Skip to main content
Research Article
Originally Published 15 June 2022
Free Access

SGLT2 Inhibition via Empagliflozin Improves Endothelial Function and Reduces Mitochondrial Oxidative Stress: Insights From Frail Hypertensive and Diabetic Patients

Abstract

Background:

Frailty is a multidimensional condition often diagnosed in older adults with hypertension and diabetes, and both these conditions are associated with endothelial dysfunction and oxidative stress. We investigated the functional role of the SGLT2 (sodium glucose cotransporter 2) inhibitor empagliflozin in frail diabetic and hypertensive older adults.

Methods:

We studied the effects of empagliflozin in consecutive hypertensive and diabetic older patients with frailty presenting at the ASL (local health unit of the Italian Ministry of Health) of Avellino, Italy, from March 2021 to January 2022. Moreover, we performed in vitro experiments in human endothelial cells to measure cell viability, permeability, mitochondrial Ca2+, and oxidative stress.

Results:

We evaluated 407 patients; 325 frail elders with diabetes successfully completed the study. We propensity-score matched 75 patients treated with empagliflozin and 75 with no empagliflozin. We observed a correlation between glycemia and Montreal Cognitive Assessment (MoCA) score and between glycemia and 5-meter gait speed (5mGS). At 3-month follow-up, we detected a significant improvement in the MoCA score and in the 5mGS in patients receiving empagliflozin compared with non-treated subjects. Mechanistically, we demonstrate that empagliflozin significantly reduces mitochondrial Ca2+ overload and reactive oxygen species production triggered by high glucose in human endothelial cells, attenuates cellular permeability, and improves cell viability in response to oxidative stress.

Conclusions:

Taken together, our data indicate that empagliflozin reduces frailty in diabetic and hypertensive patients, most likely by decreasing the mitochondrial generation of reactive oxygen species in endothelial cells.

Graphical Abstract

Novelty and Relevance

What Is New?

The SGLT2 (sodium glucose cotransporter 2) inhibitor empagliflozin improves both cognitive and physical impairment in frail patients with hypertension and diabetes.
Empagliflozin ameliorates endothelial dysfunction induced by high glucose.
Empagliflozin reduces mitochondrial Ca2+ overload and oxidative stress in human endothelial cells.

What Is Relevant?

SGLT2 inhibitors have been shown to have beneficial effects on the cardiovascular system, but the underlying mechanisms were not fully understood.
This work indicates that SGLT2 inhibitors have favorable effects on different types of human endothelial cells, i.e. umbilical vein and brain microvascular endothelial cells.

Clinical/Pathophysiological Implications

The management of frailty in older adults should consider the expected benefits given by the treatment with SGLT2 inhibitors especially taking into account the pathophysiological rationale provided by this study, in terms of improvement of endothelial function and reduced oxidative stress.
Frailty is a systemic condition that leads to functional decline with physical and cognitive impairment.1–3 Frailty is often diagnosed in older adults with hypertension and diabetes, and both these conditions are associated with endothelial dysfunction and oxidative stress.4–6
Our group has recently evidenced the detrimental effects of hyperglycemia in frail hypertensive elders.7 Similarly, several investigators have shown that hyperglycemia drives inflammation and oxidative stress leading to endothelial dysfunction, with a negative impact that is especially evident in frail patients.8–11
In this scenario, SGLT2 (sodium glucose cotransporter 2) inhibitors, which include the Food and Drug Administration–approved drugs canagliflozin, dapagliflozin, empagliflozin, and ertugliflozin,12 are considered among the most promising oral antidiabetic drugs to reach and maintain an optimal glycemic control in older subjects.13 The main mechanism of action of these drugs is the blockage of SGLT2 proteins in the renal proximal convoluted tubules to reduce the reabsorption of filtered glucose and decrease the renal threshold for glucose, thereby promoting urinary glucose excretion.14–17 Empagliflozin is a SGLT2 inhibitor that has been shown to reduce mortality and rehospitalization for heart failure in diabetic patients.18–23 Additional potential benefits of empagliflozin and other SGLT2 inhibitors include improved cardiovascular energetics, reduced vascular tone and blood pressure, decreased renal dysfunction, increased circulating levels of ketone bodies, protection against pulmonary ischemia/reperfusion injury, and overall reduced systemic inflammation.24–37
On these grounds, we investigated the effects of empagliflozin on frailty in diabetic and hypertensive older adults. To mechanistically confirm our results, we evaluated the effects of empagliflozin on mitochondrial oxidative stress and permeabilization in human endothelial cells.

Methods

Data Availability

The data that support the findings of this study are available from the first author upon reasonable request.

Study Participants

We designed a prospective study to enroll consecutive hypertensive and diabetic older adults presenting at the ASL (local health unit of the Ministry of Health) of Avellino, Italy, from March 2021 to January 2022 with a diagnosis of frailty. Inclusion criteria: age >65 years, a diagnosis of frailty, presence of diabetes and primary hypertension, Montreal Cognitive Assessment (MoCA) score <26, and glomerular filtration rate >30. Exclusion criteria: age <65 years and history of previous stroke. The study was approved by the local ethical committee (Campania Nord). All subjects or their legal representatives signed an informed consent. The study had been registered at https://www.clinicaltrials.gov (unique identifier: NCT04962841).

Clinical Evaluation and Assessment of Cognitive and Physical Impairment

Blood glucose, glycated hemoglobin, and creatinine were measured in all patients. Clinical assessment was completed at baseline and after 3 months. Diabetes was defined according to the American Diabetes Association guidelines.38 Hypertension was defined as systolic blood pressure ≥140 mm Hg or diastolic blood pressure ≥90 mm Hg on repeated measurements or use of antihypertensive medications.39

Frailty Assessment

A diagnosis of physical frailty was made with at least 3 of the 5 Fried criteria,40 as we described previously41,42:
Weight loss (unintentional loss, ≥4.5 kg in the past year)
Weakness (handgrip strength in the lowest 20% quintile at baseline, adjusted for sex and body mass index)
Exhaustion (poor endurance and energy, self-reported)
Slowness (walking speed under the lowest quintile adjusted for sex and height)
Low physical activity level (lowest quintile of kilocalories of physical activity during the past week)
Additionally, we performed a 5-meter gait speed (5mGS) test43 in all patients. Cognitive function was evaluated by applying the MoCA test41.

Ca2+ Imaging

Ca2+ imaging experiments to measure mitochondrial Ca2+ were performed as we described previously.44,45 Human umbilical vascular endothelial cells (passages 3–7) were plated in glass-bottom culture dishes using the Ca2+ imaging solution, which consists of 138 mmol/L NaCl, 5.3 mmol/L KCl, 1.2 mmol/L NaH2PO4, 1.2 mmol/L MgCl2, and 20 mmol/L HEPES (pH 7.38, adjusted with NaOH); to this solution, we added 5 mmol/L EGTA, 1.8 mmol/L CaCl2, and 5 or 30 mmol/L glucose, according to the experimental settings. Cells were loaded with Rhod-2 AM (3 μM; catolog number R1244; Thermo Fisher Scientific, Waltham, MA) at 37 °C for 30 minutes, followed by washout and 1-hour rest at room temperature for de-esterification: indeed, since Rhod-2 AM has a delocalized positive charge, it preferentially accumulates within the mitochondrial matrix, where it is hydrolyzed and trapped. Fluorescence was detected using a pass-band filter of 545 to 625 nm in response to excitation at 542 nm.

Mitochondrial Oxidative Stress

Human umbilical vein vascular endothelial cells were plated on glass-bottom dishes and treated with different concentrations of glucose and empagliflozin for 24 hours; mitochondrial reactive oxygen species (ROS) were quantified by MitoSOX Red (catalog number M36008; Thermo Fisher Scientific) as we described,7,46 using MitoTracker Green FM (catalog number M7514; Thermo Fisher Scientific) to identify mitochondria.

Real-Time PCR

RT-qPCR was performed using SYBR Green mix as described previously45,47,48; GAPDH was used as an internal standard; primer sequences are listed in Table S1.

Endothelial Permeability Assay

Human brain microvascular endothelial cells (hBMECs; passages 3–6) were cultured as we described previously.7,49 In some experiments, cells were treated with empagliflozin (1 µM), adding 5 or 30 mmol/L glucose for 48 hours. The endothelial permeability assay was performed using fibronectin-coated trans-well filters (Corning, Inc, Corning, NY), as we reported previously.7,49,50

Cell Viability Assay

Cell viability was evaluated in hBMECs using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay, as we described previously.44,51 Briefly, cells were pretreated for 24 hours with 1-μM empagliflozin and then incubated with progressively increasing doses of H2O2 for 5 hours.

Statistical Analysis

Data are presented as n (%) for categorical data (compared using the χ2 test) and mean±SD for normally distributed continuous data (compared via the Student t test or ANOVA followed by Bonferroni post hoc correction). Normality was verified using the Anderson-Darling test. A propensity score analysis was performed as described52 to minimize any selection bias due to the differences in clinical characteristics between the empagliflozin and the no-empagliflozin groups. Briefly, for each patient, a propensity score was calculated by the use of a nonparsimonious multivariable logistic regression, applying the 1:1 nearest neighbor propensity score matching method. Variables that could potentially affect treatment assignment or outcomes were selected, including sociodemographic characteristics, comorbidities, and concomitant drugs. We developed a dispersion model using the Pearson analysis to assess the correlation between glycemia and MoCA score and between glycemia and 5mGS. To adjust for potential confounders (selected a priori based on their clinical significance and possible confounding effect), we performed linear regression analyses with the MoCA score or 5mGS test as dependent variables.
Statistical significance was considered based on 2-tailed P<0.05. All calculations were computed using SPSS, version 26 (IBM Corporation, Armonk, NY), and GraphPad Prism, version 9 (GraphPad by Dotmatics, Boston, MA).

Results

Patient Characteristics

We evaluated 407 patients. Since 22 patients were unwilling to provide clinical information and 60 subjects did not fulfill eligibility criteria, 325 patients were enrolled. To minimize potential selection bias and confounding variables, we propensity score matched 150 patients, 75 treated with 10-mg empagliflozin in addition to standard therapy and 75 not receiving empagliflozin (Figure 1). At baseline, there were no significant differences in age, body mass index, sex distribution, comorbidities, and laboratory parameters between the two groups (Table 1). Notably, the MoCA score and 5mGS were not different among the two study arms at baseline (Table 1).
Table 1. Baseline Characteristics of Our Population
 EmpagliflozinNo empagliflozin
n7575
Sex (M/F)36/3935/40
Age, y78.05±7.578.62±6.2
BMI, kg/m227.7±1.627.9±1.4
SBP, mm Hg121.7±7.3121.0±7.6
DBP, mm Hg76.6±6.576.4±6.7
Heart rate, bpm82.3±9.482.6±9.0
5mGS, m/s0.60±0.120.59±0.13
MoCA score20.32±4.220.35±4.3
Comorbidities, n (%)
 Dyslipidemia39 (52.0)40 (53.0)
 COPD32 (42.7)31 (41.0)
 CKD38 (50.7)39 (52.0)
 Previous stroke12 (16.0)11 (15.0)
Laboratory analyses
 Plasma glucose, mg/dL153.87±71.6153.36±73.1
 HbA1c, mmol/mol61±7.561±6.2
 Creatinine, mg/dL1.0±0.21.0±0.2
5mGS indicates 5-m gait speed; BMI, body mass index; CKD, chronic kidney disease; COPD, chronic obstructive pulmonary disease; DBP, diastolic blood pressure; F, female; HbA1c, glycated hemoglobin; M, male; MoCA, Montreal Cognitive Assessment; and SBP, systolic blood pressure.
Figure 1. Flowchart of the study.

Empagliflozin Significantly Attenuates Cognitive and Physical Impairment

In our population, we observed a strong correlation between glycemia and MoCA score (r²=0.91, P<0.001; Figure 2A) and between glycemia and 5mGS (r²=0.79, P<0.001; Figure 2B).
Figure 2. Empagliflozin attenuates both cognitive and physical impairment in frail patients. Correlation between the Montreal Cognitive Assessment (MoCA) score and glycemia (A; r2, 0.91; P<0.001) and between 5-m gait speed (5mGS) and glycemia (B; r2, 0.79; P<0.001). Differences in the MoCA score (C), 5mGS (D), and frailty (E) at baseline and at 3-month follow-up in empagliflozin-treated and untreated groups (*P<0.05, **P<0.01, ***P<0.001).
We then evaluated the differences in MoCA score (Figure 2C) and 5mGS (Figure 2D) at baseline and at 3-month follow-up in empagliflozin-treated and nontreated patients, observing significantly different favorable effects of the empagliflozin treatment.
Lastly, we assessed how many patients in the two study arms had frailty at 3-month follow-up; the empagliflozin-treated group included only 25.3% of patients with frailty (n=19), while in the nonempagliflozin group, 73.3% of patients (n=55) had frailty (P<0.001; Figure 2E).
Importantly, the significant correlation linking glycemia with MoCA score (Figure 2C) and 5mGS (Figure 2D) was confirmed after adjusting for potential confounders in multivariable regression analyses with MoCA score or 5mGS as dependent variable (Table 2).
Table 2. Linear Regression Analysis in the Empagliflozin Group Using the Basal MoCA Score or 5mGS as the Dependent Variable
 ORSE95% CIP value
Lower boundUpper bound
Basal MoCA score
 Age, y−0.0240.019−0.0620.0140.211
 BMI0.0170.062−0.1050.1390.781
 SBP−0.0090.014−0.0360.0180.530
 DBP−0.0020.017−0.0350.0310.903
 HR0.0220.013−0.0040.0470.093
 Dyslipidemia−0.0990.212−0.5180.3190.640
 CKD−0.1400.221−0.5770.2970.527
 COPD0.0510.227−0.3970.5000.821
 HbA1c−0.7660.422−1.6000.0690.072
 Glycemia−0.0460.005−0.056−0.037<0.001
5mGS
 Age, y−0.0010.001−0.0030.0010.418
 BMI0.0020.003−0.0040.0080.451
 SBP0.0000.001−0.0010.0010.739
 DBP0.0000.001−0.0010.0020.620
 HR0.0010.0010.0000.0020.055
 Dyslipidemia−0.0100.010−0.0290.0100.339
 CKD−0.0010.010−0.0210.0200.946
 COPD0.0040.011−0.0170.0250.711
 HbA1c−0.0020.020−0.0410.0380.936
 Glycemia−0.0020.000−0.002−0.001<0.001
5mGS indicates 5-m gait speed; BMI, body mass index; CKD, chronic kidney disease; COPD, chronic obstructive pulmonary disease; DBP, diastolic blood pressure; HbA1c, glycated hemoglobin; HR, heart rate; MoCA, Montreal Cognitive Assessment; OR, odds ratio; and SBP, systolic blood pressure.

Empagliflozin Reduces Mitochondrial Ca2+ Overload and Mitochondrial Oxidative Stress

To test whether empagliflozin could attenuate mitochondrial Ca2+ overload and ROS production, we cultured human endothelial cells in normal glucose (5 mmol/L), high glucose (30 mmol/L), and high glucose plus empagliflozin for 24 hours. High-glucose condition produced mitochondrial Ca2+ overload (Figure 3AE) and significantly enhanced ROS production in these organelles (Figure 3F), while empagliflozin significantly attenuated these responses (Figure 3).
Figure 3. Empagliflozin mitigates mitochondrial Ca2+ overload and oxidative stress in human endothelial cells. Evaluation of mitochondrial Ca2+ (A–E) and mitochondrial reactive oxygen species (ROS) generation (F) in human umbilical vein endothelial cells treated for 24 hours with the indicated concentrations of glucose (Glu) and empagliflozin (Empa). Mitochondrial Ca2+ (A–E) was measured in at least 30 cells per group, in triplicate independent biological replicates, in response to thapsigargin (Tg; 1 μM), or after the addition of 1.8 mmol/L Ca2+. To assess the production of mitochondrial ROS, cells were stained with MitoTracker Green and mitoSOX Red (F; representative images from quadruplicate experiments; dimensional bar, 20 μm), and these assays have been quantified in the panel on the right; in the violin plot, median (solid line) and quartiles (dotted lines) are indicated. A.U. indicates arbitrary units; and AUC, area under curve. *P<0.05 vs 5 mmol/L Glu+vehicle; #P<0.05 vs 30 mmol/L Glu+vehicle.

SGLT2 Inhibition Mitigates Glucose-Induced Endothelial Permeabilization

Hyperglycemia has been shown to cause an augmented vascular permeability,53,54 and endothelial leakage has been associated to cognitive decline and frailty.55–60 Therefore, we tested whether inhibiting SGLT2 in hBMECs could attenuate endothelial leakage elicited by high glucose concentrations. We observed that empagliflozin significantly attenuated the endothelial permeability induced by high glucose (Figure 4A). To further confirm the effects of empagliflozin on endothelial leakage, we measured the expression levels of mRNA encoding for established junctional proteins (i.e. claudin-5 and occludin) and we found that high glucose concentrations significantly reduced the mRNA levels of both claudin-5 (Figure 4B) and occludin (Figure 4C), and these reductions were mitigated by empagliflozin (Figure 4B and 4C).
Figure 4. Empagliflozin attenuates endothelial leakage and improves cell viability. Effects of empagliflozin (Empa) on cell permeability assessed in human brain microvascular endothelial cells (A) and mRNA levels of occludin (B) and claudin-5 (C); in the violin plot, median (solid line) and quartiles (dotted lines) are indicated. *P<0.05 vs 5 mmol/L Glu+vehicle; #P<0.05 vs 30 mmol/L Glu+vehicle. Effects of Empa (1 μM) on cell viability (D) assessed in human brain microvascular endothelial cells in response to increasing doses of H2O2; the solid line indicates the median, whereas the dotted lines indicate the quartiles. *P<0.05 vs vehicle. Glu indicates glucose.

SGLT2 Inhibition Protects Human Endothelial Cells From Oxidative Stress

Since we demonstrated in human umbilical vascular endothelial cells that empagliflozin attenuates oxidative stress induced by high glucose concentrations, we sought to confirm in hBMECs that inhibiting SGLT2 could improve cell viability in response to oxidative stress. Therefore, we pretreated hBMECs with empagliflozin and then we incubated them with increasing concentrations of H2O2; after 5 hours, we observed that cell viability was significantly improved by empagliflozin (Figure 4D).

Discussion

The compelling combination of clinical data and in vitro assays provided in the present study indicates that SGLT2 inhibition significantly improves cognitive and physical impairment in diabetic and hypertensive patients, most likely reducing oxidative stress in endothelial cells (Graphic Abstract).
Oxidative stress is known to be a common pathogenic substrate in hypertension,61 diabetes,62 and frailty.63,64 Specifically, a dysregulated mitochondrial fitness has been proposed as a potential root of age-related frailty,65 and mitochondrial free radicals are crucial players in endothelial dysfunction.66,67 Henceforth, we sought to determine whether the favorable effects detected in the clinical setting could be attributable to an action on mitochondrial ROS generation at the endothelial level.
Our findings are in agreement with previous reports showing that hyperglycemia causes an augmented generation of mitochondrial ROS68,69; in fact, mitochondrial ROS are recognized as a major cause of clinical complications associated with diabetes.70,71
Hyperglycemia, alongside its actions on endothelial function, has also been associated with memory disturbances, and a potential role in the development of cognitive impairment has been proposed.72,73 Thus, a stable glycemic control is seen as imperative to reduce the incidence of functional decline and to avoid complications.74–78 Herein, we demonstrate that empagliflozin significantly improves cognitive impairment, assessed via MoCA, and physical decline, evaluated via 5mGS test, and we prove that these parameters, which we had previously shown to mutually relate with each other,79 independently correlate with blood glucose levels. Remarkably, our data also indicate a direct effect of empagliflozin on endothelial cells, which goes beyond its actions merely attributable to a reduction of blood glucose80–82 or to the offsetting of insulin resistance.83–86 Specifically, we show that SGLT2 inhibition significantly attenuates mitochondrial Ca2+ overload and the subsequent increase in ROS production in human endothelial cells.
The new 2022 guidelines on the management of heart failure87 of the American Heart Association, the American College of Cardiology, and the Heart Failure Society of America recommend SGLT2 inhibitors as class 1A in HFrEF and class 2A in HFmrEF and HFpEF (conditions in which ARNI and MRA have weaker recommendations: class 2B). These guidelines are mainly based on the protective effects of SGLT2 inhibitors on cardiovascular outcomes, independent of the glucose-lowering effects, revealed by recent clinical trials: the DAPA-HF trial (Dapagliflozin and Prevention of Adverse Outcomes in Heart Failure),88 EMPEROR-HF (Empagliflozin Outcome Trial in Patients With Chronic Heart Failure With Preserved Ejection Fraction),89 and EMPEROR-PRESERVED (Empagliflozin Outcome Trial in Patients With Chronic Heart Failure With Preserved Ejection Fraction).21 However, the exact mechanism underlying these beneficial effects on cardiovascular outcomes has remained hitherto elusive.
Our results are consistent with a pilot study testing the acute effects of dapagliflozin in 16 diabetic patients, showing a marked reduction of urinary isoprostanes, a surrogate of systemic oxidative stress, 48 hours after administration compared to baseline values.90 In our study, we also proved that empagliflozin counteracts the increased endothelial permeability induced by high glucose, by regulating at the transcriptional level the expression of occludin and claudin-5, tight junction proteins that have been shown to partake in cognitive impairment.91–93 Claudin-5 levels were also shown to be reduced in the nucleus accumbens of patients with depression.94,95 Additionally, empagliflozin significantly prevented the death of cerebral endothelial cells induced by oxidative stress, and these data are particularly relevant when considering that ROS production is fundamental in processes like senescence and aging.96–100 Therefore, our findings are highly suggestive for a potential role of SGLT2 inhibitors in preventing neurodegenerative disorders.
Strengths of our study include its prospective nature, the analysis of a real-world homogeneous population of elderly patients with diabetes and hypertension, the integration of clinical and in vitro data, and the consistency of the beneficial effects of empagliflozin in 2 different types of human endothelial cells, namely cells obtained from the umbilical vein and brain microvascular endothelial cells. Nevertheless, our study is not exempt from limitations, including the relatively brief follow-up, having tested the effects of only one SGLT2 inhibitor, and having enrolled only Caucasian patients.

Perspectives

Based on our results and on the emerging pleiotropic effects of SGLT2 inhibitors,26 we propose SGLT2 inhibitors as anti-frailty drugs. This view is strongly supported by the beneficial effects of empagliflozin on endothelial cells, in terms of mitigated mitochondrial Ca2+ overload, reduced oxidative stress, and improved cell viability.

Footnote

Nonstandard Abbreviations and Acronyms

5mGS
5-meter Gait Speed
ARNI
Angiotensin Receptor-Neprilysin Inhibitors
DAPA-HF
Dapagliflozin and Prevention of Adverse Outcomes in Heart Failure
EGTA
Ethylene Glycol-bis(β-aminoethyl ether)-N,N,N′,N′-Tetraacetic Acid
EMPA
Empagliflozin
EMPEROR-HF
Empagliflozin Outcome Trial in Patients With Chronic Heart Failure With Preserved Ejection Fraction
EMPEROR-PRESERVED
Empagliflozin Outcome Trial in Patients With Chronic Heart Failure With Preserved Ejection Fraction
Glu
Glucose
hBMEC
Human Brain Microvascular Endothelial Cell
HUVEC
Human Umbilical Vein Endothelial Cell
MoCA
Montreal Cognitive Assessment
MRA
Mineralocorticoid Receptor Antagonists
MTT
3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide
Rhod 2 AM
Rhod-2 Acetoxymethyl
ROS
Reactive Oxygen Species
RT-qPCR
Reverse Transcription quantitative Real-Time Polymerase Chain Reaction
SGLT2
Sodium-GLucose co-Transporter 2

Supplemental Material

File (hyp_hype-2022-19586_supp2.pdf)

References

1.
Tsui A, Searle SD, Bowden H, Hoffmann K, Hornby J, Goslett A, Weston-Clarke M, Howes LH, Street R, Perera R, et al. The effect of baseline cognition and delirium on long-term cognitive impairment and mortality: a prospective population-based study. Lancet Healthy Longev. 2022;3:e232–e241. doi: 10.1016/S2666-7568(22)00013-7
2.
Promislow D, Anderson RM, Scheffer M, Crespi B, DeGregori J, Harris K, Horowitz BN, Levine ME, Riolo MA, Schneider DS, et al. Resilience integrates concepts in aging research. iScience. 2022;25:104199. doi: 10.1016/j.isci.2022.104199
3.
Marinus N, Vigorito C, Giallauria F, Dendale P, Meesen R, Bokken K, Haenen L, Jansegers T, Vandenheuvel Y, Scherrenberg M, et al. Frailty test battery development including physical, socio-psychological and cognitive domains for cardiovascular disease patients: a preliminary study. J Clin Med. 2022;11:1926. doi: 10.3390/jcm11071926
4.
Kremer KM, Braisch U, Rothenbacher D, Denkinger M, Dallmeier D; ActiFE Study Group. Systolic blood pressure and mortality in community-dwelling older adults: frailty as an effect modifier. Hypertension. 2022;79:24–32. doi: 10.1161/HYPERTENSIONAHA.121.17530
5.
Rouch L, Rolland Y, Hanon O, Vidal JS, Cestac P, Sallerin B, Andrieu S, Vellas B, Barreto PS; MAPT/DSA Group. Blood pressure, antihypertensive drugs, and incident frailty: the Multidomain Alzheimer Preventive Trial (MAPT). Maturitas. 2022;162:8–14. doi: 10.1016/j.maturitas.2022.03.001
6.
Elliott ML, Caspi A, Houts RM, Ambler A, Broadbent JM, Hancox RJ, Harrington H, Hogan S, Keenan R, Knodt A, et al. Disparities in the pace of biological aging among midlife adults of the same chronological age have implications for future frailty risk and policy. Nat Aging. 2021;1:295–308. doi: 10.1038/s43587-021-00044-4
7.
Mone P, Gambardella J, Pansini A, de Donato A, Martinelli G, Boccalone E, Matarese A, Frullone S, Santulli G. Cognitive impairment in frail hypertensive elderly patients: role of hyperglycemia. Cells. 2021;10:2115. doi: 10.3390/cells10082115
8.
Li G, Prior JC, Leslie WD, Thabane L, Papaioannou A, Josse RG, Kaiser SM, Kovacs CS, Anastassiades T, Towheed T, et al; CaMos Research Group. Frailty and risk of fractures in patients with type 2 diabetes. Diabetes Care. 2019;42:507–513. doi: 10.2337/dc18-1965
9.
Ida S, Kaneko R, Imataka K, Murata K. Relationship between frailty and mortality, hospitalization, and cardiovascular diseases in diabetes: a systematic review and meta-analysis. Cardiovasc Diabetol. 2019;18:81. doi: 10.1186/s12933-019-0885-2
10.
Odegaard AO, Jacobs DR, Sanchez OA, Goff DC, Reiner AP, Gross MD. Oxidative stress, inflammation, endothelial dysfunction and incidence of type 2 diabetes. Cardiovasc Diabetol. 2016;15:51. doi: 10.1186/s12933-016-0369-6
11.
Clegg A, Hassan-Smith Z. Frailty and the endocrine system. Lancet Diabetes Endocrinol. 2018;6:743–752. doi: 10.1016/S2213-8587(18)30110-4
12.
Teo YN, Ting AZH, Teo YH, Chong EY, Tan JTA, Syn NL, Chia AZQ, Ong HT, Cheong AJY, Li TY, et al. Effects of sodium/glucose cotransporter 2 (SGLT2) inhibitors and combined SGLT1/2 inhibitors on cardiovascular, metabolic, renal, and safety outcomes in patients with diabetes: a network meta-analysis of 111 randomized controlled trials. Am J Cardiovasc Drugs. 2022;22:299–323. doi: 10.1007/s40256-022-00528-7
13.
Draznin B, Aroda VR, Bakris G, Benson G, Brown FM, Freeman R, Green J, Huang E, Isaacs D, Kahan S, et al. Older adults: standards of medical care in diabetes-2022. Diabetes Care. 2022;45:S195–S207.
14.
Nagasu H, Yano Y, Kanegae H, Heerspink HJL, Nangaku M, Hirakawa Y, Sugawara Y, Nakagawa N, Tani Y, Wada J, et al. Kidney outcomes associated with SGLT2 inhibitors versus other glucose-lowering drugs in real-world clinical practice: the Japan chronic kidney disease database. Diabetes Care. 2021;44:2542–2551. doi: 10.2337/dc21-1081
15.
Katsuno K, Fujimori Y, Takemura Y, Hiratochi M, Itoh F, Komatsu Y, Fujikura H, Isaji M. Sergliflozin, a novel selective inhibitor of low-affinity sodium glucose cotransporter (SGLT2), validates the critical role of SGLT2 in renal glucose reabsorption and modulates plasma glucose level. J Pharmacol Exp Ther. 2007;320:323–330. doi: 10.1124/jpet.106.110296
16.
Neuen BL, Young T, Heerspink HJL, Neal B, Perkovic V, Billot L, Mahaffey KW, Charytan DM, Wheeler DC, Arnott C, et al. SGLT2 inhibitors for the prevention of kidney failure in patients with type 2 diabetes: a systematic review and meta-analysis. Lancet Diabetes Endocrinol. 2019;7:845–854. doi: 10.1016/S2213-8587(19)30256-6
17.
Giorgino F, Vora J, Fenici P, Solini A. Renoprotection with SGLT2 inhibitors in type 2 diabetes over a spectrum of cardiovascular and renal risk. Cardiovasc Diabetol. 2020;19:196. doi: 10.1186/s12933-020-01163-9
18.
Wei W, Liu J, Chen S, Xu X, Guo D, He Y, Huang Z, Wang B, Huang H, Li Q, et al. Sodium glucose cotransporter type 2 inhibitors improve cardiorenal outcome of patients with coronary artery disease: a meta-analysis. Front Endocrinol (Lausanne). 2022;13:850836. doi: 10.3389/fendo.2022.850836
19.
Jiang Y, Yang P, Fu L, Sun L, Shen W, Wu Q. Comparative cardiovascular outcomes of SGLT2 inhibitors in type 2 diabetes mellitus: a network meta-analysis of randomized controlled trials. Front Endocrinol (Lausanne). 2022;13:802992. doi: 10.3389/fendo.2022.802992
20.
Braunwald E. SGLT2 inhibitors: the statins of the 21st century. Eur Heart J. 2022;43:1029–1030. doi: 10.1093/eurheartj/ehab765
21.
Anker SD, Butler J, Filippatos G, Ferreira JP, Bocchi E, Böhm M, Brunner-La Rocca HP, Choi DJ, Chopra V, Chuquiure-Valenzuela E, et al; EMPEROR-Preserved Trial Investigators. Empagliflozin in heart failure with a preserved ejection fraction. N Engl J Med. 2021;385:1451–1461. doi: 10.1056/NEJMoa2107038
22.
Santos-Gallego CG, Vargas-Delgado AP, Requena-Ibanez JA, Garcia-Ropero A, Mancini D, Pinney S, Macaluso F, Sartori S, Roque M, Sabatel-Perez F, et al; EMPA-TROPISM (ATRU-4) Investigators. Randomized trial of empagliflozin in nondiabetic patients with heart failure and reduced ejection fraction. J Am Coll Cardiol. 2021;77:243–255. doi: 10.1016/j.jacc.2020.11.008
23.
Zinman B, Wanner C, Lachin JM, Fitchett D, Bluhmki E, Hantel S, Mattheus M, Devins T, Johansen OE, Woerle HJ, et al; EMPA-REG OUTCOME Investigators. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med. 2015;373:2117–2128. doi: 10.1056/NEJMoa1504720
24.
Sciannameo V, Berchialla P, Avogaro A, Fadini GP; DARWIN-T2D Network. Transposition of cardiovascular outcome trial effects to the real-world population of patients with type 2 diabetes. Cardiovasc Diabetol. 2021;20:103. doi: 10.1186/s12933-021-01300-y
25.
Yang CT, Peng ZY, Chen YC, Ou HT, Kuo S. Cardiovascular benefits with favorable renal, amputation and hypoglycemic outcomes of SGLT-2 inhibitors in type 2 diabetes from the asian perspective: a population-based cohort study and systematic review. Front Endocrinol (Lausanne). 2022;13:836365. doi: 10.3389/fendo.2022.836365
26.
Sarafidis P, Papadopoulos CE, Kamperidis V, Giannakoulas G, Doumas M. Cardiovascular protection with sodium-glucose cotransporter-2 inhibitors and mineralocorticoid receptor antagonists in chronic kidney disease: a milestone achieved. Hypertension. 2021;77:1442–1455. doi: 10.1161/HYPERTENSIONAHA.121.17005
27.
Jun S, Aon MA, Paolocci N. Empagliflozin and HFrEF: known and possible benefits of NHE1 inhibition. JACC Basic Transl Sci. 2019;4:841–844. doi: 10.1016/j.jacbts.2019.10.005
28.
Jensen J, Omar M, Kistorp C, Tuxen C, Gustafsson I, Køber L, Gustafsson F, Faber J, Malik ME, Fosbøl EL, et al. Effects of empagliflozin on estimated extracellular volume, estimated plasma volume, and measured glomerular filtration rate in patients with heart failure (Empire HF Renal): a prespecified substudy of a double-blind, randomised, placebo-controlled trial. Lancet Diabetes Endocrinol. 2021;9:106–116. doi: 10.1016/S2213-8587(20)30382-X
29.
Prattichizzo F, De Nigris V, Micheloni S, La Sala L, Ceriello A. Increases in circulating levels of ketone bodies and cardiovascular protection with SGLT2 inhibitors: is low-grade inflammation the neglected component? Diabetes Obes Metab. 2018;20:2515–2522. doi: 10.1111/dom.13488
30.
Zhang A, Luo X, Meng H, Kang J, Qin G, Chen Y, Zhang X. Sodium glucose cotransporter 2 inhibitors reduce the risk of heart failure hospitalization in patients with type 2 diabetes mellitus: a systematic review and meta-analysis of randomized controlled trials. Front Endocrinol (Lausanne). 2020;11:604250. doi: 10.3389/fendo.2020.604250
31.
Huang D, Ju F, Du L, Liu T, Zuo Y, Abbott GW, Hu Z. Empagliflozin protects against pulmonary ischemia/reperfusion injury via an extracellular signal-regulated kinases 1 and 2-dependent mechanism. J Pharmacol Exp Ther. 2022;380:230–241. doi: 10.1124/jpet.121.000956
32.
Mone P, Lombardi A, Kansakar U, Varzideh F, Jankauskas SS, Pansini A, Marzocco S, Famiglietti M, Macina G, Frullone S, Santulli G. Empagliflozin improves the microRNA signature of endothelial dysfunction in patients with HFpEF and diabetes. J Pharmacol Exp Ther. 2022; in press.
33.
De Stefano A, Tesauro M, Di Daniele N, Vizioli G, Schinzari F, Cardillo C. Mechanisms of SGLT2 (sodium-glucose transporter type 2) inhibition-induced relaxation in arteries from human visceral adipose tissue. Hypertension. 2021;77:729–738. doi: 10.1161/HYPERTENSIONAHA.120.16466
34.
Verma S, Mazer CD, Yan AT, Mason T, Garg V, Teoh H, Zuo F, Quan A, Farkouh ME, Fitchett DH, et al. Effect of empagliflozin on left ventricular mass in patients with type 2 diabetes mellitus and coronary artery disease: the EMPA-HEART CardioLink-6 randomized clinical trial. Circulation. 2019;140:1693–1702. doi: 10.1161/CIRCULATIONAHA.119.042375
35.
Oshima H, Miki T, Kuno A, Mizuno M, Sato T, Tanno M, Yano T, Nakata K, Kimura Y, Abe K, et al. Empagliflozin, an SGLT2 inhibitor, reduced the mortality rate after acute myocardial infarction with modification of cardiac metabolomes and antioxidants in diabetic rats. J Pharmacol Exp Ther. 2019;368:524–534. doi: 10.1124/jpet.118.253666
36.
Salvatore T, Galiero R, Caturano A, Rinaldi L, Di Martino A, Albanese G, Di Salvo J, Epifani R, Marfella R, Docimo G, et al. An overview of the cardiorenal protective mechanisms of SGLT2 inhibitors. Int J Mol Sci. 2022;23:3651. doi: 10.3390/ijms23073651
37.
Pietschner R, Kolwelter J, Bosch A, Striepe K, Jung S, Kannenkeril D, Ott C, Schiffer M, Achenbach S, Schmieder RE. Effect of empagliflozin on ketone bodies in patients with stable chronic heart failure. Cardiovasc Diabetol. 2021;20:219. doi: 10.1186/s12933-021-01410-7
38.
American Diabetes Association. Classification and diagnosis of diabetes. Diabetes Care. 2017;40:S11–S24.
39.
Unger T, Borghi C, Charchar F, Khan NA, Poulter NR, Prabhakaran D, Ramirez A, Schlaich M, Stergiou GS, Tomaszewski M, et al. 2020 International Society of Hypertension global hypertension practice guidelines. Hypertension. 2020;75:1334–1357. doi: 10.1161/HYPERTENSIONAHA.120.15026
40.
Fried LP, Tangen CM, Walston J, Newman AB, Hirsch C, Gottdiener J, Seeman T, Tracy R, Kop WJ, Burke G, et al; Cardiovascular Health Study Collaborative Research Group. Frailty in older adults: evidence for a phenotype. J Gerontol A Biol Sci Med Sci. 2001;56:M146–M156. doi: 10.1093/gerona/56.3.m146
41.
Mone P, Lombardi A, Gambardella J, Pansini A, Macina G, Morgante M, Frullone S, Santulli G. Empagliflozin improves cognitive impairment in frail older adults with type 2 diabetes and heart failure with preserved ejection fraction. Diabetes Care. 2022;45:1247–1251. doi: 10.2337/dc21-2434
42.
Pansini A, Lombardi A, Morgante M, Frullone S, Marro A, Rizzo M, Martinelli G, Boccalone E, De Luca A, Santulli G, et al. Hyperglycemia and physical impairment in frail hypertensive older adults. Front Endocrinol (Lausanne). 2022;13:831556. doi: 10.3389/fendo.2022.831556
43.
Afilalo J, Alexander KP, Mack MJ, Maurer MS, Green P, Allen LA, Popma JJ, Ferrucci L, Forman DE. Frailty assessment in the cardiovascular care of older adults. J Am Coll Cardiol. 2014;63:747–762. doi: 10.1016/j.jacc.2013.09.070
44.
Lombardi A, Trimarco B, Iaccarino G, Santulli G. Impaired mitochondrial calcium uptake caused by tacrolimus underlies beta-cell failure. Cell Commun Signal. 2017;15:47. doi: 10.1186/s12964-017-0203-0
45.
Lombardi A, Gambardella J, Du XL, Sorriento D, Mauro M, Iaccarino G, Trimarco B, Santulli G. Sirolimus induces depletion of intracellular calcium stores and mitochondrial dysfunction in pancreatic beta cells. Sci Rep. 2017;7:15823. doi: 10.1038/s41598-017-15283-y
46.
Santulli G, Xie W, Reiken SR, Marks AR. Mitochondrial calcium overload is a key determinant in heart failure. Proc Natl Acad Sci USA. 2015;112:11389–11394. doi: 10.1073/pnas.1513047112
47.
Morelli MB, Shu J, Sardu C, Matarese A, Santulli G. Cardiosomal microRNAs are essential in post-infarction myofibroblast phenoconversion. Int J Mol Sci. 2019;21:E201. doi: 10.3390/ijms21010201
48.
Yuan Q, Chen Z, Santulli G, Gu L, Yang ZG, Yuan ZQ, Zhao YT, Xin HB, Deng KY, Wang SQ, et al. Functional role of calstabin2 in age-related cardiac alterations. Sci Rep. 2014;4:7425. doi: 10.1038/srep07425
49.
Mone P, Gambardella J, Wang X, Jankauskas SS, Matarese A, Santulli G. miR-24 targets the transmembrane glycoprotein neuropilin-1 in human brain microvascular endothelial cells. Noncoding RNA. 2021;7:9. doi: 10.3390/ncrna7010009
50.
Gambardella J, Sorriento D, Bova M, Rusciano M, Loffredo S, Wang X, Petraroli A, Carucci L, Mormile I, Oliveti M, et al. Role of endothelial G protein-coupled receptor kinase 2 in angioedema. Hypertension. 2020;76:1625–1636. doi: 10.1161/HYPERTENSIONAHA.120.15130
51.
De Vitis S, Sonia Treglia A, Ulianich L, Turco S, Terrazzano G, Lombardi A, Miele C, Garbi C, Beguinot F, Di Jeso B. Tyr phosphatase-mediated P-ERK inhibition suppresses senescence in EIA + v-raf transformed cells, which, paradoxically, are apoptosis-protected in a MEK-dependent manner. Neoplasia. 2011;13:120–130. doi: 10.1593/neo.101152
52.
Mone P, Gambardella J, Minicucci F, Lombardi A, Mauro C, Santulli G. Hyperglycemia drives stent restenosis in STEMI patients. Diabetes Care. 2021;44:e192–e193. doi: 10.2337/dc21-0939
53.
Aronson D, Rayfield EJ. How hyperglycemia promotes atherosclerosis: molecular mechanisms. Cardiovasc Diabetol. 2002;1:1. doi: 10.1186/1475-2840-1-1
54.
Clemmer JS, Xiang L, Lu S, Mittwede PN, Hester RL. Hyperglycemia-mediated oxidative stress increases pulmonary vascular permeability. Microcirculation. 2016;23:221–229. doi: 10.1111/micc.12267
55.
de Montgolfier O, Pinçon A, Pouliot P, Gillis MA, Bishop J, Sled JG, Villeneuve L, Ferland G, Lévy BI, Lesage F, et al. High systolic blood pressure induces cerebral microvascular endothelial dysfunction, neurovascular unit damage, and cognitive decline in mice. Hypertension. 2019;73:217–228. doi: 10.1161/HYPERTENSIONAHA.118.12048
56.
Chen YC, Lu BZ, Shu YC, Sun YT. Spatiotemporal dynamics of cerebral vascular permeability in type 2 diabetes-related cerebral microangiopathy. Front Endocrinol (Lausanne). 2021;12:805637. doi: 10.3389/fendo.2021.805637
57.
Rajeev V, Fann DY, Dinh QN, Kim HA, De Silva TM, Lai MKP, Chen CL, Drummond GR, Sobey CG, Arumugam TV. Pathophysiology of blood brain barrier dysfunction during chronic cerebral hypoperfusion in vascular cognitive impairment. Theranostics. 2022;12:1639–1658. doi: 10.7150/thno.68304
58.
Adamski MG, Sternak M, Mohaissen T, Kaczor D, Wierońska JM, Malinowska M, Czaban I, Byk K, Lyngsø KS, Przyborowski K, et al. Vascular cognitive impairment linked to brain endothelium inflammation in early stages of heart failure in mice. J Am Heart Assoc. 2018;7:e007694. doi: 10.1161/JAHA.117.007694
59.
Li M, Li Y, Zuo L, Hu W, Jiang T. Increase of blood-brain barrier leakage is related to cognitive decline in vascular mild cognitive impairment. BMC Neurol. 2021;21:159. doi: 10.1186/s12883-021-02189-6
60.
Wang F, Cao Y, Ma L, Pei H, Rausch WD, Li H. Dysfunction of cerebrovascular endothelial cells: prelude to vascular dementia. Front Aging Neurosci. 2018;10:376. doi: 10.3389/fnagi.2018.00376
61.
Guzik TJ, Touyz RM. Oxidative stress, inflammation, and vascular aging in hypertension. Hypertension. 2017;70:660–667. doi: 10.1161/HYPERTENSIONAHA.117.07802
62.
Vaidya AR, Wolska N, Vara D, Mailer RK, Schröder K, Pula G. Diabetes and thrombosis: a central role for vascular oxidative stress. Antioxidants (Basel). 2021;10:706. doi: 10.3390/antiox10050706
63.
Alabadi B, Civera M, De la Rosa A, Martinez-Hervas S, Gomez-Cabrera MC, Real JT. Frailty is associated with oxidative stress in older patients with type 2 diabetes. Nutrients. 2021;13:3983. doi: 10.3390/nu13113983
64.
Bernabeu-Wittel M, Gómez-Díaz R, González-Molina Á, Vidal-Serrano S, Díez-Manglano J, Salgado F, Soto-Martín M, Ollero-Baturone M, On Behalf Of The Proteo Researchers. Oxidative stress, telomere shortening, and apoptosis associated to sarcopenia and frailty in patients with multimorbidity. J Clin Med. 2020;9:E2669. doi: 10.3390/jcm9082669
65.
Ferrucci L, Zampino M. A mitochondrial root to accelerated ageing and frailty. Nat Rev Endocrinol. 2020;16:133–134. doi: 10.1038/s41574-020-0319-y
66.
Shenouda SM, Widlansky ME, Chen K, Xu G, Holbrook M, Tabit CE, Hamburg NM, Frame AA, Caiano TL, Kluge MA, et al. Altered mitochondrial dynamics contributes to endothelial dysfunction in diabetes mellitus. Circulation. 2011;124:444–453. doi: 10.1161/CIRCULATIONAHA.110.014506
67.
Widlansky ME, Gutterman DD. Regulation of endothelial function by mitochondrial reactive oxygen species. Antioxid Redox Signal. 2011;15:1517–1530. doi: 10.1089/ars.2010.3642
68.
Zhang H, Shen Y, Kim IM, Weintraub NL, Tang Y. The impaired bioenergetics of diabetic cardiac microvascular endothelial cells. Front Endocrinol (Lausanne). 2021;12:642857. doi: 10.3389/fendo.2021.642857
69.
Suzuki K, Olah G, Modis K, Coletta C, Kulp G, Gerö D, Szoleczky P, Chang T, Zhou Z, Wu L, et al. Hydrogen sulfide replacement therapy protects the vascular endothelium in hyperglycemia by preserving mitochondrial function. Proc Natl Acad Sci USA. 2011;108:13829–13834. doi: 10.1073/pnas.1105121108
70.
Kaludercic N, Di Lisa F. Mitochondrial ROS formation in the pathogenesis of diabetic cardiomyopathy. Front Cardiovasc Med. 2020;7:12. doi: 10.3389/fcvm.2020.00012
71.
Nishikawa T, Araki E. Impact of mitochondrial ROS production in the pathogenesis of diabetes mellitus and its complications. Antioxid Redox Signal. 2007;9:343–353. doi: 10.1089/ars.2006.1458
72.
Kerti L, Witte AV, Winkler A, Grittner U, Rujescu D, Flöel A. Higher glucose levels associated with lower memory and reduced hippocampal microstructure. Neurology. 2013;81:1746–1752. doi: 10.1212/01.wnl.0000435561.00234.ee
73.
Rom S, Zuluaga-Ramirez V, Gajghate S, Seliga A, Winfield M, Heldt NA, Kolpakov MA, Bashkirova YV, Sabri AK, Persidsky Y. Hyperglycemia-driven neuroinflammation compromises BBB leading to memory loss in both diabetes mellitus (DM) type 1 and type 2 mouse models. Mol Neurobiol. 2019;56:1883–1896. doi: 10.1007/s12035-018-1195-5
74.
ADA. Older adults: standards of medical care in diabetes-2021. Diabetes Care. 2021;44:S168–S179.
75.
Laiteerapong N, Karter AJ, Liu JY, Moffet HH, Sudore R, Schillinger D, John PM, Huang ES. Correlates of quality of life in older adults with diabetes: the diabetes & aging study. Diabetes Care. 2011;34:1749–1753. doi: 10.2337/dc10-2424
76.
Shaw KM. Overcoming the hurdles to achieving glycemic control. Metabolism. 2006;55(5 suppl 1):S6–S9. doi: 10.1016/j.metabol.2006.02.001
77.
Zaslavsky O, Walker RL, Crane PK, Gray SL, Larson EB. Glucose levels and risk of frailty. J Gerontol A Biol Sci Med Sci. 2016;71:1223–1229. doi: 10.1093/gerona/glw024
78.
Tamura Y, Omura T, Toyoshima K, Araki A. Nutrition management in older adults with diabetes: a review on the importance of shifting prevention strategies from metabolic syndrome to frailty. Nutrients. 2020;12:E3367. doi: 10.3390/nu12113367
79.
Mone P, Gambardella J, Lombardi A, Pansini A, De Gennaro S, Leo AL, Famiglietti M, Marro A, Morgante M, Frullone S, et al. Correlation of physical and cognitive impairment in diabetic and hypertensive frail older adults. Cardiovasc Diabetol. 2022;21:10. doi: 10.1186/s12933-021-01442-z
80.
Vallon V, Gerasimova M, Rose MA, Masuda T, Satriano J, Mayoux E, Koepsell H, Thomson SC, Rieg T. SGLT2 inhibitor empagliflozin reduces renal growth and albuminuria in proportion to hyperglycemia and prevents glomerular hyperfiltration in diabetic Akita mice. Am J Physiol Renal Physiol. 2014;306:F194–F204. doi: 10.1152/ajprenal.00520.2013
81.
Musso G, Gambino R, Cassader M, Pagano G. A novel approach to control hyperglycemia in type 2 diabetes: sodium glucose co-transport (SGLT) inhibitors: systematic review and meta-analysis of randomized trials. Ann Med. 2012;44:375–393. doi: 10.3109/07853890.2011.560181
82.
O’Brien TP, Jenkins EC, Estes SK, Castaneda AV, Ueta K, Farmer TD, Puglisi AE, Swift LL, Printz RL, Shiota M. Correcting postprandial hyperglycemia in zucker diabetic fatty rats with an SGLT2 inhibitor restores glucose effectiveness in the liver and reduces insulin resistance in skeletal muscle. Diabetes. 2017;66:1172–1184. doi: 10.2337/db16-1410
83.
Al-Wakeel DE, El-Kashef DH, Nader MA. Renoprotective effect of empagliflozin in cafeteria diet-induced insulin resistance in rats: modulation of HMGB-1/TLR-4/NF-κB axis. Life Sci. 2022;301:120633. doi: 10.1016/j.lfs.2022.120633
84.
Kullmann S, Hummel J, Wagner R, Dannecker C, Vosseler A, Fritsche L, Veit R, Kantartzis K, Machann J, Birkenfeld AL, et al. Empagliflozin improves insulin sensitivity of the hypothalamus in humans with prediabetes: a randomized, double-blind, placebo-controlled, phase 2 trial. Diabetes Care. 2022;45:398–406. doi: 10.2337/dc21-1136
85.
Xu L, Nagata N, Chen G, Nagashimada M, Zhuge F, Ni Y, Sakai Y, Kaneko S, Ota T. Empagliflozin reverses obesity and insulin resistance through fat browning and alternative macrophage activation in mice fed a high-fat diet. BMJ Open Diabetes Res Care. 2019;7:e000783. doi: 10.1136/bmjdrc-2019-000783
86.
Paolisso P, Bergamaschi L, Santulli G, Gallinoro E, Cesaro A, Gragnano F, Sardu C, Mileva N, Foà A, Armillotta M, et al. Infarct size, inflammatory burden, and admission hyperglycemia in diabetic patients with acute myocardial infarction treated with SGLT2-inhibitors: a multicenter international registry. Cardiovasc Diabetol. 2022;21:77. doi: 10.1186/s12933-022-01506-8
87.
Santulli G, Wang X, Mone P. Updated ACC/AHA/HFSA 2022 guidelines on heart failure: what’s new? from epidemiology to clinical management. Eur Heart J Cardiovasc Pharmacother. doi: 10.1093/ehjcvp/pvac029
88.
McMurray JJV, Solomon SD, Inzucchi SE, Køber L, Kosiborod MN, Martinez FA, Ponikowski P, Sabatine MS, Anand IS, Bělohlávek J, et al; DAPA-HF Trial Committees and Investigators. Dapagliflozin in patients with heart failure and reduced ejection fraction. N Engl J Med. 2019;381:1995–2008. doi: 10.1056/NEJMoa1911303
89.
Packer M, Anker SD, Butler J, Filippatos G, Pocock SJ, Carson P, Januzzi J, Verma S, Tsutsui H, Brueckmann M, et al; EMPEROR-Reduced Trial Investigators. Cardiovascular and renal outcomes with empagliflozin in heart failure. N Engl J Med. 2020;383:1413–1424. doi: 10.1056/NEJMoa2022190
90.
Solini A, Giannini L, Seghieri M, Vitolo E, Taddei S, Ghiadoni L, Bruno RM. Dapagliflozin acutely improves endothelial dysfunction, reduces aortic stiffness and renal resistive index in type 2 diabetic patients: a pilot study. Cardiovasc Diabetol. 2017;16:138. doi: 10.1186/s12933-017-0621-8
91.
Yamazaki Y, Shinohara M, Shinohara M, Yamazaki A, Murray ME, Liesinger AM, Heckman MG, Lesser ER, Parisi JE, Petersen RC, et al. Selective loss of cortical endothelial tight junction proteins during Alzheimer’s disease progression. Brain. 2019;142:1077–1092. doi: 10.1093/brain/awz011
92.
Greene C, Hanley N, Campbell M. Claudin-5: gatekeeper of neurological function. Fluids Barriers CNS. 2019;16:3. doi: 10.1186/s12987-019-0123-z
93.
Hussain B, Fang C, Chang J. Blood-brain barrier breakdown: an emerging biomarker of cognitive impairment in normal aging and dementia. Front Neurosci. 2021;15:688090. doi: 10.3389/fnins.2021.688090
94.
Menard C, Pfau ML, Hodes GE, Kana V, Wang VX, Bouchard S, Takahashi A, Flanigan ME, Aleyasin H, LeClair KB, et al. Social stress induces neurovascular pathology promoting depression. Nat Neurosci. 2017;20:1752–1760. doi: 10.1038/s41593-017-0010-3
95.
Dudek KA, Dion-Albert L, Lebel M, LeClair K, Labrecque S, Tuck E, Ferrer Perez C, Golden SA, Tamminga C, Turecki G, et al. Molecular adaptations of the blood-brain barrier promote stress resilience vs. depression. Proc Natl Acad Sci USA. 2020;117:3326–3336. doi: 10.1073/pnas.1914655117
96.
Reskiawan A Kadir R, Alwjwaj M, Ahmad Othman O, Rakkar K, Sprigg N, Bath PM, Bayraktutan U. Inhibition of oxidative stress delays senescence and augments functional capacity of endothelial progenitor cells. Brain Res. 2022;1787:147925. doi: 10.1016/j.brainres.2022.147925
97.
Chandrasekaran A, Idelchik MDPS, Melendez JA. Redox control of senescence and age-related disease. Redox Biol. 2017;11:91–102. doi: 10.1016/j.redox.2016.11.005
98.
Kiss T, Tarantini S, Csipo T, Balasubramanian P, Nyúl-Tóth Á, Yabluchanskiy A, Wren JD, Garman L, Huffman DM, Csiszar A, et al. Circulating anti-geronic factors from heterochonic parabionts promote vascular rejuvenation in aged mice: transcriptional footprint of mitochondrial protection, attenuation of oxidative stress, and rescue of endothelial function by young blood. Geroscience. 2020;42:727–748. doi: 10.1007/s11357-020-00180-6
99.
Valeri A, Chiricosta L, Calcaterra V, Biasin M, Cappelletti G, Carelli S, Zuccotti GV, Bramanti P, Pelizzo G, Mazzon E, et al. Transcriptomic analysis of HCN-2 cells suggests connection among oxidative stress, senescence, and neuron death after SARS-CoV-2 infection. Cells. 2021;10:2189. doi: 10.3390/cells10092189
100.
Liguori I, Russo G, Curcio F, Bulli G, Aran L, Della-Morte D, Gargiulo G, Testa G, Cacciatore F, Bonaduce D, et al. Oxidative stress, aging, and diseases. Clin Interv Aging. 2018;13:757–772. doi: 10.2147/CIA.S158513

eLetters(0)

eLetters should relate to an article recently published in the journal and are not a forum for providing unpublished data. Comments are reviewed for appropriate use of tone and language. Comments are not peer-reviewed. Acceptable comments are posted to the journal website only. Comments are not published in an issue and are not indexed in PubMed. Comments should be no longer than 500 words and will only be posted online. References are limited to 10. Authors of the article cited in the comment will be invited to reply, as appropriate.

Comments and feedback on AHA/ASA Scientific Statements and Guidelines should be directed to the AHA/ASA Manuscript Oversight Committee via its Correspondence page.

Information & Authors

Information

Published In

Go to Hypertension
Go to Hypertension
Hypertension
Pages: 1633 - 1643
PubMed: 35703100

Versions

You are viewing the most recent version of this article.

History

Received: 22 April 2022
Accepted: 1 June 2022
Published online: 15 June 2022
Published in print: August 2022

Permissions

Request permissions for this article.

Keywords

  1. aging
  2. diabetes mellitus
  3. frailty
  4. hyperglycemia
  5. mitochondria
  6. occludin
  7. sodium-glucose transporter 2 inhibitors

Subjects

Authors

Affiliations

Pasquale Mone* [email protected]
Department of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Aging Research, Einstein-Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York City, NY (P.M., F.V., S.S.J., A.L., G.S.).
ASL Avellino, Italy (P.M., A.P., S.F.).
Fahimeh Varzideh*
Department of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Aging Research, Einstein-Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York City, NY (P.M., F.V., S.S.J., A.L., G.S.).
Stanislovas S. Jankauskas
Department of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Aging Research, Einstein-Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York City, NY (P.M., F.V., S.S.J., A.L., G.S.).
Antonella Pansini
ASL Avellino, Italy (P.M., A.P., S.F.).
Angela Lombardi
Department of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Aging Research, Einstein-Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York City, NY (P.M., F.V., S.S.J., A.L., G.S.).
Salvatore Frullone
ASL Avellino, Italy (P.M., A.P., S.F.).
Department of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Aging Research, Einstein-Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York City, NY (P.M., F.V., S.S.J., A.L., G.S.).
Department of Molecular Pharmacology, Institute for Neuroimmunology and Inflammation (INI), Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York City, NY (G.S.).

Notes

*
P. Mone and F. Varzideh contributed equally.
Supplemental Material is available at Supplemental Material.
For Sources of Funding and Disclosures, see page 1641.
Correspondence to: Gaetano Santulli, Albert Einstein College of Medicine, 1300 Morris Park Ave, 10461 New York, NY, Email [email protected]
Pasquale Mone, Albert Einstein College of Medicine, 1300 Morris Park Ave, 10461 New York, NY, Email [email protected]

Disclosures

Disclosures None.

Sources of Funding

The Santulli Lab is supported, in part, by the National Institutes of Health (NIH; R01-DK123259, R01-DK033823, R01-HL146691, R01-HL159062, and T32-HL144456, to G. Santulli), by the Irma T. Hirschl and Monique Weill-Caulier Trusts (to G. Santulli), by the Diabetes Action Research and Education Foundation (to G. Santulli), and by the American Heart Association (AHA-22POST915561 to F. Varzideh and AHA-21POST836407 to S.S. Jankauskas).

Metrics & Citations

Metrics

Citations

Download Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Select your manager software from the list below and click Download.

  1. Endothelial dysfunction in the kidney transplant population: Current evidence and management strategies, World Journal of Transplantation, 15, 1, (2025).https://doi.org/10.5500/wjt.v15.i1.97458
    Crossref
  2. Elucidating the cardioprotective mechanisms of sodium-glucose cotransporter-2 inhibitors beyond glycemic control, World Journal of Diabetes, 15, 2, (137-141), (2024).https://doi.org/10.4239/wjd.v15.i2.137
    Crossref
  3. Endothelial Dysfunction as a Key Link between Cardiovascular Disease and Frailty: A Systematic Review, Journal of Clinical Medicine, 13, 9, (2686), (2024).https://doi.org/10.3390/jcm13092686
    Crossref
  4. SGLT2 Inhibitors in Kidney Diseases—A Narrative Review, International Journal of Molecular Sciences, 25, 9, (4959), (2024).https://doi.org/10.3390/ijms25094959
    Crossref
  5. Understanding the Role of Oxidative Stress in Platelet Alterations and Thrombosis Risk among Frail Older Adults, Biomedicines, 12, 9, (2004), (2024).https://doi.org/10.3390/biomedicines12092004
    Crossref
  6. Mitochondrial Dysfunction in Cardiac Disease: The Fort Fell, Biomolecules, 14, 12, (1534), (2024).https://doi.org/10.3390/biom14121534
    Crossref
  7. Editorial: Frailty and oxidative stress, Frontiers in Aging, 4, (2024).https://doi.org/10.3389/fragi.2023.1345486
    Crossref
  8. Frailty and Parkinson’s disease: the role of diabetes mellitus, Frontiers in Medicine, 11, (2024).https://doi.org/10.3389/fmed.2024.1377975
    Crossref
  9. Cross-sectional comparison of the association between three different insulin resistance surrogates and frailty: NHANES 1999-2018, Frontiers in Endocrinology, 15, (2024).https://doi.org/10.3389/fendo.2024.1439326
    Crossref
  10. The association between frailty and in-hospital mortality in critically ill patients with congestive heart failure: results from MIMIC-IV database, Frontiers in Cardiovascular Medicine, 11, (2024).https://doi.org/10.3389/fcvm.2024.1361542
    Crossref
  11. See more
Loading...

View Options

View options

PDF and All Supplements

Download PDF and All Supplements

PDF/EPUB

View PDF/EPUB
Login options

Check if you have access through your login credentials or your institution to get full access on this article.

Personal login Institutional Login
Purchase Options

Purchase this article to access the full text.

Purchase access to this article for 24 hours

SGLT2 Inhibition via Empagliflozin Improves Endothelial Function and Reduces Mitochondrial Oxidative Stress: Insights From Frail Hypertensive and Diabetic Patients
Hypertension
  • Vol. 79
  • No. 8

Purchase access to this journal for 24 hours

Hypertension
  • Vol. 79
  • No. 8
Restore your content access

Enter your email address to restore your content access:

Note: This functionality works only for purchases done as a guest. If you already have an account, log in to access the content to which you are entitled.

Media

Figures

Other

Tables

Share

Share

Share article link

Share

Comment Response