinct effects of SGLT2 inhibitors on plasma glucagon levels. Mol. Metab. 2019, 19, 1–12. [Google Scholar] [CrossRef]
  19. Vrhovac, I.; Eror, D.; Klessen, D.; Burger, C.; Breljak, D.; Kraus, O.; Radovic, N.; Jadrijevic, S.; Aleksic, I.; Walles, T.; et al. Localizations of Na+-d-glucose cotransporters SGLT1 and SGLT2 in human kidney and of SGLT1 in human small intestine, liver, lung and heart. Pflug. Arch. 2015, 467, 1881–1898. [Google Scholar] [CrossRef]
  20. Herat, L.Y.; Matthews, V.; Rakoczy, E.; Carnagarin, R.; Schlaich, M. Focusing on Sodium Glucose Co-transporter-2 (SGLT2) and its potential impact in diabetic retinopathy. Int. J. Endocrinol. 2018, 2018, 9254126. [Google Scholar] [CrossRef]
  21. Matthews, J.; Hibbs, M.; Herat, L.; Schlaich, M.; Matthews, V. The Sympathetic Nervous System Regulates Sodium Glucose Co-Transporter 1 Expression in the Kidney. Biomedicines 2023, 11, 819. [Google Scholar] [CrossRef] [PubMed]
  22. Herat, L.; Magno, A.; Rudnicka, C.; Hricova, J.; Carnagarin, R.; Ward, N.; Arcambal, A.; Kiuchi, M.; Head, G.; Schlaich, M.; et al. SGLT2 Inhibitor-Induced Sympathoinhibition: A Novel Mechanism for Cardiorenal Protection. JACC Basic Transl. Sci. 2020, 5, 169–179. [Google Scholar] [CrossRef]
  23. Bailey, C.; Day, C.; Bellary, S. Renal Protection with SGLT2 inhibitors: Effects in Acute and Chronic Kidney Disease. Curr. Diabetes Rep. 2022, 22, 39–52. [Google Scholar] [CrossRef]
  24. Chao, E.; Henry, R. SGLT2 Inhibition—A novel strategy for diabetes treatment. Nat. Rev. Drug Discov. 2010, 9, 551–559. [Google Scholar] [CrossRef]
  25. Zinman, B.; Wanner, C.; Lachin, J.; Fitchett, D.; Bluhmki, E.; Hantel, S.; Mattheus, M.; Devins, T.; Johanssen, E.; Woerle, H.; et al. Empagliflozin, Cardiovascular Outcomes, and Mortality in Type 2 Diabetes. N. Engl. J. Med. 2015, 373, 2117–2128. [Google Scholar] [CrossRef]
  26. Wiviott, S.D.; Raz, I.; Bonaca, M.; Mosenzon, O.; Kato, E.; Cahn, A.; Silverman, M.; Zelniker, T.; Kuder, J.; Murphy, S.; et al. Dapagliflozin and Cardiovascular Outcomes in Type 2 Diabetes. N. Engl. J. Med. 2019, 380, 347–357. [Google Scholar] [CrossRef]
  27. Neal, B.; Perkovic, V.; Mahaffey, K.; Zeeuw, D.; Fulcher, G.; Erondu, N.; Shaw, W.; Law, G.; Desai, M.; Matthews, D. Canagliflozin and Cardiovascular and Renal Events in Type 2 Diabetes. N. Engl. J. Med. 2017, 377, 644–657. [Google Scholar] [CrossRef]
  28. Xiang, B.; Zhao, X.; Zhou, X. Cardiovascular benefits of sodium glucose cotransporter 2 inhibitors in diabetic and nondiabetic patients. Cardiovasc. Diabetol. 2021, 20, 78. [Google Scholar] [CrossRef]
  29. Dutka, M.; Bobinski, R.; Ulman-Wlodarz, I.; Hajduga, M.; Bujok, J.; Pajak, C.; Cwiertnia, M. Sodium glucose cotransporter 2 inhibitors: Mechanisms of action in heart failure. Heart Fail. Rev. 2021, 26, 603–622. [Google Scholar] [CrossRef]
  30. Elsayed, N.; Aleppo, G.; Aroda, V.; Bannuru, R.; Brown, F.; Bruemmer, D.; Collins, B.; Hilliard, M.; Isaacs, D.; Johnson, E.; et al. Chronic Kidney Disease and Risk Management: Standards of Care in Diabetes—2023. Diabetes Care 2023, 46 (Suppl. S1), S191–S202. [Google Scholar] [CrossRef]
  31. Tahara, A.; Takasu, T.; Yokono, M.; Imamura, M.; Kurosaki, E. Characterization and comparison of sodium-glucose cotransporter 2 inhibitors in pharmacokinetics, pharmacodynamics, and pharmacologic effects. J. Pharmacol. Sci. 2016, 130, 159–169. [Google Scholar] [CrossRef]
  32. Tahara, A.; Takasu, T.; Yokono, M.; Imamura, M.; Kurosaki, E. Characterization and comparison of sodium-glucose cotransporter 2 inhibitors: Part 2. Antidiabetic effects in type 2 diabetic mice. J. Pharmacol. Sci. 2016, 131, 198–208. [Google Scholar] [CrossRef]
  33. Tahara, A.; Takasu, T.; Yokono, M.; Imamura, M.; Kurosaki, E. Characterization and comparison of sodium-glucose cotransporter 2 inhibitors: Part 3. Effects on diabetic complications in type 2 diabetic mice. Eur. J. Pharmacol. 2017, 809, 163–171. [Google Scholar] [CrossRef] [PubMed]
  34. Vallon, V.; Verma, S. Effects of SGLT2 Inhibitors on Kidney and Cardiovascular Function. Annu. Rev. Physiol. 2021, 83, 503–528. [Google Scholar] [CrossRef] [PubMed]
  35. Novikov, A.; Fu, Y.; Huang, W.; Freeman, B.; Patel, R.; van Ginkel, C.; Koepsell, H.; Busslinger, M.; Onishi, A.; Nespoux, J.; et al. SGLT2 inhibition and renal urate excretion: Role of luminal glucose, GLUT9, and URAT1. Am. J. Physiol. Ren. Physiol. 2019, 316, F173–F185. [Google Scholar] [CrossRef] [PubMed]
  36. Majewski, C.; Bakris, G. Blood Pressure Reduction: An Added Benefit of Sodium-Glucose Cotransporter 2 Inhibitors in Patients with Type 2 Diabetes. Diabetes Care 2015, 38, 429–430. [Google Scholar] [CrossRef] [PubMed]
  37. Tikkanen, I.; Narko, K.; Zeller, C.; Green, A.; Salsali, A.; Broedl, U.; Woerle, H. Empagliflozin reduces blood pressure in patients with type 2 diabetes and hypertension. Diabetes Care 2015, 38, 420–428. [Google Scholar] [CrossRef] [PubMed]
  38. Sjostrom, C.; Johansson, P.; Ptaszynska, A.; List, J.; Johnsson, E. Dapagliflozin lowers blood pressure in hypertensive and non-hypertensive patients with type 2 diabetes. Diabetes Vasc. Dis. Res. 2015, 12, 352–358. [Google Scholar] [CrossRef]
  39. Matthews, V.; Elliot, R.; Rudnicka, C.; Hricova, J.; Herat, L.; Schlaich, M. Role of the sympathetic nervous system in regulation of the sodium glucose cotransporter 2. J. Hypertens. 2017, 35, 2059–2068. [Google Scholar] [CrossRef]
  40. Wu, J.; Chen, Y.; Yang, H.; Gu, L.; Ni, Z.; Mou, S.; Shen, J.; Che, X. Sodium glucose co-transporter 2 (SGLT2) inhibition via dapagliflozin improves diabetic kidney disease (DKD) over time associated with increasing effect on the gut microbiota in db/db mice. Front. Endocrinol. 2023, 14, 1026040. [Google Scholar] [CrossRef] [PubMed]
  41. Lee, D.; Battson, M.; Jarrell, D.; Hou, S.; Ecton, K.; Weir, T.; Gentile, C. SGLT2 inhibition via dapagliflozin improves generalized vascular dysfunction and alters the gut microbiota in type 2 diabetic mice. Cardiovasc. Diabetol. 2018, 17, 62. [Google Scholar] [CrossRef] [PubMed]
  42. Herat, L.; Ward, N.; Magno, A.; Rakoczy, E.; Kiuchi, M.; Schlaich, M.; Matthews, V. Sodium glucose co-transporter 2 inhibition reduces succinate levels in diabetic mice. World J. Gastroenterol. 2020, 26, 3225–3235. [Google Scholar] [CrossRef] [PubMed]
  43. Deng, L.; Yang, Y.; Xu, G. Empagliflozin ameliorates type 2 diabetes mellitus-related diabetic nephrophathy via altering the gut microbiota. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2022, 1867, 159234. [Google Scholar] [CrossRef]
  44. Hata, S.; Okamura, T.; Kobayashi, A.; Bamba, R.; Miyoshi, T.; Nakajima, H.; Kitagawa, N.; Hashimoto, Y.; Majima, S.; Senmaru, T.; et al. Gut Microbiota Changes by an SGLT2 Inhibitor, Luseogliflozin, Alters Metabolites Compared with Those in a Low Carbohydrate Diet in db/db Mice. Nutrients 2022, 14, 3531. [Google Scholar] [CrossRef]
  45. Gong, Q.; Zhang, R.; Wei, F.; Fang, J.; Zhang, J.; Sun, J.; Sun, Q.; Wang, H. SGLT2 inhibitor-empagliflozin treatment ameliorates diabetic retinopathy manifestations and exerts protective effects associated with augmenting branched chain amino acids catabolism and transportation in db/db mice. Biomed. Pharmacother. 2022, 152, 113222. [Google Scholar] [CrossRef] [PubMed]
  46. Luo, Q.; Leley, S.; Bello, E.; Dhami, H.; Mathew, D.; Bhatwadekar, A. Dapagliflozin protects neural and vascular dysfunciton of the retinas in diabetes. BMJ Open Diabetes Res. Care 2022, 10, e002801. [Google Scholar] [CrossRef]
  47. Tang, L.; Wu, Y.; Tian, M.; Sjostrom, C.; Johansson, U.; Peng, X.; Smith, D.; Huang, Y. Dapagliflozin slows the progression of the renal and liver fibrosis associated with type 2 diabetes. Am. J. Physiol. Endocrinol. Metab. 2017, 313, E563–E576. [Google Scholar] [CrossRef] [PubMed]
  48. Shin, S.; Chung, S.; Kim, S.; Lee, E.; Yoo, Y.; Kim, J.; Ahn, Y.; Kim, E.; Moon, S.; Kim, M.; et al. Effect of Sodium-Glucose Co-Transporter 2 Inhibitor, Dapagliflozin, on Renal Renin-Angiotensin System in an Animal Model of Type 2 Diabetes. PLoS ONE 2016, 11, e0165703. [Google Scholar] [CrossRef] [PubMed]
  49. Kojima, N.; Williams, J.; Takahashi, T.; Miyata, N.; Roman, R. Effects of a new SGLT2 inhibitor, Luseogliflozin, on diabetic nephropathy in T2D rats. J. Pharmacol. Exp. Ther. 2013, 345, 464–472. [Google Scholar] [CrossRef] [PubMed]
  50. Lin, B.; Koibuchi, N.; Hasegawa, Y.; Sueta, D.; Toyama, K.; Uekawa, K.; Ma, M.; Nakagawa, T.; Husaka, H.; Kim-Mitsuyama, S. Glycemic control with empagliflozin, a novel selective SGLT2 inhibitor, ameliorates cardiovascular injury and cognitive dysfunction in obese and type 2 diabetic mice. Cardiovasc. Diabetol. 2014, 13, 148. [Google Scholar] [CrossRef] [PubMed]
  51. Wanner, C.; Inzucchi, S.; Lachin, J.; Fitchett, D.; Von Eynatten, M.; Mattheus, M.; Johansen, E.; Woerle, H.; Broedl, U.; Zinman, B. Empagliflozin and Progression of Kidney Disease in Type 2 Diabetes. N. Engl. J. Med. 2016, 375, 323–334. [Google Scholar] [CrossRef] [PubMed]
  52. Perkovic, V.; Jardine, M.; Neal, B.; Bompoint, S.; Heerspink, H.; Charytan, D.; Edwards, R.; Agarwal, R.; Bakris, G.; Bull, S.; et al. Canagliflozin and Renal Outcomes in Type 2 Diabetes and Nephropathy. N. Engl. J. Med. 2019, 380, 2295–2306. [Google Scholar] [CrossRef] [PubMed]
  53. Cannon, C.; Pratley, R.; Dagogo-Jack, S.; Mancuso, J.; Huyck, S.; Masiukiewicz, U.; Charbonnel, B.; Frederich, R.; Gallo, S.; Cosentino, F.; et al. Cardiovascular Outcomes with Ertugliflozin in Type 2 Diabetes. N. Engl. J. Med. 2020, 383, 1425–1435. [Google Scholar] [CrossRef]
  54. McMurray, J.; Solomon, S.; Inzucchi, S.; Kober, L.; Kosiborod, M.; Martinez, F.; Ponikowski, P.; Sabatine, M.; Anand, I.; Belohlavek, J.; et al. Dapagliflozin in Patients with Heart Failure and Reduced Ejection Fraction. N. Engl. J. Med. 2019, 381, 1995–2008. [Google Scholar] [CrossRef]
  55. Cherney, D.; Dekkers, C.; Barbour, S.; Cattran, D.; Abdul Gafor, A.H.; Greasley, P.; Laverman, G.; Kun Lim, S.; Di Tanna, G.; Reich, H.; et al. Effects of the SGLT2 inhibitor dapagliflozin on proteinuria in non-diabetic patients with chronic kidney disease (DIAMOND): A randomized, double-blind, crossover trial. Lancet Diabetes Endocrinol. 2020, 8, 582–593. [Google Scholar] [CrossRef] [PubMed]
  56. Packer, M.; Anker, S.; Butler, J.; Filippatos, G.; Pocock, S.; Carson, P.; Januzzi, J.; Subodh, V.; Tsutsui, H.; Brueckmann, M.; et al. Cardiovascular and Renal Outcomes with Empagliflozin in Heart Failure. N. Engl. J. Med. 2020, 383, 1413–1424. [Google Scholar] [CrossRef]
  57. Heerspink, H.; Stefansson, B.; Correa-Rotter, R.; Chertow, G.; Greene, T.; Hou, F.; Mann, J.; McMurray, J.; Lindberg, M.; Rossing, P.; et al. Dapagliflozin in Patients with Chronic Kidney Disease. N. Engl. J. Med. 2020, 383, 1436–1446. [Google Scholar] [CrossRef] [PubMed]
  58. Packer, M.; Butler, J.; Zannad, F.; Filippatos, G.; Ferreira, J.; Pocock, S.; Carson, P.; Anand, I.; Doehner, W.; Haass, M.; et al. Effect of Empagliflozin on Worsening Heart Failure Events in Patients With Heart Failure and Preserved Ejection Fraction: EMPEROR-Preserved Trial. Circulation 2021, 144, 1284–1294. [Google Scholar] [CrossRef] [PubMed]
  59. Lewinski, D.; Kolesnik, E.; Tripolt, N.; Pferschy, P.; Benedikt, M.; Wallner, M.; Alber, H.; Berger, R.; Lichtnauer, M.; Saely, C.; et al. Empagliflozin in acute myocardial infarction: The EMMY trial. Eur. Heart J. 2022, 43, 4421–4432. [Google Scholar] [CrossRef] [PubMed]
  60. Herrington, W.; Staplin, N.; Wanner, C.; Green, J.; Hauske, S.; Emberson, J.; Preiss, D.; Judge, P.; Mayne, K.; Ng, S.; et al. Empagliflozin in Patients with Chronic Kidney Disease. N. Engl. J. Med. 2023, 388, 117–127. [Google Scholar] [CrossRef]
  61. Solomon, S.; McMurray, J.; Claggett, B.; Boer, R.; DeMets, D.; Hernandez, A.; Inzucchi, S.; Kosiborod, M.; Lam, C.; Martinez, F.; et al. Dapagliflozin in Heart Failure with Mildly Reduced or Preserved Ejection Fraction. N. Engl. J. Med. 2022, 387, 1089–1098. [Google Scholar] [CrossRef] [PubMed]
  62. Herat, L.; Matthews, J.; Azzam, O.; Schlaich, M.; Matthews, V. Targeting features of the metabolic syndrome through sympatholytic effects of SGLT inhibition. Curr. Hypertens. Rep. 2022, 24, 67–74. [Google Scholar] [CrossRef] [PubMed]
  63. Liu, Y.; Wu, M.; Xu, B.; Kang, L. Empagliflozin Alleviates Atherosclerosis Progression by Inhibiting Inflammation and Sympathetic Activity in a Normoglycemic Mouse Model. J. Inflamm. Res. 2021, 14, 2277–2287. [Google Scholar] [CrossRef] [PubMed]
  64. Matthews, J.; Herat, L.; Magno, A.; Gorman, S.; Schlaich, M.; Matthews, V. SGLT-2 Inhibitor-Induced Sympathoexcitation in White Adipose Tissue: A Novel Mechanism of Beiging. Biomedicines 2020, 8, 514. [Google Scholar] [CrossRef]
  65. Machado, S.; Pasquarelli-do-Nascimento, G.; Santos da Silva, D.; Farias, G.; de Oliveira Santos, I.; Baptista, L.; Magalhaes, K. Browning of the white adipose tissue regulation: New insights into nutritional and metabolic relevance in health and diseases. Nutr. Metab. 2022, 19, 61. [Google Scholar] [CrossRef] [PubMed]
  66. Leng, W.; Ouyang, X.; Lei, X.; Wu, M.; Chen, L.; Wu, Q.; Deng, W.; Liang, Z. The SGLT-2 Inhibitor Dapagliflozin Has a Therapeutic Effect on Atherosclerosis in Diabetic ApoE−/− Mice. Mediat. Inflamm. 2016, 2016, 6305735. [Google Scholar] [CrossRef] [PubMed]
  67. Papazafiropoulou, A.; Georgopoulos, M.; Katsilambros, N. Ketone bodies and the heart. Arch. Med. Sci. Atheroscler. Dis. 2021, 6, e209–e214. [Google Scholar] [CrossRef] [PubMed]
  68. Qiu, Y.; Hu, X.; Xu, C.; Lu, C.; Cao, R.; Xie, Y.; Yang, J. Ketogenic diet alleviates renal fibrosis in mice by enhancing fatty acid oxidation through the free fatty acid receptor 3 pathway. Front. Nutr. 2023, 10, 1127845. [Google Scholar] [CrossRef] [PubMed]
  69. Pietschner, R.; Kolwelter, J.; Bosch, A.; Striepe, K.; Jung, S.; Kannenkeril, D.; Ott, C.; Schiffer, M.; Achenbach, S.; Schmieder, R. Effect of empagliflozin on ketone bodies in patients with chronic heart failure. Cardiovasc. Diabetol. 2021, 20, 219. [Google Scholar] [CrossRef]
  70. Park, S.; Farooq, M.; Gaertner, S.; Bruckert, C.; Qureshi, A.; Lee, H.; Benrahla, D.; Pollet, B.; Stephan, D.; Ohlmann, P.; et al. Empaglifozin improved systolic blood pressure, endothelial dysfunction and heart remodeling in the metabolic syndrome ZSF1 rat. Cardiovasc. Diabetol. 2020, 19, 19. [Google Scholar] [CrossRef]
  71. Tomita, I.; Kume, S.; Sugahara, S.; Osawa, N.; Yamahara, K.; Yasuda-Yamahara, M.; Takeda, N.; Chin-Kanasaki, M.; Kaneko, T.; Mayoux, E.; et al. SGLT2 inhibition mediates protection from diabetic kidney disease by promoting ketone body-induced mTORC1 inhibition. Cell Metab. 2020, 32, 404–419.e6. [Google Scholar] [CrossRef] [PubMed]
  72. Khunti, K.; Ruan, Y.; Davies, J.; Field, B.; Harris, S.; Kosiborod, M.; Nagi, D.; Narendran, P.; Patel, D.; Ryder, R.; et al. Association between SGLT2 Inhibitor Treatment and Diabetic Ketoacidosis and Mortality in People with Type 2 Diabetes Admitted to Hospital with COVID-19. Diabetes Care 2022, 45, 2838–2843. [Google Scholar] [CrossRef] [PubMed]
  73. Kimura, I.; Inoue, D.; Maeda, T.; Hara, T.; Ichimura, A.; Miyauchi, S.; Kobayashi, M.; Hirasawa, A.; Tsujimoto, G. Short-chain fatty acids and ketones directly regulate sympathetic nervous system via G protein-coupled receptor 41 (GPR41). Proc. Natl. Acad. Sci. USA 2011, 108, 8030–8035. [Google Scholar] [CrossRef] [PubMed]
  74. Kolanowski, J.; Young, J.; Landsberg, L. Stimulatory influence of D(-)3-hydroxybutyrate feeding on sympathetic nervous system activity in the rat. Metabolism 1994, 43, 180–185. [Google Scholar] [CrossRef] [PubMed]
  75. Lin, K.; Yang, N.; Luo, W.; Qian, J.; Zhu, W.; Ye, S.; Yuan, C.; Xu, D.; Liang, G.; Huang, W.; et al. Direct cardio-protection of Dapagliflozin against obesity-related cardiomyopathy via NHE1/MAPK signaling. Acta Pharmacol. Sin. 2022, 43, 2624–2635. [Google Scholar] [CrossRef]
  76. Xu, L.; Nagata, N.; Nagashimada, M.; Zhuge, F.; Ni, Y.; Chen, G.; Mayoux, E.; Kaneko, S.; Ota, T. 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, 137–149. [Google Scholar] [CrossRef] [PubMed]
  77. Radlinger, B.; Ress, C.; Folie, S.; Salzmann, K.; Lechuga, A.; Weiss, B.; Salvenmoser, W.; Graber, M.; Hirsch, J.; Holfeld, J.; et al. Empagliflozin protects mice against diet-induced obesity, insulin resistance and hepatic steatosis. Diabetologia 2023, 66, 754–767. [Google Scholar] [CrossRef]
  78. Ji, W.; Zhao, M.; Wang, M.; Yan, W.; Liu, Y.; Ren, S.; Lu, J.; Wang, B.; Chen, L. Effects of canagliflozin on weight loss in high-fat-diet-induced obese mice. PLoS ONE 2017, 12, e0179960. [Google Scholar] [CrossRef] [PubMed]
  79. Heerspink, H.; Cherney, D. Clinical Implications of an Acute Dip in eGFR after SGLT2 Inhibitor Initiation. Clin. J. Am. Soc. Nephrol. 2021, 16, 1278–1280. [Google Scholar] [CrossRef] [PubMed]
  80. Kiuchi, S.; Hisatake, S.; Kabuki, T.; Fujii, T.; Oka, T.; Dobashi, S.; Hashimoto, H.; Ikeda, T. Long-term use of ipragliflozin improved cardiac sympathetic nerve activity in a patient with heart failure: A case report. Drug Discov. Ther. 2018, 12, 51–54. [Google Scholar] [CrossRef] [PubMed]
  81. Young, S.; Ryan, L.; Mullins, T.; Flint, M.; Steane, S.; Walton, S.; Bielefeldt-Ohmann, H.; Carter, D.; Reichelt, M.; Gallo, L. Sotagliflozin, a dual SGLT1/2 Inhibitor, improves cardiac outcomes in a mouse model of early heart failure without diabetes. Front. Physiol. 2021, 12, 738594. [Google Scholar] [CrossRef] [PubMed]
  82. Bhatt, D.; Szarek, M.; Steg, P.; Cannon, C.; Leiter, L.; McGuire, D.; Lewis, J.; Riddle, M.; Voors, A.; Metra, M.; et al. Sotagliflozin in Patients with Diabetes and Recent Worsening Heart Failure. N. Engl. J. Med. 2021, 384, 117–128. [Google Scholar] [CrossRef]
  83. Bhatt, D.; Szarek, M.; Pitt, B.; Cannon, C.; Leiter, L.; McGuire, D.; Lewis, J.; Riddle, M.; Inzucchi, S.; Kosiborod, M.; et al. Sotagliflozin in Patients with Diabetes and Chronic Kidney Disease. N. Engl. J. Med. 2021, 384, 129–139. [Google Scholar] [CrossRef] [PubMed]
  84. Perez, M.; Rodriguez-Capitan, J.; Requena-Ibanez, J.; Santos-Gallego, C.; Zafar, M.; Escolar, G.; Mancini, D.; Mitter, S.; Lam, D.; Contreras, J.; et al. Rationale and Design of the SOTA-P-CARDIA Trial (ATRU-V): Sotagliflozin in HFpEF Patients Without Diabetes. Cardiovasc. Drugs Ther. online ahead of print. 2023. [Google Scholar] [CrossRef] [PubMed]
  85. Herat, L.; Matthews, J.; Ong, W.; Rakoczy, E.; Schlaich, M.; Matthews, V. Determining the Role of SGLT2 Inhibition with Dapagliflozin in the Development of Diabetic Retinopathy. Front. Biosci. 2022, 27, 320. [Google Scholar] [CrossRef] [PubMed]
  86. Herat, L.; Matthews, J.; Rakoczy, E.; Schlaich, M.; Matthews, V. Comparing and Contrasting the Effects of the SGLT Inhibitors Canagliflozin and Empagliflozin on the Progression of Retinopathy. Front. Biosci. 2023, 28, 83. [Google Scholar] [CrossRef] [PubMed]
  87. Matthews, J.; Herat, L.; Rooney, J.; Rakoczy, E.; Schlaich, M.; Matthews, V. Determining the Role of SGLT2 inhibition with Empagliflozin in the development of diabetic retinopathy. Biosci. Rep. 2022, 42, BSR20212209. [Google Scholar] [CrossRef] [PubMed]
  88. Matthews, J.; Schlaich, M.; Rakoczy, E.; Matthews, V.; Herat, L. The Effect of SGLT2 Inhibition on Diabetic Kidney Disease in a Model of Diabetic Retinopathy. Biomedicines 2022, 10, 522. [Google Scholar] [CrossRef] [PubMed]
  89. Kesherwani, V.; Shahshahan, H.; Mishra, P. Cardiac transcriptome profiling of diabetic Akita mice using microarray and next generation sequencing. PLoS ONE 2017, 12, e0182828. [Google Scholar] [CrossRef] [PubMed]
  90. Herat, L.; Matthews, J.; Hibbs, M.; Rakoczy, E.; Schlaich, M.; Matthews, V. SGLT1/2 inhibition improves glycemic control and multi-organ protection in type 1 diabetes. iScience 2023, 26, 107260. [Google Scholar] [CrossRef]
  91. Henry, R.; Thakkar, P.; Tong, C.; Polidori, D.; Alba, M. Efficacy and Safety of Canagliflozin, a Sodium-Glucose Cotransporter 2 Inhibitor, as Add-on to Insulin in Patients with Type 1 Diabetes. Diabetes Care 2015, 38, 2258–2265. [Google Scholar] [CrossRef] [PubMed]
  92. Dandona, P.; Mathieu, C.; Phillip, M.; Hansen, L.; Griffen, S.; Tschope, D.; Thoren, F.; Xu, J.; Langkilde, A. Efficacy and safety of dapagliflozin in patients with inadequately controlled type 1 diabetes (DEPICT-1): 24 week results from a multicentre, double-blind, phase 3, randomised controlled trial. Lancet 2017, 5, 864–876. [Google Scholar] [CrossRef] [PubMed]
  93. Mathieu, C.; Rudofsky, G.; Phillip, M.; Araki, E.; Lind, M.; Arya, N.; Thoren, F.; Scheerer, M.; Iqbal, N.; Dandona, P. Long-term efficacy and safety of dapagliflozin in patients with inadequately controlled type 1 diabetes (the DEPICT-2 study): 52-week results from a randomized controlled trial. Diabetes Obes. Metab. 2020, 22, 1516–1526. [Google Scholar] [CrossRef] [PubMed]
  94. Rosenstock, J.; Marquard, J.; Laffel, L.; Neubacher, D.; Kaspers, S.; Cherney, D.; Zinman, B.; Skyler, J.; George, J.; Soleymanlou, N.; et al. Empagliflozin as Adjunctive to Insulin Therapy in Type 1 Diabetes: The EASE Trials. Diabetes Care 2018, 41, 2560–2569. [Google Scholar] [CrossRef] [PubMed]
  95. Paik, J.; Blair, H. Dapagliflozin: A Review in Type 1 Diabetes. Drugs 2019, 79, 1877–1884. [Google Scholar] [CrossRef] [PubMed]
  96. Zinman, B.; Lachin, J.; Inzucchi, S. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N. Engl. J. Med. 2016, 374, 1094. [Google Scholar] [CrossRef] [PubMed]
  97. Cefalo, C.; Cinti, F.; Moffa, S.; Impronta, F.; Sorice, G.; Mezza, T.; Pontecorvi, A.; Giaccari, A. Sotagliflozin, the first dual SGLT inhibitor: Current outlook and perspectives. Cardiovasc. Diabetol. 2019, 18, 20. [Google Scholar] [CrossRef] [PubMed]
Figure 1. SGLT1 and SGLT2 sodium and glucose transport in intestinal lumen (SGLT1) and proximal tubule lumen in the kidneys (SGLT2).
Figure 1. SGLT1 and SGLT2 sodium and glucose transport in intestinal lumen (SGLT1) and proximal tubule lumen in the kidneys (SGLT2).
Ijms 24 14243 g001
Figure 2. Traditional and nontraditional risk factors for atherosclerosis. Red arrow indicates an increase in cardiovascular morbidity and mortality.
Figure 2. Traditional and nontraditional risk factors for atherosclerosis. Red arrow indicates an increase in cardiovascular morbidity and mortality.
Ijms 24 14243 g002
Figure 3. SGLT2 inhibition with DAPA treatment prevents the increase in blood pressure in neurogenically hypertensive mice: Effects of DAPA treatment on (A) systolic blood pressure, (B) diastolic blood pressure and (C) mean arterial blood pressure were measured using a tail-cuff apparatus. n = 10–12 mice/group; * p = 0.006; ** p = 0.0003; *** p = 0.0008; All data represented as mean ± SEM. Figure taken from [22].
Figure 3. SGLT2 inhibition with DAPA treatment prevents the increase in blood pressure in neurogenically hypertensive mice: Effects of DAPA treatment on (A) systolic blood pressure, (B) diastolic blood pressure and (C) mean arterial blood pressure were measured using a tail-cuff apparatus. n = 10–12 mice/group; * p = 0.006; ** p = 0.0003; *** p = 0.0008; All data represented as mean ± SEM. Figure taken from [22].
Ijms 24 14243 g003
Figure 5. Cardiorenal benefits of Sodium–Glucose Cotransporter 2 (SGLT2) inhibitors and proposed effects on SGLT1 activity: Sodium–Glucose Cotransporter 2, SGLT2; Sodium–Glucose Cotransporter 1, SGLT1; Empagliflozin, EMPA; Dapagliflozin, DAPA; Canagliflozin, CANA; CV, cardiovascular; HF, heart failure and MI, myocardial infarction. SGLT1/2 Inhibition: [83,97], SGLT1 Activity: [85,90], Cardio-Renal Protection: [25,27,51,54,96]. Red arrows indicate an increase in the parameter. Green arrows indicate a decrease in the parameter. Blue arrows indicate the flow of the figure.
Figure 5. Cardiorenal benefits of Sodium–Glucose Cotransporter 2 (SGLT2) inhibitors and proposed effects on SGLT1 activity: Sodium–Glucose Cotransporter 2, SGLT2; Sodium–Glucose Cotransporter 1, SGLT1; Empagliflozin, EMPA; Dapagliflozin, DAPA; Canagliflozin, CANA; CV, cardiovascular; HF, heart failure and MI, myocardial infarction. SGLT1/2 Inhibition: [83,97], SGLT1 Activity: [85,90], Cardio-Renal Protection: [25,27,51,54,96]. Red arrows indicate an increase in the parameter. Green arrows indicate a decrease in the parameter. Blue arrows indicate the flow of the figure.
Ijms 24 14243 g005
Table 3. Clinical trials in patients irrespective of diabetic status.
Table 3. Clinical trials in patients irrespective of diabetic status.
Clinical Trials in Patients with Diabetes Only
Trial/YearSGLT2 InhibitorPatient CohortPatient #OutcomeRef #
2015
EMPA-REG
EmpagliflozinT2D—High Risk of Cardiovascular Events7064↓ Mortality & Hospitalisation due to HF
↓ Risk of Clinically Relevant Renal Events
[25,51]
2017
CANVAS
CanagliflozinT2D—High Risk of CVD10,143↓ Major Adverse Cardiovascular Events
↓ Albuminuria Levels
↓ Renal Replacement Therapy/Death
[27]
2018
DELCARE-TIMI
DapagliflozinT2D—High Risk of Atherosclerotic CVD17,190↓ Cardiovascular Death or Hospitalisation for HF.[26]
2018
CREDENCE
CanagliflozinT2D and Kidney Disease4401↓ Risk of Kidney Failure/Cardiovascular Events[52]
2019
VERTIS
ErtugliflozinT2D and CVD8246↓ First/Total Hospitalization
↓ Risk of death from HF/CVD
[53]
Clinical Trials in Patients Irrespective of Diabetic Status
Trial/YearSGLT2 InhibitorPatient CohortPatient #OutcomeRef #
2019
DAPA-HF
DapagliflozinClass II, III or IV Heart Failure & EF < 40%4744↓ Worsening Heart Failure/Death from CV Events[54]
2019
DIAMOND
DapagliflozinNon Diabetic Patients with CKD50Induced an acute and reversible decline in mGFR levels.[55]
2020
EMPEROR-REDUCED
EmpagliflozinClass II, III or IV Heart Failure & EF < 40%3730↓ Mortality & Hospitalisation due to HF[56]
2020
DAPA-CKD
DapagliflozinHigh Risk of Kidney & CDV Outcomes4304↓ Risk of a declining GFR level
↓ End Stage Renal Disease/Death from Renal Causes
[57]
2021
EMPEROR-PRESERVED
EmpagliflozinClass II, III or IV Heart Failure & EF > 40%5988↓ Cardiovascular Death and Hospitalisation for HF.[58]
2022
EMMY
EmpagliflozinRecent acute myocardial infarction476↓ Risk of a declining GFR level
↓ ES Renal Disease/Death from Renal Causes
[59]
2022
EMPA-KIDNEY
EmpagliflozinPatients with CKD6609↓ Progression of Kidney Disease
↓ Death from Cardiovascular Causes
[60]
2022
DELIVER
DapagliflozinHeart Failure, Left Ventricular EF > 40%6263↓ Worsening Heart Failure/Death from Cardiovascular Events[61]
Arrows represent decreases of parameters in the table. # represents the number of patients.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Matthews, J.; Herat, L.; Schlaich, M.P.; Matthews, V. The Impact of SGLT2 Inhibitors in the Heart and Kidneys Regardless of Diabetes Status. Int. J. Mol. Sci. 2023, 24, 14243. https://doi.org/10.3390/ijms241814243

AMA Style

Matthews J, Herat L, Schlaich MP, Matthews V. The Impact of SGLT2 Inhibitors in the Heart and Kidneys Regardless of Diabetes Status. International Journal of Molecular Sciences. 2023; 24(18):14243. https://doi.org/10.3390/ijms241814243

Chicago/Turabian Style

Matthews, Jennifer, Lakshini Herat, Markus P. Schlaich, and Vance Matthews. 2023. "The Impact of SGLT2 Inhibitors in the Heart and Kidneys Regardless of Diabetes Status" International Journal of Molecular Sciences 24, no. 18: 14243. https://doi.org/10.3390/ijms241814243

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop