1
|
Hong JH, Kim MJ, Min KW, Won JC, Kim TN, Lee B, Kang JG, Kim JH, Park JH, Ku BJ, Lee CB, Kim SY, Shon HS, Lee WJ, Park J. Efficacy and safety of a fixed-dose combination of dapagliflozin and linagliptin (AJU-A51) in patients with type 2 diabetes mellitus: A multicentre, randomized, double-blind, parallel-group, placebo-controlled phase III study. Diabetes Obes Metab 2025; 27:81-91. [PMID: 39375869 PMCID: PMC11618241 DOI: 10.1111/dom.15985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/16/2024] [Accepted: 09/16/2024] [Indexed: 10/09/2024]
Abstract
AIMS To evaluate the efficacy and safety of add-on dapagliflozin in patients with type 2 diabetes mellitus (T2D) who had inadequate glycaemic control with metformin and linagliptin. MATERIALS AND METHODS A total of 235 patients with inadequate response to metformin (≥1000 mg/day) plus linagliptin (5 mg/day) were randomized to receive either dapagliflozin/linagliptin fixed-dose combination (FDC [AJU-A51]) 10/5 mg/day (n = 117) or linagliptin 5 mg plus placebo (n = 118) for 24 weeks. After the main treatment period, patients who received linagliptin plus placebo were treated with AJU-A51 for an additional 28 weeks. Change in glycated haemoglobin (HbA1c) from baseline to Week 24 was the primary endpoint. RESULTS AJU-A51 significantly reduced HbA1c levels (from 7.93% ± 0.82% to 7.11% ± 0.61%) compared with linagliptin plus placebo (from 7.80% ± 0.71% to 7.87% ± 0.94%), with a least squares mean difference of -0.88% (95% confidence interval -1.07 to -0.68; p < 0.0001) at 24 weeks. The AJU-A51 group had a significantly higher proportion of patients who achieved HbA1c <7.0% at Week 24 than the control group (44.8% vs. 18.6%; p < 0.001). The AJU-A51 group maintained glycaemic efficacy up to 52 weeks, whereas the control group showed a substantial reduction in HbA1c after switching to AJU-A51 in the extension study period. Both groups had similar incidence of treatment-emergent and serious adverse events, and no cases of symptomatic hypoglycaemia were reported. CONCLUSIONS Dapagliflozin and linagliptin FDC (AJU-A51) showed potent glucose-lowering effects, with good tolerability, in patients with T2D who had poor glycaemic control on metformin and linagliptin (ClinicalTrials.gov [NCT06329674]).
Collapse
Affiliation(s)
- Jun Hwa Hong
- Department of Internal Medicine, Daejeon Eulji Medical CenterEulji UniversityDaejeonKorea
| | - Myung Jin Kim
- Department of Internal Medicine, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulKorea
- Asan Diabetes CenterAsan Medical CenterSeoulKorea
| | - Kyung Wan Min
- Department of Internal MedicineEulji University School of MedicineSeoulKorea
| | - Jong Chul Won
- Department of Internal Medicine, Sanggye Paik Hospital, Cardiovascular and Metabolic Disease CenterInje University College of MedicineSeoulKorea
| | - Tae Nyun Kim
- Department of Internal Medicine, Cardiovascular and Metabolic Disease CenterInje University College of MedicineBusanKorea
| | - Byung‐Wan Lee
- Division of Endocrinology and Metabolism, Department of Internal MedicineYonsei University College of MedicineSeoulRepublic of Korea
| | - Jun Goo Kang
- Department of Internal MedicineHallym University Sacred Heart HospitalAnyangKorea
| | - Jae Hyeon Kim
- Department of Internal Medicine, Samsung Medical CenterSungkyunkwan University School of MedicineSeoulKorea
| | - Jung Hwan Park
- Department of Internal MedicineHanyang University College of MedicineSeoulKorea
| | - Bon Jeong Ku
- Department of Internal MedicineChungnam National University College of MedicineDaejeonKorea
| | - Chang Beom Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Hanyang University Guri HospitalHanyang University College of MedicineGuriKorea
| | - Sang Yong Kim
- Department of Internal Medicine, Chosun University HospitalChosun University College of MedicineGwangjuKorea
| | - Ho Sang Shon
- Department of Internal MedicineCatholic University of Daegu School of MedicineDaeguKorea
| | - Woo Je Lee
- Department of Internal Medicine, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulKorea
- Asan Diabetes CenterAsan Medical CenterSeoulKorea
| | - Joong‐Yeol Park
- Department of Internal Medicine, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulKorea
- Asan Diabetes CenterAsan Medical CenterSeoulKorea
| |
Collapse
|
2
|
Forouzanmehr B, Hedayati AH, Gholami E, Hemmati MA, Maleki M, Butler AE, Jamialahmadi T, Kesharwani P, Yaribeygi H, Sahebkar A. Sodium-glucose cotransporter 2 inhibitors and renin-angiotensin-aldosterone system, possible cellular interactions and benefits. Cell Signal 2024; 122:111335. [PMID: 39117253 DOI: 10.1016/j.cellsig.2024.111335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 07/30/2024] [Accepted: 08/04/2024] [Indexed: 08/10/2024]
Abstract
Sodium glucose cotransporter 2 inhibitors (SGLT2is) are a newly developed class of anti-diabetics which exert potent hypoglycemic effects in the diabetic milieu. However, the evidence suggests that they also have extra-glycemic effects. The renin-angiotensin-aldosterone system (RAAS) is a hormonal system widely distributed in the body that is important for water and electrolyte homeostasis as well as renal and cardiovascular function. Therefore, modulating RAAS activity is a main goal in patients, notably diabetic patients, which are at higher risk of complications involving these organ systems. Some studies have suggested that SGLT2is have modulatory effects on RAAS activity in addition to their hypoglycemic effects and, thus, these drugs can be considered as promising therapeutic agents for renal and cardiovascular disorders. However, the exact molecular interactions between SGLT2 inhibition and RAAS activity are not clearly understood. Therefore, in the current study we surveyed the literature for possible molecular mechanisms by which SGLT2is modulate RAAS activity.
Collapse
Affiliation(s)
- Behina Forouzanmehr
- Student Research Committee, Semnan University of Medical Sciences, Semnan, Iran
| | | | - Emad Gholami
- Student Research Committee, Semnan University of Medical Sciences, Semnan, Iran
| | | | - Mina Maleki
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alexandra E Butler
- Research Department, Royal College of Surgeons in Ireland Bahrain, Adliya 15503, Bahrain
| | - Tannaz Jamialahmadi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| | - Habib Yaribeygi
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran.
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
3
|
Nagayama A, Inokuchi T, Ashida K, Inada C, Homma T, Miyazaki H, Adachi T, Iwata S, Motomura S, Nomura M. Assessing the Metabolic and Physical Effects of Combined DPP4 and SGLT2 Inhibitor Therapy in Patients with Type-2 Diabetes Mellitus: An Observational Prospective Pilot Study. JMA J 2024; 7:387-400. [PMID: 39114607 PMCID: PMC11301034 DOI: 10.31662/jmaj.2023-0214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 03/06/2024] [Indexed: 08/10/2024] Open
Abstract
Introduction This study aimed to assess the efficacy of combined administration of dipeptidyl peptide-4 (DPP4) and sodium-glucose cotransporter-2 (SGLT2) inhibitors on metabolic disorders and their preferable and complementary effects. Methods The effectiveness of a 24-week intervention on metabolic parameters (including glucose profile), physical functions (grip strength and calf circumference), and health-related quality of life (HR-QOL) was analyzed using the International Physical Activity Questionnaire and Geriatric Depression Scale 5. A total of 39 patients with type-2 diabetes mellitus (T2DM) treated with the combination of DPP4 and SGLT2 inhibitors were included in this multicenter pilot study. Results Combination therapy significantly reduced the HbA1c level (median [interquartile range]) after 24 weeks (pretreatment: 7.7% [7.3-8.2] vs. posttreatment: 7.1% [6.6-7.9], P < 0.001). The grip strength significantly increased after 24 weeks (1.7 ± 2.7 kg, P < 0.001), while the mean calf circumference and body mass index significantly decreased. In particular, administration of the SGLT2 inhibitor significantly increased total physical activity in participants aged ≥65 years (P = 0.003), while psychological QOL did not significantly improve. Conclusions Combination therapy with DPP4 and SGLT2 inhibitors decreased HbA1c levels and improved physical function in patients with T2DM. This study confirmed the effectiveness of combination therapy for metabolic disorders and suggested its beneficial and complementary effects. Therefore, advances in treatment plans to achieve further improvements in glucose profiles using DPP4 and SGLT2 inhibitors are recommended to enhance the QOL of patients with T2DM. Clinical trial number: University Hospital Medical Information Network Center: UMIN000045375.
Collapse
Affiliation(s)
- Ayako Nagayama
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | | | - Kenji Ashida
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | | | - Tomoki Homma
- Homma Clinic of Cardiology and Internal Medicine, Kurume, Japan
| | - Hiroshi Miyazaki
- Miyazaki Clinic of Cardiology and Internal Medicine, Kurume, Japan
| | | | - Shimpei Iwata
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Seiichi Motomura
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Masatoshi Nomura
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| |
Collapse
|
4
|
Matthews JR, Herat LY, Schlaich MP, Matthews VB. Sympathetic Activation Promotes Sodium Glucose Co-Transporter-1 Protein Expression in Rodent Skeletal Muscle. Biomedicines 2024; 12:1456. [PMID: 39062029 PMCID: PMC11275186 DOI: 10.3390/biomedicines12071456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/21/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024] Open
Abstract
The hyperactivation of the sympathetic nervous system (SNS) is linked to obesity, hypertension, and type 2 diabetes, which are characterized by elevated norepinephrine (NE) levels. Previous research has shown increased sodium-dependent glucose cotransporter 1 (SGLT1) protein levels in kidneys of hypertensive rodents, prompting investigation into the expression of SGLT1 in various tissues, such as skeletal muscle. This study aimed to assess (i) whether skeletal muscle cells and tissue express SGLT1 and SGLT2 proteins; (ii) if NE increases SGLT1 levels in skeletal muscle cells, and (iii) whether the skeletal muscle of neurogenically hypertensive mice exhibits increased SGLT1 expression. We found that (i) skeletal muscle cells and tissue are a novel source of the SGLT2 protein and that (ii) NE significantly elevated SGLT1 levels in skeletal muscle cells. As SGLT2 inhibition (SGLT2i) with Empagliflozin increased SGLT1 levels, in vivo studies with the dual inhibitor SGLT1/2i, Sotagliflozin were warranted. The treatment of neurogenically hypertensive mice using Sotagliflozin significantly reduced blood pressure. Our findings suggest that SNS activity upregulates the therapeutic target, SGLT1, in skeletal muscle, potentially worsening cardiometabolic control. As clinical trial data suggest cardiorenal benefits from SGLT2i, future studies should aim to utilize SGLT1i by itself, which may offer a therapeutic strategy for conditions with heightened SNS activity, such as hypertension, diabetes, and obesity.
Collapse
Affiliation(s)
- Jennifer R. Matthews
- Dobney Hypertension Centre, School of Biomedical Sciences—Royal Perth Hospital Unit, University of Western Australia, and Royal Perth Hospital Research Foundation, Crawley, WA 6000, Australia; (J.R.M.); (L.Y.H.)
| | - Lakshini Y. Herat
- Dobney Hypertension Centre, School of Biomedical Sciences—Royal Perth Hospital Unit, University of Western Australia, and Royal Perth Hospital Research Foundation, Crawley, WA 6000, Australia; (J.R.M.); (L.Y.H.)
| | - Markus P. Schlaich
- Dobney Hypertension Centre, School of Medicine—Royal Perth Hospital Unit, University of Western Australia, Perth, WA 6000, Australia;
- Departments of Cardiology and Nephrology, Royal Perth Hospital, Perth, WA 6000, Australia
- Neurovascular Hypertension & Kidney Disease Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Vance B. Matthews
- Dobney Hypertension Centre, School of Biomedical Sciences—Royal Perth Hospital Unit, University of Western Australia, and Royal Perth Hospital Research Foundation, Crawley, WA 6000, Australia; (J.R.M.); (L.Y.H.)
| |
Collapse
|
5
|
Zhang L, Lin H, Yang X, Shi J, Sheng X, Wang L, Li T, Quan H, Zhai X, Li W. Effects of dapagliflozin monotherapy and combined aerobic exercise on skeletal muscle mitochondrial quality control and insulin resistance in type 2 diabetes mellitus rats. Biomed Pharmacother 2023; 169:115852. [PMID: 37944441 DOI: 10.1016/j.biopha.2023.115852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/31/2023] [Accepted: 11/05/2023] [Indexed: 11/12/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a prevalent, chronic metabolic disease. Sodium-glucose cotransporter-2 (SGLT2) inhibitors and aerobic exercise (AE) have shown promise in mitigating insulin resistance (IR) and T2DM. This study investigated the effects of dapagliflozin (Dapa) monotherapy and combined AE on mitochondrial quality control (MQC) in skeletal muscle and IR in T2DM rats. T2DM rats, induced by a high-fat diet/streptozotocin model, were randomly assigned to the following groups: T2DM+vehicle group (DMV), T2DM rats treated with Dapa (DMDa, 10 mg/kg/d), T2DM rats subjected to combined Dapa treatment and AE (DMDa+AE), and the standard control group (CON). Blood and skeletal muscle samples were collected after 6 weeks of intragastric administration and treadmill exercise. The results showed that DMDa monotherapy could reduce the accumulation of white adipose tissue and skeletal muscle lipid droplets and improve HOMA-IR. While the combined AE led to further reductions in subcutaneous white adipose tissue and fasting glucose levels, it did not confer additional benefits in terms of HOMA-IR. Furthermore, Dapa monotherapy enhanced skeletal muscle mitochondrial biogenesis (PGC-1α, NRF1, TFAM, and COX IV), mitochondrial dynamics (OPA1, DRP1, and MFN2), and mitophagy (PGAM5 and PINK1) related protein levels. Nevertheless, the combination of Dapa with AE treatment did not yield an additive effect. In conclusion, this study highlights the potential of SGLT2 inhibitors, specifically Dapa, in ameliorating IR and maintaining MQC in skeletal muscle in rats with T2DM. However, combined AE did not produce an additive effect, indicating the need for further research.
Collapse
Affiliation(s)
- Liangzhi Zhang
- Exercise and Metabolism Research Center, College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, Zhejiang, China
| | - Hengjun Lin
- Department of Colorectal anal Surgery, Jinhua People's Hospital, Jinhua, Zhejiang, China
| | - Xudong Yang
- Exercise and Metabolism Research Center, College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, Zhejiang, China
| | - Jipeng Shi
- Exercise Capacity Assessment and Promotion Research Center, School of Physical Education, Northeast Normal University, Changchun, Jilin, China
| | - Xiusheng Sheng
- Medical Molecular Biology Laboratory, School of Medicine, Jinhua Polytechnic, Jinhua, Zhejiang, China
| | - Lifeng Wang
- Exercise and Metabolism Research Center, College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, Zhejiang, China
| | - Ting Li
- Exercise and Metabolism Research Center, College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, Zhejiang, China
| | - Helong Quan
- Exercise Capacity Assessment and Promotion Research Center, School of Physical Education, Northeast Normal University, Changchun, Jilin, China.
| | - Xia Zhai
- Medical Molecular Biology Laboratory, School of Medicine, Jinhua Polytechnic, Jinhua, Zhejiang, China.
| | - Wei Li
- Exercise and Metabolism Research Center, College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, Zhejiang, China.
| |
Collapse
|
6
|
Afsar B, Afsar RE. Sodium-glucose co-transporter 2 inhibitors and Sarcopenia: A controversy that must be solved. Clin Nutr 2023; 42:2338-2352. [PMID: 37862820 DOI: 10.1016/j.clnu.2023.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/23/2023] [Accepted: 10/02/2023] [Indexed: 10/22/2023]
Abstract
Diabetes mellitus is a risk factor for muscle loss and sarcopenia. Sodium-glucose co-transporter 2 inhibitors (SGLT2i) or "gliflozins" are one of the newest anti-hyperglycemic drugs. They reduce blood glucose levels by inhibiting renal glucose reabsorption in the early proximal convoluted tubule. Various randomized trials showed that SGLT2i have cardio-protective and reno-protective action. SGLT2i also affect body composition. They usually decrease body fat percentage, visceral and subcutaneous adipose tissue. However, regarding the muscle mass, there are conflicting findings some studies showing detrimental effects and others showed neutral or beneficial effects. This issue is extremely important not only because of the wide use of SGLT2i around globe; but also skeletal muscle mass consumes large amounts of calories during exercise and is an important determinant of resting metabolic rate and skeletal muscle loss hinders energy consumption leading to obesity. In this systematic review, we extensively reviewed the experimental and clinical studies regarding the impact of SGLT2i on muscle mass and related metabolic alterations. Importantly, studies are heterogeneous and there is unmet need to highlight the alterations in muscle during SGLT2i use.
Collapse
Affiliation(s)
- Baris Afsar
- Suleyman Demirel University, School of Medicine, Department of Nephrology, Isparta, Turkey.
| | - Rengin Elsurer Afsar
- Suleyman Demirel University, School of Medicine, Department of Nephrology, Isparta, Turkey
| |
Collapse
|
7
|
Chandrasekar B, Mummidi S, DeMarco VG, Higashi Y. Empagliflozin Reverses Oxidized LDL-Induced RECK Suppression, Cardiotrophin-1 Expression, MMP Activation, and Human Aortic Smooth Muscle Cell Proliferation and Migration. Mediators Inflamm 2023; 2023:6112301. [PMID: 37830075 PMCID: PMC10567511 DOI: 10.1155/2023/6112301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 09/04/2023] [Accepted: 09/07/2023] [Indexed: 10/14/2023] Open
Abstract
Persistent oxidative stress and inflammation contribute causally to smooth muscle cell (SMC) proliferation and migration, the characteristic features of vascular proliferative diseases. Oxidatively modified low-density lipoproteins (OxLDL) elevate oxidative stress levels, inflammatory responses, and matrix metallopeptidase (MMP) activation, resulting ultimately in SMC migration, proliferation, and phenotype change. Reversion-inducing cysteine-rich protein with Kazal motifs (RECK) is a membrane-anchored MMP inhibitor. Empagliflozin is an SGLT2 inhibitor and exerts pleiotropic cardiovascular protective effects, including antioxidant and anti-inflammatory effects. Here, we investigated (i) whether OxLDL regulates RECK expression, (ii) whether ectopic expression of RECK reverses OxLDL-induced SMC migration and proliferation, and (iii) whether pretreatment with empagliflozin reverses OxLDL-induced RECK suppression, MMP activation, and SMC migration, proliferation, and differentiation. Indeed, results show that OxLDL at pathophysiological concentration promotes SMC migration and proliferation via NF-κB/miR-30b-dependent RECK suppression. Moreover, OxLDL changed the SMC phenotype to a more pro-inflammatory type, and this effect is blunted by RECK overexpression. Further, treatment with empagliflozin reversed OxLDL-induced miR-30b induction, RECK suppression, MMP activation, SMC migration, proliferation, and proinflammatory phenotype changes. OxLDL-induced cardiotrophin (CT)-1 expression and CT-1 stimulated SMC proliferation and migration in part via leukemia inhibitory factor receptor (LIFR) and glycoprotein 130 (gp130). Ectopic expression of RECK inhibited these effects by physically associating with LIFR and gp130, as evidenced by immunoprecipitation/immunoblotting and double immunofluorescence. Importantly, empagliflozin inhibited CT-1-induced mitogenic and migratory effects. Together, these results suggest the therapeutic potential of sustaining RECK expression or empagliflozin in vascular diseases characterized by SMC proliferation and migration.
Collapse
Affiliation(s)
- Bysani Chandrasekar
- Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA
- Medicine, University of Missouri School of Medicine, Columbia, MO, USA
- Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
- Dalton Cardiovascular Center, University of Missouri, Columbia, MO, USA
| | - Srinivas Mummidi
- Life Sciences, Texas A&M University-San Antonio, San Antonio, TX, USA
| | - Vincent G. DeMarco
- Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA
- Medicine, University of Missouri School of Medicine, Columbia, MO, USA
- Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| | - Yusuke Higashi
- Medicine/Cardiology, Tulane University School of Medicine, New Orleans, LA, USA
| |
Collapse
|
8
|
Yang X, Wang L, Zhang L, Zhai X, Sheng X, Quan H, Lin H. Exercise mitigates Dapagliflozin-induced skeletal muscle atrophy in STZ-induced diabetic rats. Diabetol Metab Syndr 2023; 15:154. [PMID: 37438792 PMCID: PMC10337193 DOI: 10.1186/s13098-023-01130-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 07/01/2023] [Indexed: 07/14/2023] Open
Abstract
BACKGROUND Sodium-glucose cotransporter 2 inhibitors (SGLT2i) are commonly used in the management of type 2 diabetes mellitus (T2DM) and have been found to worsen the reduction of skeletal muscle mass in individuals with T2DM. This study aims to examine the potential of exercise in mitigating the skeletal muscle atrophy induced by SGLT2i treatment. METHODS A rat model of T2DM (40 male Sprague-Dawley rats; T2DM induced by a combination of high-fat diet and streptozotocin) was used to examine the effects of six-week treatment with Dapagliflozin (DAPA, SGLT2i) in combination with either aerobic exercise (AE) or resistance training (RT) on skeletal muscle. T2DM-eligible rats were randomized into the T2DM control group (CON, n = 6), DAPA treatment group (DAPA, n = 6), DAPA combined with aerobic exercise intervention group (DAPA + AE, n = 6), and DAPA combined with resistance training intervention group (DAPA + RT, n = 6). To assess the morphological changes in skeletal muscle, myosin ATPase and HE staining were performed. mRNA expression levels of Atrogin-1, MuRF1, and Myostatin were determined using quantitative PCR. Furthermore, protein expression levels of AKT, p70S6K, mTOR, FoXO1/3A, NF-κB, and MuRF1 were examined through western blotting. RESULTS Both the administration of DAPA alone and the combined exercise intervention with DAPA resulted in significant reductions in blood glucose levels and body weight in rats. However, DAPA alone administration led to a decrease in skeletal muscle mass, whereas RT significantly increased skeletal muscle mass and muscle fiber cross-sectional area. The DAPA + RT group exhibited notable increases in both total protein levels and phosphorylation levels of AKT and p70S6K in skeletal muscle. Moreover, the DAPA, DAPA + AE, and DAPA + RT groups demonstrated downregulation of protein expression (FoXO1/3A) and mRNA levels (Atrogin-1, MuRF1, and Myostatin) associated with muscle atrophy. CONCLUSIONS Our findings provide support for the notion that dapagliflozin may induce skeletal muscle atrophy through mechanisms unrelated to protein metabolism impairment in skeletal muscle, as it does not hinder protein metabolic pathways while reduces muscle atrophy-related genes. Additionally, our observations reveal that RT proves more effective than AE in enhancing skeletal muscle mass and muscle fiber cross-sectional area in rats with T2DM by stimulating protein anabolism within the skeletal muscle.
Collapse
Affiliation(s)
- Xudong Yang
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, Zhejiang, China
- Exercise and Metabolism Research Center, Zhejiang Normal University, Jinhua, Zhejiang, China
| | - Lifeng Wang
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, Zhejiang, China
- Exercise and Metabolism Research Center, Zhejiang Normal University, Jinhua, Zhejiang, China
| | - Liangzhi Zhang
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, Zhejiang, China
- Exercise and Metabolism Research Center, Zhejiang Normal University, Jinhua, Zhejiang, China
| | - Xia Zhai
- Medical Molecular Biology Laboratory, School of Medicine, Jinhua Polytechnic, Jinhua, Zhejiang, China
| | - Xiusheng Sheng
- Medical Molecular Biology Laboratory, School of Medicine, Jinhua Polytechnic, Jinhua, Zhejiang, China
| | - Helong Quan
- Exercise and Metabolism Research Center, Zhejiang Normal University, Jinhua, Zhejiang, China.
- School of Sports Science and Physical Education, Research Center of Sports and Health Science, Northeast Normal University, 5268 Renmin Street, Changchun, Jilin, 130024, China.
| | - Hengjun Lin
- Department of Colorectal anal surgery, Jinhua people's hospital, 267 Danxi East Road, Jinhua, Zhejiang, 321007, China.
| |
Collapse
|
9
|
De La Flor JC, Villa D, Cruzado L, Apaza J, Valga F, Zamora R, Marschall A, Cieza M, Deira J, Rodeles M. Efficacy and Safety of the Use of SGLT2 Inhibitors in Patients on Incremental Hemodialysis: Maximizing Residual Renal Function, Is There a Role for SGLT2 Inhibitors? Biomedicines 2023; 11:1908. [PMID: 37509547 PMCID: PMC10377393 DOI: 10.3390/biomedicines11071908] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/11/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023] Open
Abstract
SGLT-2i are the new standard of care for diabetic kidney disease (DKD), but previous studies have not included patients on kidney replacement therapy (KRT). Due to their high risk of cardiovascular, renal complications, and mortality, these patients would benefit the most from this therapy. Residual kidney function (RKF) conveys a survival benefit and cardiovascular health among hemodialysis (HD) patients, especially those on incremental hemodialysis (iHD). We retrospectively describe the safety and efficacy of SGLT2i regarding RKF preservation in seven diabetic patients with different clinical backgrounds who underwent iHD (one or two sessions per week) during a 12-month follow-up. All patients preserved RKF, measured as residual kidney urea clearance (KrU) in 24 h after the introduction of SGLT2i. KrU levels improved significantly from 4.91 ± 1.14 mL/min to 7.28 ± 1.68 mL/min at 12 months (p = 0.028). Pre-hemodialysis blood pressure improved 9.95% in mean systolic blood pressure (SBP) (p = 0.015) and 10.95% in mean diastolic blood pressure (DBP) (p = 0.041); as a result, antihypertensive medication was modified. Improvements in blood uric acid, hemoglobin A1c, urine albumin/creatinine ratio (UACR), and 24 h proteinuria were also significant. Regarding side effects, two patients developed uncomplicated urinary tract infections that were resolved. No other complications were reported. The use of SGLT2i in our sample of DKD patients starting iHD on a 1-2 weekly regimen appears to be safe and effective in preserving RKF.
Collapse
Affiliation(s)
- José C De La Flor
- Department of Nephrology, Hospital Central Defense Gomez Ulla, 28047 Madrid, Spain
| | - Daniel Villa
- Department of Nephrology, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Leónidas Cruzado
- Department of Nephrology, Hospital General Elche, 03203 Elche, Spain
| | - Jacqueline Apaza
- Department of Nephrology, Hospital Fuensanta, 28942 Madrid, Spain
| | - Francisco Valga
- Department of Nephrology, Hospital Universitario Doctor Negrin de Gran Canarias, 35016 Las Palmas de Gran Canarias, Spain
| | - Rocío Zamora
- Department of Nephrology, Hospital Universitario General Villalba, 28400 Madrid, Spain
| | - Alexander Marschall
- Department of Cardiology, Central Defense Gomez Ulla Hospital, 28047 Madrid, Spain
| | - Michael Cieza
- Teaching Coordination Unit, Universidad Peruana Cayetano Heredia, Lima 15012, Peru
| | - Javier Deira
- Department of Nephrology, Hospital San Pedro de Alcántara, 10003 Cáceres, Spain
| | - Miguel Rodeles
- Department of Nephrology, Hospital Central Defense Gomez Ulla, 28047 Madrid, Spain
| |
Collapse
|
10
|
Quiroga B, Díez J. Estimation of glomerular filtration rate in cardiorenal patients: a step forward. Clin Kidney J 2023; 16:1049-1055. [PMID: 37398687 PMCID: PMC10310511 DOI: 10.1093/ckj/sfad083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Indexed: 07/04/2023] Open
Abstract
The progressive reduction in estimated glomerular filtration rate (eGFR) resulting in chronic kidney disease (CKD) is associated with increased risk of cardiovascular disease (CVD) (i.e., cardiorenal disease). Cardiorenal disease is associated with poor outcomes, mainly due to increased cardiovascular (CV) complications and CV death. Data from general population-based studies and studies of cohorts with CKD and/or CVD show that compared with creatinine-based eGFR, cystatin C-based eGFR and creatinine plus cystatin C-based eGFR detect higher risks of adverse CV outcomes and add predictive discrimination to current CVD risk scores. On the other hand, growing clinical evidence supports kidney and CV protective effects of sodium-glucose cotransporter-2 (SGLT2) inhibitors in cardiorenal patients. However, recent data suggest that some detrimental effects of SGLT2 inhibitors on skeletal muscle mass may lead to overestimation of creatinine-based eGFR and subsequent misinterpretation of associated CV risk in patients treated with these agents. Within this framework, we suggest the advisability of using cystatin C and/or creatinine plus cystatin C-based eGFR for routine clinical practice in cardiorenal patients to more accurately stratify CV risk and evaluate the kidney and CV protective effects of SGLT2 inhibitors. In this regard, we make a call to action to investigate the protective effects of these pharmacological agents using cystatin C-based eGFR.
Collapse
Affiliation(s)
| | - Javier Díez
- Working Group on Cardiorenal Medicine (CaReSEN), Sociedad Española de Nefrología, Madrid, Spain
- Center of Applied Medical Research and School of Medicine, University of Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de la Enfermedades Cardiovasculares (CIBERCV), Carlos III Institute of Health, Madrid, Spain
| |
Collapse
|
11
|
Stöllberger C, Finsterer J, Schneider B. Adverse events and drug-drug interactions of sodium glucose co-transporter 2 inhibitors in patients treated for heart failure. Expert Rev Cardiovasc Ther 2023; 21:803-816. [PMID: 37856368 DOI: 10.1080/14779072.2023.2273900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 10/18/2023] [Indexed: 10/21/2023]
Abstract
INTRODUCTION Sodium glucose co-transporter 2-inhibitors (SGLT2-I), antihyperglycemic agents, are increasingly prescribed in chronic heart failure (CHF). Their risk for drug-drug interactions (DDI) seems low. Safety-data derive mainly from diabetes-patients. This review aims to summarize adverse-events (AE) and DDI of the SGLT2-I dapagliflozin, empagliflozin and sotagliflozin in patients with CHF. AREAS COVERED Literature-search-terms in PubMed were 'adverse event/drug-drug interaction' and 'heart failure AND 'dapagliflozin' OR 'empagliflozin' OR 'sotagliflozin.'AEreported in randomized controlled trials (RCT) comprisegenitaland urinary-tract infections, hypotension, ketoacidosis, renal impairment, hypoglycemia, limb-amputations, Fournier's gangrene, bone-fractures, hepatopathy, pancreatitis, diarrhea, malignancy and venous thromboembolism. Their incidence is largely unknown, since they were not consistently evaluated in RCT of CHF. Further AE from meta-analyses, pharmacovigilance reports, case-series and case-reports include erythrocytosis, hypertriglyceridemia, myopathy, sarcopenia, skin problems, ventricular tachycardia, and urinary retention. The maximal observation period of RCT in CHF was 26 months.DDI were mainly studied in healthy volunteers for 3-8 days. In CHF or diabetes-patients, DDI were reported with interleukin-17-inhibitors, linezolid, lithium, tacrolimus, valproate, angiotensin-receptor-neprilysin-inhibitors and intravenous iron. EXPERT OPINION Guidelines recommend treatment with SGLT2-I for CHF but no data on AE during long-term therapy and only little information on DDI are available, which stresses the need for further research. Evidence-based recommendations for ketoacidosis-prevention are desirable.
Collapse
|
12
|
Legaard GE, Lyngbæk MPP, Almdal TP, Karstoft K, Bennetsen SL, Feineis CS, Nielsen NS, Durrer CG, Liebetrau B, Nystrup U, Østergaard M, Thomsen K, Trinh B, Solomon TPJ, Van Hall G, Brønd JC, Holst JJ, Hartmann B, Christensen R, Pedersen BK, Ried-Larsen M. Effects of different doses of exercise and diet-induced weight loss on beta-cell function in type 2 diabetes (DOSE-EX): a randomized clinical trial. Nat Metab 2023; 5:880-895. [PMID: 37127822 PMCID: PMC10229430 DOI: 10.1038/s42255-023-00799-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 04/04/2023] [Indexed: 05/03/2023]
Abstract
Diet-induced weight loss is associated with improved beta-cell function in people with type 2 diabetes (T2D) with remaining secretory capacity. It is unknown if adding exercise to diet-induced weight loss improves beta-cell function and if exercise volume is important for improving beta-cell function in this context. Here, we carried out a four-armed randomized trial with a total of 82 persons (35% females, mean age (s.d.) of 58.2 years (9.8)) with newly diagnosed T2D (<7 years). Participants were randomly allocated to standard care (n = 20), calorie restriction (25% energy reduction; n = 21), calorie restriction and exercise three times per week (n = 20), or calorie restriction and exercise six times per week (n = 21) for 16 weeks. The primary outcome was beta-cell function as indicated by the late-phase disposition index (insulin secretion multiplied by insulin sensitivity) at steady-state hyperglycemia during a hyperglycemic clamp. Secondary outcomes included glucose-stimulated insulin secretion and sensitivity as well as the disposition, insulin sensitivity, and secretion indices derived from a liquid mixed meal tolerance test. We show that the late-phase disposition index during the clamp increases more in all three intervention groups than in standard care (diet control group, 58%; 95% confidence interval (CI), 16 to 116; moderate exercise dose group, 105%; 95% CI, 49 to 182; high exercise dose group, 137%; 95% CI, 73 to 225) and follows a linear dose-response relationship (P > 0.001 for trend). We report three serious adverse events (two in the control group and one in the diet control group), as well as adverse events in two participants in the diet control group, and five participants each in the moderate and high exercise dose groups. Overall, adding an exercise intervention to diet-induced weight loss improves glucose-stimulated beta-cell function in people with newly diagnosed T2D in an exercise dose-dependent manner (NCT03769883).
Collapse
Affiliation(s)
- Grit E Legaard
- Centre for Physical Activity Research, Rigshospitalet, Copenhagen, Denmark
| | - Mark P P Lyngbæk
- Centre for Physical Activity Research, Rigshospitalet, Copenhagen, Denmark
| | - Thomas P Almdal
- Department of Endocrinology PE, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Department of Immunology & Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Kristian Karstoft
- Centre for Physical Activity Research, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Pharmacology, Bispebjerg-Frederiksberg Hospital, University of Copenhagen, Copenhagen, Denmark
| | | | - Camilla S Feineis
- Centre for Physical Activity Research, Rigshospitalet, Copenhagen, Denmark
| | - Nina S Nielsen
- Centre for Physical Activity Research, Rigshospitalet, Copenhagen, Denmark
| | - Cody G Durrer
- Centre for Physical Activity Research, Rigshospitalet, Copenhagen, Denmark
| | | | - Ulrikke Nystrup
- Centre for Physical Activity Research, Rigshospitalet, Copenhagen, Denmark
| | - Martin Østergaard
- Centre for Physical Activity Research, Rigshospitalet, Copenhagen, Denmark
| | - Katja Thomsen
- Centre for Physical Activity Research, Rigshospitalet, Copenhagen, Denmark
| | - Beckey Trinh
- Centre for Physical Activity Research, Rigshospitalet, Copenhagen, Denmark
| | | | - Gerrit Van Hall
- Biomedical Sciences, Faculty of Health & Medical Science, University of Copenhagen, Rigshospitalet, Copenhagen, Denmark
- Clinical Metabolomics Core Facility, Clinical Biochemistry, University of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Jan Christian Brønd
- Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences and the Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Bolette Hartmann
- Department of Biomedical Sciences and the Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Robin Christensen
- Section for Biostatistics and Evidence-Based Research, the Parker Institute, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
- Research Unit of Rheumatology, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Bente K Pedersen
- Centre for Physical Activity Research, Rigshospitalet, Copenhagen, Denmark
| | - Mathias Ried-Larsen
- Centre for Physical Activity Research, Rigshospitalet, Copenhagen, Denmark.
- Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
13
|
Sun H, Wang Z, Wang Y, Rong H, Wang D, Liu X, Jin K, Sun Z, Fan Q. Bibliometric and visualized analysis of sodium-Glucose cotransporter 2 inhibitors. Front Pharmacol 2023; 13:1009025. [PMID: 36686683 PMCID: PMC9846544 DOI: 10.3389/fphar.2022.1009025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 12/13/2022] [Indexed: 01/06/2023] Open
Abstract
Background: Sodium-glucose cotransporter 2 inhibitors have proved to be extremely effective and reliable in reducing hyperglycemia, and have also been used for the treatment of cardiovascular and renal disease in patients with or without type 2 diabetes. Thousands of research articles on SGLT2 inhibitors have been published in the past, but few bibliometric analyses have systematically been studied this field. We aimed to visualize the global research hotspots and trends of SGLT2 inhibitors using a bibliometric analysis to provide new evidence and ideas for researchers and clinicians in this field. Methods: We retrieved publications from Science Citation Index Expanded of Web of Science Core Collection in 2004-2022 on 1 July 2022. Microsoft Excel, CiteSpace and VOSviewer were employed to collect publication data, analyze publication trends, and visualize relevant results. Results: We identified 4,419 original research articles on SGLT2 inhibitors published between 2004 and the first half of 2022. Global SGLT2 inhibitors-related research increased rapidly from 2004 to 2022, especially recently. United States made the greatest contribution to the topic, with (1,629, 36.86%) publications and citations (88,892). AstraZeneca was the most prolific institutions (272, 6.16%). Heerspink HJL published the most related articles (98), whereas Zinman B was cited the most frequently (1,784 citations). Diabetes Obesity and Metabolism was the journal with the most studies (406, 9.19%), and The New England Journal of Medicine was the most commonly cited journal (11,617 citations), with nine of the top 10 co-cited references published in this journal. The emerging keywords "heart failure," "diabetic cardiomyopathy," "ejection fraction," "mortality," "biomarker," "fibrosis," "ampk," and "guideline" appeared the most recently as research frontiers. Conclusion: United States is the leader in SGLT2 inhibitor research. Recently, the research on SGLT2 inhibitors has focused on clinical trials, related mechanisms, and therapy. In the future, the research on SGLT2 inhibitors will delve into molecular mechanisms, especially those related to fibrosis and AMPK, revealing the link between SGLT2 inhibitors and heart failure and diabetic cardiomyopathy will be the next research hotspot.
Collapse
Affiliation(s)
- He Sun
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, China,Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhongqing Wang
- Department of Information Center, The First Hospital of China Medical University, Shenyang, China
| | - Yuxi Wang
- Department of Clinical Medicine, China Medical University, Shenyang, China
| | - Haichuan Rong
- Department of Clinical Medicine, China Medical University, Shenyang, China
| | - Danyang Wang
- Department of Clinical Medicine, China Medical University, Shenyang, China
| | - Xiangnian Liu
- Department of Clinical Medicine, China Medical University, Shenyang, China
| | - Ke Jin
- Department of Clinical Medicine, China Medical University, Shenyang, China
| | - Zhicheng Sun
- Department of Clinical Medicine, China Medical University, Shenyang, China
| | - Qiuling Fan
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, China,Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shenyang, China,*Correspondence: Qiuling Fan,
| |
Collapse
|
14
|
Beneficial effects of SGLT2 inhibitor on metabolic inflexibility and visceral fat amount in animal model of obese type 2 diabetes. Heliyon 2022; 8:e11012. [PMID: 36281369 PMCID: PMC9587290 DOI: 10.1016/j.heliyon.2022.e11012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/24/2022] [Accepted: 10/05/2022] [Indexed: 11/05/2022] Open
Abstract
Background Obesity and type 2 diabetes mellitus (T2DM) are often accompanied with a disrupted diurnal rhythm of eating and sustained anabolic state, leading to metabolic inflexibility. In the present study, we plan to investigate effects of a sodium glucose co-transporter 2 (SGLT2) inhibitor, canagliflozin (CANA), on such a metabolic inflexibility, especially on fat metabolism, in the obese type 2 diabetic rats. Materials and methods Five-week-old male SDT (Spontaneously Diabetic Torii) fatty rats as a model of obesity and T2DM and Sprague-Dawley (SD) rats were treated by either CANA (10 mg/kg) or saline (vehicle) orally for 14 days. Then, after the measurement of respiratory quotient (RQ) and visceral and subcutaneous fat volumes, rats were euthanized and blood and tissue samples were collected. Results The treatment by CANA significantly enhanced β-ketone concentration in the blood during light period in the SDT fatty rats with no effect on blood glucose concentration. The CANA treatment significantly reduced visceral fat volume in the SDT fatty rats. A diurnal rhythm of RQ was severely disrupted and persistently high throughout the day in the vehicle-treated SDT fatty rats. By the administration of CANA clearly restored the disrupted diurnal rhythm of RQ with a revival of the nadir during light period. Quantitative real-time RT-PCR revealed a significant increase of AMP-activated protein kinase and decrease of acetyl-CoA carboxylase-1 expression in the liver, and a significant increase of hormone sensitive lipase and uncoupling protein-2 expression in the white adipose tissue by the treatment of CANA in the SDT fatty rats. Conclusion CANA as a SGLT2i reduced visceral fat amount via the enhancement of fat oxidation during the light period, leading to an amelioration of metabolic inflexibility in an obese diabetic model. A novel mechanism of CANA prompts the possibility that this new class of anti-diabetic agent could be a promising anti-obesity agent as well. SGLT2i reduced visceral fat independent of its effect on urinary glucose excretion. SGLT2i increased blood ketone concentration during light period. Concomitantly, RQ during light period was decreased, reviving a diurnal RQ rhythm. Changes in relevant molecules were observed towards fat oxidation. SGLT2i effectively corrected metabolic inflexibility in an obese diabetic model.
Collapse
|
15
|
Voorrips SN, Saucedo-Orozco H, Sánchez-Aguilera PI, De Boer RA, Van der Meer P, Westenbrink BD. Could SGLT2 Inhibitors Improve Exercise Intolerance in Chronic Heart Failure? Int J Mol Sci 2022; 23:8631. [PMID: 35955784 PMCID: PMC9369142 DOI: 10.3390/ijms23158631] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 12/04/2022] Open
Abstract
Despite the constant improvement of therapeutical options, heart failure (HF) remains associated with high mortality and morbidity. While new developments in guideline-recommended therapies can prolong survival and postpone HF hospitalizations, impaired exercise capacity remains one of the most debilitating symptoms of HF. Exercise intolerance in HF is multifactorial in origin, as the underlying cardiovascular pathology and reactive changes in skeletal muscle composition and metabolism both contribute. Recently, sodium-related glucose transporter 2 (SGLT2) inhibitors were found to improve cardiovascular outcomes significantly. Whilst much effort has been devoted to untangling the mechanisms responsible for these cardiovascular benefits of SGLT2 inhibitors, little is known about the effect of SGLT2 inhibitors on exercise performance in HF. This review provides an overview of the pathophysiological mechanisms that are responsible for exercise intolerance in HF, elaborates on the potential SGLT2-inhibitor-mediated effects on these phenomena, and provides an up-to-date overview of existing studies on the effect of SGLT2 inhibitors on clinical outcome parameters that are relevant to the assessment of exercise capacity. Finally, current gaps in the evidence and potential future perspectives on the effects of SGLT2 inhibitors on exercise intolerance in chronic HF are discussed.
Collapse
Affiliation(s)
- Suzanne N. Voorrips
- Department of Cardiology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (H.S.-O.); (P.I.S.-A.); (R.A.D.B.); (P.V.d.M.)
| | | | | | | | | | - B. Daan Westenbrink
- Department of Cardiology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (H.S.-O.); (P.I.S.-A.); (R.A.D.B.); (P.V.d.M.)
| |
Collapse
|
16
|
Giannattasio S, Citarella A, Trocchianesi S, Filardi T, Morano S, Lenzi A, Ferretti E, Crescioli C. Cell-Target-Specific Anti-Inflammatory Effect of Empagliflozin: In Vitro Evidence in Human Cardiomyocytes. Front Mol Biosci 2022; 9:879522. [PMID: 35712355 PMCID: PMC9194473 DOI: 10.3389/fmolb.2022.879522] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 04/11/2022] [Indexed: 12/21/2022] Open
Abstract
The antidiabetic sodium–glucose cotransporter type 2 inhibitor (SGLT2i) empagliflozin efficiently reduces heart failure (HF) hospitalization and cardiovascular death in type 2 diabetes (T2D). Empagliflozin-cardioprotection likely includes anti-inflammatory effects, regardless glucose lowering, but the underlying mechanisms remain unclear. Inflammation is a primary event in diabetic cardiomyopathy (DCM) and HF development. The interferon (IFN)γ-induced 10-kDa protein (IP-10/CXCL10), a T helper 1 (Th1)-type chemokine, promotes cardiac inflammation, fibrosis, and diseases, including DCM, ideally representing a therapeutic target. This preliminary study aims to explore whether empagliflozin directly affects Th1-challenged human cardiomyocytes, in terms of CXCL10 targeting. To this purpose, empagliflozin dose–response curves were performed in cultured human cardiomyocytes maintained within a Th1-dominant inflammatory microenvironment (IFNγ/TNFα), and CXCL10 release with the intracellular IFNγ-dependent signaling pathway (Stat-1) was investigated. To verify possible drug–cell-target specificity, the same assays were run in human skeletal muscle cells. Empagliflozin dose dependently inhibited CXCL10 secretion (IC50 = 76,14 × 10-9 M) in association with Stat-1 pathway impairment only in Th1-induced human cardiomyocytes, suggesting drug-selective cell-type-targeting. As CXCL10 plays multifaceted functions in cardiac remodeling toward HF and currently there is no effective method to prevent it, these preliminary data might be hypothesis generating to open new scenarios in the translational approach to SGLT2i-dependent cardioprotection.
Collapse
Affiliation(s)
- Silvia Giannattasio
- Laboratory of Endocrine Research, Department of Movement, Human and Health Sciences, Section of Health Sciences, University of Rome “Foro Italico”, Rome, Italy
- Laboratory of Nutrigenetic and Nutrigenomic, Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Anna Citarella
- Laboratory of Oncogemics, Department of Experimental Medicine, “Sapienza” University of Rome, Rome, Italy
| | - Sofia Trocchianesi
- Laboratory of Molecular Medicine “Alberto Gulino” Group, Department of Molecular Medicine, “Sapienza” University of Rome, Rome, Italy
| | - Tiziana Filardi
- Laboratory of Oncogemics, Department of Experimental Medicine, “Sapienza” University of Rome, Rome, Italy
| | - Susanna Morano
- Laboratory of Oncogemics, Department of Experimental Medicine, “Sapienza” University of Rome, Rome, Italy
| | - Andrea Lenzi
- Laboratory of Oncogemics, Department of Experimental Medicine, “Sapienza” University of Rome, Rome, Italy
| | - Elisabetta Ferretti
- Laboratory of Oncogemics, Department of Experimental Medicine, “Sapienza” University of Rome, Rome, Italy
- *Correspondence: Elisabetta Ferretti, ; Clara Crescioli,
| | - Clara Crescioli
- Laboratory of Endocrine Research, Department of Movement, Human and Health Sciences, Section of Health Sciences, University of Rome “Foro Italico”, Rome, Italy
- *Correspondence: Elisabetta Ferretti, ; Clara Crescioli,
| |
Collapse
|
17
|
Hoong CWS, Chua MWJ. SGLT2 Inhibitors as Calorie Restriction Mimetics: Insights on Longevity Pathways and Age-Related Diseases. Endocrinology 2021; 162:6226811. [PMID: 33857309 DOI: 10.1210/endocr/bqab079] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Indexed: 02/08/2023]
Abstract
Sodium-glucose cotransporter-2 (SGLT2) inhibitors induce glycosuria, reduce insulin levels, and promote fatty acid oxidation and ketogenesis. By promoting a nutrient deprivation state, SGLT2 inhibitors upregulate the energy deprivation sensors AMPK and SIRT1, inhibit the nutrient sensors mTOR and insulin/IGF1, and modulate the closely linked hypoxia-inducible factor (HIF)-2α/HIF-1α pathways. Phosphorylation of AMPK and upregulation of adiponectin and PPAR-α favor a reversal of the metabolic syndrome which have been linked to suppression of chronic inflammation. Downregulation of insulin/IGF1 pathways and mTOR signaling from a reduction in glucose and circulating amino acids promote cellular repair mechanisms, including autophagy and proteostasis which confer cellular stress resistance and attenuate cellular senescence. SIRT1, another energy sensor activated by NAD+ in nutrient-deficient states, is reciprocally activated by AMPK, and can deacetylate and activate transcription factors, such as PCG-1α, mitochondrial transcription factor A (TFAM), and nuclear factor E2-related factor (NRF)-2, that regulate mitochondrial biogenesis. FOXO3 transcription factor which target genes in stress resistance, is also activated by AMPK and SIRT1. Modulation of these pathways by SGLT2 inhibitors have been shown to alleviate metabolic diseases, attenuate vascular inflammation and arterial stiffness, improve mitochondrial function and reduce oxidative stress-induced tissue damage. Compared with other calorie restriction mimetics such as metformin, rapamycin, resveratrol, and NAD+ precursors, SGLT2 inhibitors appear to be the most promising in the treatment of aging-related diseases, due to their regulation of multiple longevity pathways that closely resembles that achieved by calorie restriction and their established efficacy in reducing cardiovascular events and all-cause mortality. Evidence is compelling for the role of SGLT2 inhibitors as a calorie restriction mimetic in anti-aging therapeutics.
Collapse
Affiliation(s)
- Caroline W S Hoong
- Division of Endocrinology, Department of General Medicine, Woodlands Health Campus, National Healthcare Group Singapore, Woodlands Health Campus Singapore, 768024, Singapore
| | - Marvin W J Chua
- Endocrinology Service, Department of General Medicine, Sengkang General Hospital, SingHealth Group Singapore, Sengkang General Hospital Singapore, 544886, Singapore
| |
Collapse
|
18
|
Tsai KF, Chen YL, Chiou TTY, Chu TH, Li LC, Ng HY, Lee WC, Lee CT. Emergence of SGLT2 Inhibitors as Powerful Antioxidants in Human Diseases. Antioxidants (Basel) 2021; 10:1166. [PMID: 34439414 PMCID: PMC8388972 DOI: 10.3390/antiox10081166] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/10/2021] [Accepted: 07/19/2021] [Indexed: 12/14/2022] Open
Abstract
Sodium-glucose cotransporter 2 (SGLT2) inhibitors are a new class of oral glucose-lowering agents. Apart from their glucose-lowering effects, large clinical trials assessing certain SGLT2 inhibitors have revealed cardiac and renal protective effects in non-diabetic patients. These excellent outcomes motivated scientists and clinical professionals to revisit their underlying mechanisms. In addition to the heart and kidney, redox homeostasis is crucial in several human diseases, including liver diseases, neural disorders, and cancers, with accumulating preclinical studies demonstrating the therapeutic benefits of SGLT2 inhibitors. In the present review, we aimed to update recent advances in the antioxidant roles of SGLT2 inhibitors in common but debilitating human diseases. We anticipate that this review will guide new research directions and novel therapeutic strategies for diabetes, cardiovascular diseases, nephropathies, liver diseases, neural disorders, and cancers in the era of SGLT2 inhibitors.
Collapse
Affiliation(s)
- Kai-Fan Tsai
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (K.-F.T.); (T.T.-Y.C.); (L.-C.L.); (H.-Y.N.)
| | - Yung-Lung Chen
- Section of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan;
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Terry Ting-Yu Chiou
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (K.-F.T.); (T.T.-Y.C.); (L.-C.L.); (H.-Y.N.)
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Tian-Huei Chu
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan;
- Biobank and Tissue Bank, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Lung-Chih Li
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (K.-F.T.); (T.T.-Y.C.); (L.-C.L.); (H.-Y.N.)
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Hwee-Yeong Ng
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (K.-F.T.); (T.T.-Y.C.); (L.-C.L.); (H.-Y.N.)
| | - Wen-Chin Lee
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (K.-F.T.); (T.T.-Y.C.); (L.-C.L.); (H.-Y.N.)
| | - Chien-Te Lee
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (K.-F.T.); (T.T.-Y.C.); (L.-C.L.); (H.-Y.N.)
| |
Collapse
|
19
|
Yabe D, Shiki K, Suzaki K, Meinicke T, Kotobuki Y, Nishida K, Clark D, Yasui A, Seino Y. Rationale and design of the EMPA-ELDERLY trial: a randomised, double-blind, placebo-controlled, 52-week clinical trial of the efficacy and safety of the sodium-glucose cotransporter-2 inhibitor empagliflozin in elderly Japanese patients with type 2 diabetes. BMJ Open 2021; 11:e045844. [PMID: 33827843 PMCID: PMC8031078 DOI: 10.1136/bmjopen-2020-045844] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
INTRODUCTION Elderly people (≥65 years) with type 2 diabetes mellitus (T2DM) are becoming increasingly prevalent, notably in Japan. As cardiovascular (CV) risk increases with age and sodium-glucose cotransporter-2 (SGLT2) inhibitors reduce CV risk, elderly patients with T2DM are increasingly likely to be prescribed these glucose-lowering drugs. There is controversy surrounding the effects of SGLT2 inhibitors on muscle mass, particularly in elderly patients for whom loss of muscle is especially undesirable; however, robust evidence on this important issue is lacking. Consequently, we have designed a clinical trial of the SGLT2 inhibitor empagliflozin in elderly Japanese patients with T2DM (Empagliflozin in Elderly T2DM Patients (EMPA-ELDERLY)) to assess its effects on body composition as well as glycaemic control. EMPA-ELDERLY will be the first randomised clinical trial of an SGLT2 inhibitor in elderly patients with T2DM to evaluate effects on skeletal muscle mass, muscle strength and physical performance concurrently. METHODS AND ANALYSIS EMPA-ELDERLY is a randomised, double-blind, placebo-controlled, parallel-group clinical trial to be conducted in Japan. Patients with T2DM aged ≥65 years are eligible if they are Japanese with a body mass index of ≥22 kg/m2 and glycated haemoglobin (HbA1c) levels from ≥7.0% to ≤10.0% from either diet and exercise alone or treatment with oral glucose-lowering drugs. Approximately 128 participants will be randomised 1:1 to once per day, oral, double-blind treatment with empagliflozin 10 mg or matching placebo for 52 weeks. The primary endpoint is the change in HbA1c level from baseline at week 52. Secondary endpoints include changes from baseline to 52 weeks in body composition, including muscle mass and body fat, measured by bioelectrical impedance analysis, as well as skeletal muscle index, grip strength and time in the five-time chair stand test. Other endpoints include changes in patient-reported outcomes (including quality of life), cognitive function and safety. ETHICS AND DISSEMINATION We will submit the trial results to conferences and peer-reviewed journals. TRIAL REGISTRATION NUMBER NCT04531462.
Collapse
Affiliation(s)
- Daisuke Yabe
- Department of Diabetes and Endocrinology, Gifu University Graduate School of Medicine, Gifu, Japan
- Yutaka Seino Distinguished Centre for Diabetes Research, Kansai Electric Power Medical Research Institute, Kobe, Japan
- Division of Molecular and Metabolic Medicine, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kosuke Shiki
- Medicine Division, Nippon Boehringer Ingelheim Co. Ltd, Tokyo, Japan
| | - Keiko Suzaki
- Medicine Division, Nippon Boehringer Ingelheim Co. Ltd, Tokyo, Japan
| | - Thomas Meinicke
- Medicine Division, Boehringer Ingelheim International GmbH, Biberach, Germany
| | - Yutaro Kotobuki
- Medicine Division, Nippon Boehringer Ingelheim Co. Ltd, Tokyo, Japan
| | - Kenichiro Nishida
- Biostatistics and Data Science, Nippon Boehringer Ingelheim Co. Ltd, Tokyo, Japan
| | - Douglas Clark
- Medicine Division, Boehringer Ingelheim UK Ltd, Bracknell, UK
| | - Atsutaka Yasui
- Medicine Division, Nippon Boehringer Ingelheim Co. Ltd, Tokyo, Japan
| | - Yutaka Seino
- Yutaka Seino Distinguished Centre for Diabetes Research, Kansai Electric Power Medical Research Institute, Kobe, Japan
- Center for Diabetes, Endocrinology and Metabolism, Kansai Electric Power Hospital, Osaka, Japan
| |
Collapse
|
20
|
Nagayama A, Ashida K, Watanabe M, Moritaka K, Sonezaki A, Kitajima Y, Takahashi H, Yoshinobu S, Iwata S, Yasuda J, Hasuzawa N, Ozono S, Motomura S, Nomura M. Case Report: Metreleptin and SGLT2 Inhibitor Combination Therapy Is Effective for Acquired Incomplete Lipodystrophy. Front Endocrinol (Lausanne) 2021; 12:690996. [PMID: 34135866 PMCID: PMC8201990 DOI: 10.3389/fendo.2021.690996] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 05/13/2021] [Indexed: 12/30/2022] Open
Abstract
Childhood cancer survivors (CCSs) who have undergone bone marrow transplantation with systemic chemotherapy and whole-body irradiation often experience impaired glucose tolerance with marked insulin resistance. Incomplete acquired diabetic lipodystrophy should be considered as a late complication of bone marrow transplantation. A 24-year-old Japanese female patient with incomplete acquired lipodystrophy, a CCS of acute lymphocytic leukemia at the age of 3 years, was treated for diabetes mellitus and dyslipidemia at our hospital. Administration of multiple daily insulin injections (70 units/day), and oral administration of 500 mg/day metformin, 15 mg/day pioglitazone, and 200 mg/day bezafibrate had proven ineffective for her metabolic disorders. Subcutaneous administration of metreleptin improved her insulin resistance and hypertriglyceridemia within a month; however, it failed to maintain adequate plasma glucose levels in the long term. When oral administration of 10 mg/day empagliflozin was added to the metreleptin supplementation, her HbA1c value (National Glycohemoglobin Standardization Program) improved from 11% to 8%, which was maintained for an additional 18 months. This is the first case report of incomplete lipodystrophy that shows efficacy of a combination therapy with metreleptin and a sodium glucose cotransporter 2 (SGLT2) inhibitor for the treatment of diabetes and dyslipidemia. An SGLT2 inhibitor attenuates hyperglycemia through urinary glucose excretion and has been suggested to enhance lipid catabolism in the extra-adipose tissues, especially in the liver and skeletal muscles. Furthermore, metreleptin supplementation could enhance the action of the SGLT2 inhibitor by promoting satiety and lipolysis through the central nervous system. Combination therapy with metreleptin and an SGLT2 inhibitor was suggested to recover the volume of adipose tissue, possibly through improvement of insulin resistance in the adipose tissue. This report highlights the pathophysiological mechanism of an SGLT2 inhibitor in the improvement of glucose metabolism in non-healthy lean CCSs with insulin resistance. Administration of SGLT2 inhibitor, along with metreleptin supplementation, could be a good alternative therapy for diabetic lipodystrophy observed in CCSs.
Collapse
Affiliation(s)
- Ayako Nagayama
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Kenji Ashida
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
- *Correspondence: Kenji Ashida,
| | - Miki Watanabe
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Kanoko Moritaka
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Aya Sonezaki
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | | | - Hirokazu Takahashi
- Division of Hepatology, Diabetes Mellitus, and Endocrinology, Department of Internal Medicine, Saga University, Saga, Japan
| | - Satoko Yoshinobu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Shimpei Iwata
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Junichi Yasuda
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Nao Hasuzawa
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Shuichi Ozono
- Department of Pediatrics and Child Health, Kurume University School of Medicine, Kurume, Japan
| | - Seiichi Motomura
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Masatoshi Nomura
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| |
Collapse
|