1
|
Angermann CE, Sehner S, Gerhardt LMS, Santos-Gallego CG, Requena-Ibanez JA, Zeller T, Maack C, Sanz J, Frantz S, Ertl G, Badimon JJ. Anaemia predicts iron homoeostasis dysregulation and modulates the response to empagliflozin in heart failure with reduced ejection fraction: the EMPATROPISM-FE trial. Eur Heart J 2025; 46:1507-1523. [PMID: 39907687 PMCID: PMC12011522 DOI: 10.1093/eurheartj/ehae917] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/18/2024] [Accepted: 09/27/2024] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND AND AIMS Sodium-glucose cotransporter 2 inhibitors (SGLT2i) impact iron metabolism in patients with heart failure but mechanisms are incompletely understood. This post hoc analysis explored interrelations between iron homeostasis, cardiac structure/function, exercise capacity, haematopoiesis, and sympathetic activity at baseline, and the effects of 6-month treatment with empagliflozin vs. placebo by anaemia status in EMPATROPISM-FE study participants. METHODS Myocardial iron content (MIC, estimated by cardiac magnetic resonance T2* imaging), left ventricular (LV) volumes and LV ejection fraction (LVEF), exercise capacity, laboratory iron markers (LIM), haemoglobin/haematocrit, erythropoietin, and plasma norepinephrine were determined at baseline and 6 months. RESULTS At baseline, 24/80 participants (30%) had anaemia (haemoglobin < 13/<12 mg/dL in men/women). Patients with vs. without anaemia had higher T2* (indicating lower MIC, P < .001), lower peak oxygen consumption (VO2max, P = .024) and hepcidin (P = .017), and higher erythropoietin (P = .040) and norepinephrine (P = .016). Across subgroups, lower MIC correlated with higher LV volumes (P < .01) and norepinephrine (P < .001), and lower LVEF (P < .01), VO2max (P < .001) and haemoglobin/haematocrit (P < .001). Associations with LIM were poor (all P > .10). Empagliflozin increased MIC (P < .012), improved exercise capacity, and activated haematopoiesis. Changes in LIM and norepinephrine suggested progressive systemic iron depletion and sympatholysis. LV reverse remodelling was greater in individuals with anaemia. CONCLUSIONS Dysregulated cellular iron uptake/availability may be a shared mechanism in myocardial structural/functional impairment, reduced exercise capacity, and restricted haematopoiesis in heart failure, which are worse in patients with anaemia, and improve with empagliflozin. Empagliflozin increases MIC and decreases norepinephrine. Given this inverse association, sympatholysis may help explain the diverse cardiac and systemic benefits from SGLT2i therapy. CLINICAL TRIAL REGISTRATION NCT03485222 (www.clinicaltrials.gov).
Collapse
Affiliation(s)
- Christiane E Angermann
- Comprehensive Heart Failure Center Würzburg, University and University Hospital Würzburg, Am Schwarzenberg 15, 97078 Würzburg, Germany
- Department of Medicine 1, University Hospital Würzburg, Oberdürrbacherstraße 6, 97080 Würzburg, Germany
| | - Susanne Sehner
- Institute of Medical Biometry and Epidemiology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Louisa M S Gerhardt
- Fifth Department of Medicine, University Medical Center Mannheim, Faculty of Medicine Mannheim of the University of Heidelberg, Mannheim, Germany
| | | | | | - Tanja Zeller
- University Center of Cardiovascular Science, University Heart and Vascular Center, University Hospital Hamburg-Eppendorf, and German Center of Cardiovascular Research, Partner Site Hamburg–Kiel–Lübeck, Hamburg, Germany
| | - Christoph Maack
- Comprehensive Heart Failure Center Würzburg, University and University Hospital Würzburg, Am Schwarzenberg 15, 97078 Würzburg, Germany
| | - Javier Sanz
- Icahn School of Medicine at Mount Sinai, Cardiovascular Institute, New York, NY, USA
| | - Stefan Frantz
- Comprehensive Heart Failure Center Würzburg, University and University Hospital Würzburg, Am Schwarzenberg 15, 97078 Würzburg, Germany
- Department of Medicine 1, University Hospital Würzburg, Oberdürrbacherstraße 6, 97080 Würzburg, Germany
| | - Georg Ertl
- Comprehensive Heart Failure Center Würzburg, University and University Hospital Würzburg, Am Schwarzenberg 15, 97078 Würzburg, Germany
- Department of Medicine 1, University Hospital Würzburg, Oberdürrbacherstraße 6, 97080 Würzburg, Germany
| | - Juan J Badimon
- Icahn School of Medicine at Mount Sinai, Cardiovascular Institute, New York, NY, USA
| |
Collapse
|
2
|
López-González ÁA, Martínez-Almoyna Rifá E, Paublini Oliveira H, Martorell Sánchez C, Tárraga López PJ, Ramírez-Manent JI. [Association between sociodemographic variables, healthy habits and stress with metabolic syndrome. A descriptive, cross-sectional study]. Semergen 2025; 51:102455. [PMID: 39922044 DOI: 10.1016/j.semerg.2025.102455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 12/17/2024] [Accepted: 01/07/2025] [Indexed: 02/10/2025]
Abstract
INTRODUCTION Metabolic syndrome (MS) is a pathological condition that encompasses various cardiometabolic risk factors, such as obesity, dyslipidemia, hyperglycemia, and elevated blood pressure levels. It is considered a multifactorial pathological condition. The aim of this study is to assess how variables such as age, sex, socioeconomic status, tobacco and alcohol consumption, physical activity, adherence to the Mediterranean diet, and stress are associated with the prevalence of MS, determined using two different criteria. MATERIALS AND METHODS This is a descriptive, cross-sectional study conducted on 24,224 Spanish workers, evaluating the association between sociodemographic variables, health habits, and stress with MS, determined using two criteria: the National Cholesterol Education Program, Adult Treatment Panel III (NCEP ATP-III), and the International Diabetes Federation (IDF). RESULTS All the variables analyzed showed an association with the presence of MS when applying both criteria. Among them, the variables with the strongest association were age: odds ratio 5.55 (95% CI: 4.80-6.30) for MS using the NCEP ATP-III criteria and 6.71 (95% CI: 5.30-8.13) for IDF criteria; and type of job: odds ratio 3.42 (95% CI: 2.95-3.90) for NCEP ATP-III and 3.57 (95% CI: 3.12-4.03) for IDF. CONCLUSIONS The profile of an individual at higher risk of developing MS under both criteria would be an older male, manual laborer, smoker, habitual alcohol consumer, sedentary, with low adherence to the Mediterranean diet, and experiencing high levels of stress.
Collapse
Affiliation(s)
- Á A López-González
- Grupo ADEMA-Salud del Instituto Universitario de Ciencias de la Salud (IUNICS) de Baleares, Palma, Islas Baleares, España; Facultad de Odontología, Escuela Universitaria ADEMA-UIB, Palma, Islas Baleares, España; Servicio de Salud de las Islas Baleares, España
| | - E Martínez-Almoyna Rifá
- Grupo ADEMA-Salud del Instituto Universitario de Ciencias de la Salud (IUNICS) de Baleares, Palma, Islas Baleares, España; Facultad de Odontología, Escuela Universitaria ADEMA-UIB, Palma, Islas Baleares, España
| | - H Paublini Oliveira
- Grupo ADEMA-Salud del Instituto Universitario de Ciencias de la Salud (IUNICS) de Baleares, Palma, Islas Baleares, España; Facultad de Odontología, Escuela Universitaria ADEMA-UIB, Palma, Islas Baleares, España
| | - C Martorell Sánchez
- Grupo ADEMA-Salud del Instituto Universitario de Ciencias de la Salud (IUNICS) de Baleares, Palma, Islas Baleares, España; Facultad de Odontología, Escuela Universitaria ADEMA-UIB, Palma, Islas Baleares, España
| | - P J Tárraga López
- Facultad de Medicina, Universidad de Castilla-La Mancha, Albacete, España.
| | - J I Ramírez-Manent
- Grupo ADEMA-Salud del Instituto Universitario de Ciencias de la Salud (IUNICS) de Baleares, Palma, Islas Baleares, España; Servicio de Salud de las Islas Baleares, España; Facultad de Medicina, Universidad de las Islas Baleares, Palma, Islas Baleares, España
| |
Collapse
|
3
|
Li X, Huang B, Liu Y, Wang M, Cui JQ. Uric acid in diabetic microvascular complications: Mechanisms and therapy. J Diabetes Complications 2025; 39:108929. [PMID: 39689504 DOI: 10.1016/j.jdiacomp.2024.108929] [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: 09/12/2024] [Revised: 11/30/2024] [Accepted: 12/03/2024] [Indexed: 12/19/2024]
Abstract
Uric acid (UA) is mainly synthesized in the liver, intestine, and vascular endothelium and excreted by the kidney (70 %) and intestine (30 %). Hyperuricemia (HUA) occurs when UA production exceeds excretion. Many studies have found that elevated UA is associated with diabetic microvascular complications (DMC), including diabetic retinopathy (DR), diabetic nephropathy (DN), and diabetic peripheral neuropathy (DPN). In addition, too high or too low UA levels will promote the occurrence and development of chronic diseases, but the relationship between UA and diabetic microvascular complications (DMC) is not clear. Therefore, the rational treatment of UA in patients with diabetes is essential. In this review, we summarize and discuss the mechanism and treatment of UA and DMC and may provide potential advice for rational drug selection.
Collapse
Affiliation(s)
- Xin Li
- Tianjin Medical University General Hospital, People's Republic of China
| | - Bo Huang
- Tianjin Medical University General Hospital, People's Republic of China
| | - Yue Liu
- Tianjin Medical University General Hospital, People's Republic of China
| | - Meng Wang
- Tianjin Medical University General Hospital, People's Republic of China
| | - Jing-Qiu Cui
- Tianjin Medical University General Hospital, People's Republic of China.
| |
Collapse
|
4
|
Zhang S, Huang Y, Han C, Wang F, Chen M, Yang Z, Yang S, Wang C. Central SGLT2 mediate sympathoexcitation in hypertensive heart failure via attenuating subfornical organ endothelial cGAS ubiquitination to amplify neuroinflammation: Molecular mechanism behind sympatholytic effect of Empagliflozin. Int Immunopharmacol 2025; 145:113711. [PMID: 39647283 DOI: 10.1016/j.intimp.2024.113711] [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: 10/13/2024] [Revised: 11/22/2024] [Accepted: 11/22/2024] [Indexed: 12/10/2024]
Abstract
BACKGROUND Sodium/glucose co-transporter 2 (SGLT2) inhibitors have transformed heart failure (HF) treatment, offering sympatholytic effects whose mechanisms are not fully understood. Our previous studies identified Cyclic GMP-AMP synthase (cGAS)-derived neuroinflammation in the Subfornical organ (SFO) as a promoter of sympathoexcitation, worsening myocardial remodeling in HF. This research explored the role of central SGLT2 in inducing endothelial cGAS-driven neuroinflammation in the SFO during HF and assessed the impact of SGLT2 inhibitors on this process. METHODS Hypertensive HF was induced in mice via Angiotensin II infusion for four weeks. SGLT2 expression and localization in the SFO were determined through immunoblotting and double-immunofluorescence staining. AAV9-TIE-shRNA (SGLT2) facilitated targeted SGLT2 knockdown in SFO endothelial cells (ECs), with subsequent analyses via immunoblotting, staining, and co-immunoprecipitation to investigate interactions with cGAS, mitochondrial alterations, and pro-inflammatory pathway activation. Renal sympathetic nerve activity and heart rate variability were measured to assess sympathetic output, alongside evaluations of cardiac function in HF mice. RESULTS In HF model mice, SGLT2 levels are markedly raised in SFO ECs, disrupting mitochondrial function and elevating oxidative stress. SGLT2 knockdown preserved mitochondrial integrity and function, reduced inflammation, and highlighted the influence of SGLT2 on mitochondrial health. SGLT2's interaction with cGAS prevented its ubiquitination and degradation, amplifying neuroinflammation and HF progression. Conversely, Empagliflozin counteracted these effects, suggesting that targeting the SGLT2-cGAS interaction as a novel HF treatment avenue. CONCLUSION This study revealed that SGLT2 directly reduced cGAS degradation in brain ECs, enhancing neuroinflammation in the SFO, and promoting sympathoexcitation and myocardial remodeling. The significance of the central SGLT2-cGAS interaction in cardiovascular disease mechanisms is emphasized.
Collapse
Affiliation(s)
- Shutian Zhang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China.
| | - Yijun Huang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Chengzhi Han
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Fanshun Wang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Maoxiang Chen
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Zhaohua Yang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Shouguo Yang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China.
| | - Chunsheng Wang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China.
| |
Collapse
|
5
|
Azmy Nabeh O, Amr A, Faoosa AM, Esmat E, Osama A, Khedr AS, Amin B, Saud AI, Elmorsy SA. Emerging Perspectives on the Impact of Diabetes Mellitus and Anti-Diabetic Drugs on Premenstrual Syndrome. A Narrative Review. Diabetes Ther 2024; 15:1279-1299. [PMID: 38668996 PMCID: PMC11096298 DOI: 10.1007/s13300-024-01585-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 04/08/2024] [Indexed: 05/16/2024] Open
Abstract
Diabetes mellitus (DM) and premenstrual syndrome (PMS) are global health challenges. Both disorders are often linked to a range of physical and psychological symptoms that significantly impact the quality of life of many women. Yet, the exact relation between DM and PMS is not clear, and the management of both conditions poses a considerable challenge. In this review, we aimed to investigate the interplay between DM, anti-diabetic drugs, and the different theories and symptoms of PMS. Female sex hormones are implicated in the pathophysiology of PMS and can also impair blood glucose control. In addition, patients with diabetes face a higher susceptibility to anxiety and depression disorders, with a significant number of patients experiencing symptoms such as fatigue and difficulty concentrating, which are reported in patients with PMS as well. Complications related to diabetic medications, such as hypoglycemia (with sulfonylurea) and fluid retention (with thiazolidinediones) may also mediate PMS-like symptoms. DM can, in addition, disturb the normal gut microbiota (GM), with a consequent loss of beneficial GM metabolites that guard against PMS, particularly the short-chain fatty acids and serotonin. Among the several available anti-diabetic drugs, those (1) with an anti-inflammatory potential, (2) that can preserve the beneficial GM, and (3) possessing a lower risk for hypoglycemia, might have a favorable outcome in PMS women. Yet, well-designed clinical trials are needed to investigate the anti-diabetic drug(s) of choice for patients with diabetes and PMS.
Collapse
Affiliation(s)
- Omnia Azmy Nabeh
- Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt.
| | - Alaa Amr
- Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | | | - Eshraka Esmat
- Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Alaa Osama
- Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | | | - Basma Amin
- Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Alaa I Saud
- Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | | |
Collapse
|
6
|
Mondal S, Pramanik S, Khare VR, Fernandez CJ, Pappachan JM. Sodium glucose cotransporter-2 inhibitors and heart disease: Current perspectives. World J Cardiol 2024; 16:240-259. [PMID: 38817648 PMCID: PMC11135334 DOI: 10.4330/wjc.v16.i5.240] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 04/08/2024] [Accepted: 04/28/2024] [Indexed: 05/23/2024] Open
Abstract
Sodium glucose cotransporter-2 inhibitors (SGLT-2i) are antidiabetic medications with remarkable cardiovascular (CV) benefits proven by multiple randomised controlled trials and real-world data. These drugs are also useful in the prevention of CV disease (CVD) in patients with diabetes mellitus (DM). Although DM as such is a huge risk factor for CVD, the CV benefits of SGLT-2i are not just because of antidiabetic effects. These molecules have proven beneficial roles in prevention and management of nondiabetic CVD and renal disease as well. There are various molecular mechanisms for the organ protective effects of SGLT-2i which are still being elucidated. Proper understanding of the role of SGLT-2i in prevention and management of CVD is important not only for the cardiologists but also for other specialists caring for various illnesses which can directly or indirectly impact care of heart diseases. This clinical review compiles the current evidence on the rational use of SGLT-2i in clinical practice.
Collapse
Affiliation(s)
- Sunetra Mondal
- Department of Endocrinology, NRS Medical College, Kolkata 700020, West Bengal, India
| | - Subhodip Pramanik
- Department of Endocrinology, Neotia Getwel Multispecialty Hospitals, Siliguri 734010, West Bengal, India
| | - Vibhu Ranjan Khare
- Department of Endocrinology, NRS Medical College, Kolkata 700020, West Bengal, India
| | - Cornelius James Fernandez
- Department of Endocrinology and Metabolism, Pilgrim Hospital, United Lincolnshire Hospitals NHS Trust, Boston PE21 9QS, United Kingdom
| | - Joseph M Pappachan
- Department of Endocrinology and Metabolism, Lancashire Teaching Hospitals NHS Trust, Preston PR2 9HT, United Kingdom
- Faculty of Science, Manchester Metropolitan University, Manchester M15 6BH, United Kingdom
- Faculty of Biology, Medicine & Health, The University of Manchester, Manchester M13 9PL, United Kingdom.
| |
Collapse
|
7
|
Zhang Q, Deng Z, Li T, Chen K, Zeng Z. SGLT2 inhibitor improves the prognosis of patients with coronary heart disease and prevents in-stent restenosis. Front Cardiovasc Med 2024; 10:1280547. [PMID: 38274313 PMCID: PMC10808651 DOI: 10.3389/fcvm.2023.1280547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 12/27/2023] [Indexed: 01/27/2024] Open
Abstract
Coronary heart disease is a narrowing or obstruction of the vascular cavity caused by atherosclerosis of the coronary arteries, which leads to myocardial ischemia and hypoxia. At present, percutaneous coronary intervention (PCI) is an effective treatment for coronary atherosclerotic heart disease. Restenosis is the main limiting factor of the long-term success of PCI, and it is also a difficult problem in the field of intervention. Sodium-glucose cotransporter 2 (SGLT2) inhibitor is a new oral glucose-lowering agent used in the treatment of diabetes in recent years. Recent studies have shown that SGLT2 inhibitors can effectively improve the prognosis of patients after PCI and reduce the occurrence of restenosis. This review provides an overview of the clinical studies and mechanisms of SGLT2 inhibitors in the prevention of restenosis, providing a new option for improving the clinical prognosis of patients after PCI.
Collapse
Affiliation(s)
| | | | | | | | - Zhihuan Zeng
- Department of Cardiovascular Diseases, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, China
| |
Collapse
|
8
|
Oshima N, Onimaru H, Yamashiro A, Goto H, Tanoue K, Fukunaga T, Sato H, Uto A, Matsubara H, Imakiire T, Kumagai H. SGLT2 and SGLT1 inhibitors suppress the activities of the RVLM neurons in newborn Wistar rats. Hypertens Res 2024; 47:46-54. [PMID: 37710035 DOI: 10.1038/s41440-023-01417-5] [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: 07/05/2023] [Revised: 08/11/2023] [Accepted: 08/20/2023] [Indexed: 09/16/2023]
Abstract
Hypertension is well-known to often coexist with diabetes mellitus (DM) in humans. Treatment with sodium-glucose cotransporter 2 (SGLT2) inhibitors has been shown to decrease both the blood glucose and the blood pressure (BP) in such patients. Some reports show that SGLT2 inhibitors improve the BP by decreasing the activities of the sympathetic nervous system. Therefore, we hypothesized that SGLT2 inhibitors might alleviate hypertension via attenuating sympathetic nervous activity. Combined SGLT2/SGLT1 inhibitor therapy is also reported as being rather effective for decreasing the BP. In this study, we examined the effects of SGLT2 and SGLT1 inhibitors on the bulbospinal neurons of the rostral ventrolateral medulla (RVLM). To investigate whether bulbospinal RVLM neurons are sensitive to SGLT2 and SGLT1 inhibitors, we examined the changes in the neuronal membrane potentials (MPs) of these neurons using the whole-cell patch-clamp technique during superfusion of the cells with the SGLT2 and SGLT1 inhibitors. A brainstem-spinal cord preparation was used for the experiments. Our results showed that superfusion of the RVLM neurons with SGLT2 and SGLT1 inhibitor solutions induced hyperpolarization of the neurons. Histological examination revealed the presence of SGLT2s and SGLT1s in the RVLM neurons, and also colocalization of SGLT2s with SGLT1s. These results suggest the involvement of SGLT2s and SGLT1s in regulating the activities of the RVLM neurons, so that SGLT2 and SGLT1 inhibitors may inactivate the RVLM neurons hyperpolarized by empagliflozin. SGLT2 and SGLT1 inhibitors suppressed the activities of the bulbospinal RVLM neurons in the brainstem-spinal preparations, suggesting the possibilities of lowering BP by decreasing the sympathetic nerve activities. RVLM, rostral ventrolateral medulla. IML, intralateral cell column. aCSF, artificial cerebrospinal fluid.
Collapse
Affiliation(s)
- Naoki Oshima
- Department of Nephrology and Endocrinology, National Defense Medical College, Tokorozawa, Saitama, Japan.
| | - Hiroshi Onimaru
- Department of Physiology, Showa University School of Medicine, Tokyo, Japan
| | - Aoi Yamashiro
- Department of Nephrology and Endocrinology, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Hiroyasu Goto
- Department of Nephrology and Endocrinology, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Keiko Tanoue
- Department of Nephrology and Endocrinology, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Tsugumi Fukunaga
- Department of Nephrology and Endocrinology, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Hiroki Sato
- Department of Nephrology and Endocrinology, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Asuka Uto
- Department of Nephrology and Endocrinology, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Hidehito Matsubara
- Department of Nephrology and Endocrinology, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Toshihiko Imakiire
- Department of Nephrology and Endocrinology, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Hiroo Kumagai
- Department of Nephrology, Sayama General Clinic, Iruma, Saitama, Japan
| |
Collapse
|
9
|
Manolis AA, Manolis TA, Melita H, Manolis AS. Sodium-glucose cotransporter type 2 inhibitors and cardiac arrhythmias. Trends Cardiovasc Med 2023; 33:418-428. [PMID: 35447305 DOI: 10.1016/j.tcm.2022.04.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/02/2022] [Accepted: 04/15/2022] [Indexed: 02/06/2023]
Abstract
The introduction of sodium-glucose cotransporter 2 (SGLT2) inhibitors as a new and effective class of therapeutic agents for type 2 diabetes (T2D) preventing the reabsorption of glucose in the kidneys and thus facilitating glucose excretion in the urine, but also as agents with cardiovascular benefits, particularly in patients with heart failure (HF), regardless of the diabetic status, has ushered in a new era in treating patients with T2D and/or HF. In addition, data have recently emerged indicating an antiarrhythmic effect of the SGLT2 inhibitors in patients with and without diabetes. Prospective studies, randomized controlled trials and meta-analyses have provided robust evidence for a protective and beneficial effect of these agents against atrial fibrillation, ventricular arrhythmias and sudden cardiac death. The antiarrhythmic mechanisms involved include reverse atrial and ventricular remodeling, amelioration of mitochondrial function, reduction of hypoglycemic episodes with their attendant arrhythmogenic effects, attenuated sympathetic nervous system activity, regulation of sodium and calcium homeostasis, and suppression of prolonged ventricular repolarization. These new data on antiarrhythmic actions of SGLT2 inhibitors are herein reviewed, potential mechanisms involved are discussed and pictorially illustrated, and treatment results on specific arrhythmias are described and tabulated.
Collapse
Affiliation(s)
| | | | | | - Antonis S Manolis
- First Department of Cardiology, Athens University School of Medicine, Athens, Greece.
| |
Collapse
|
10
|
Matthews J, Herat L, Schlaich MP, Matthews V. The Impact of SGLT2 Inhibitors in the Heart and Kidneys Regardless of Diabetes Status. Int J Mol Sci 2023; 24:14243. [PMID: 37762542 PMCID: PMC10532235 DOI: 10.3390/ijms241814243] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/12/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Chronic Kidney Disease (CKD) and Cardiovascular Disease (CVD) are two devastating diseases that may occur in nondiabetics or individuals with diabetes and, when combined, it is referred to as cardiorenal disease. The impact of cardiorenal disease on society, the economy and the healthcare system is enormous. Although there are numerous therapies for cardiorenal disease, one therapy showing a great deal of promise is sodium-dependent glucose cotransporter 2 (SGLT2) inhibitors. The SGLT family member, SGLT2, is often implicated in the pathogenesis of a range of diseases, and the dysregulation of the activity of SGLT2 markedly effects the transport of glucose and sodium across the luminal membrane of renal cells. Inhibitors of SGLT2 were developed based on the antidiabetic action initiated by inhibiting renal glucose reabsorption, thereby increasing glucosuria. Of great medical significance, large-scale clinical trials utilizing a range of SGLT2 inhibitors have demonstrated both metabolic and biochemical benefits via numerous novel mechanisms, such as sympathoinhibition, which will be discussed in this review. In summary, SGLT2 inhibitors clearly exert cardio-renal protection in people with and without diabetes in both preclinical and clinical settings. This exciting class of inhibitors improve hyperglycemia, high blood pressure, hyperlipidemia and diabetic retinopathy via multiple mechanisms, of which many are yet to be elucidated.
Collapse
Affiliation(s)
- Jennifer Matthews
- Royal Perth Hospital Unit, Dobney Hypertension Centre, School of Biomedical Sciences, University of Western Australia, Crawley, WA 6009, Australia; (J.M.); (L.H.)
| | - Lakshini Herat
- Royal Perth Hospital Unit, Dobney Hypertension Centre, School of Biomedical Sciences, University of Western Australia, Crawley, WA 6009, Australia; (J.M.); (L.H.)
| | - Markus P. Schlaich
- Royal Perth Hospital Unit, Dobney Hypertension Centre, School of Medicine, University of Western Australia, Crawley, WA 6009, Australia;
- Department of Cardiology and Department of Nephrology, Royal Perth Hospital, Perth, WA 6000, Australia
| | - Vance Matthews
- Royal Perth Hospital Unit, Dobney Hypertension Centre, School of Biomedical Sciences, University of Western Australia, Crawley, WA 6009, Australia; (J.M.); (L.H.)
| |
Collapse
|
11
|
Bahat G, Ozkok S, Petrovic M. Management of Type 2 Diabetes in Frail Older Adults. Drugs Aging 2023; 40:751-761. [PMID: 37434085 DOI: 10.1007/s40266-023-01049-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2023] [Indexed: 07/13/2023]
Abstract
Aging is one of the most important factors associated with the dramatic increase in the prevalence of type 2 diabetes mellitus (T2DM) globally. In addition to traditional micro- and macrovascular complications, diabetes mellitus (DM) in older adults is of great importance due to its independent relationship with frailty, which is defined as a decline in functional reserves and vulnerability to stressors. Frailty assessment enables the determination of biological age, thus predicting potential complications in older adults and identifying tailored treatment strategies. Although the latest guidelines have acknowledged the frailty concept and provided recommendations specific to this subgroup of older adults, frail older adults are particularly considered only as anorexic, malnourished people for whom relaxed treatment targets should be set. However, this approach bypasses other metabolic phenotypes in the context of diabetes and frailty. Recently, a spectrum of metabolic phenotypes in the context of frailty in DM was suggested, and the two edges of this spectrum were defined as "anorexic malnourished (AM)" and "sarcopenic obese (SO)." These two edges were suggested to require different strategies: Opposite to the AM phenotype requiring less stringent targets and de-intensification of treatments, tight blood glucose control with agents promoting weight loss was recommended in the SO group. Our suggestion is that, regardless of their phenotype, weight loss should not be the primary goal in DM management in older adults who are overweight or obese, because of the increased malnutrition prevalence in older adults suffering from DM compared with standard older adults. Furthermore, overweight older adults have been reported to have the lowest risk of mortality compared with other groups. On the other hand, obese older individuals may benefit from intensive lifestyle interventions including caloric restriction and regular exercise with the assurance of at least 1 g/kg/day high-quality protein intake. Besides metformin (MF), sodium-glucose cotransporter-2 inhibitors (SGLT-2i) or glucagon-like peptide-1 receptor agonists (GLP-1RA) should be considered in appropriate SO cases, due to high evidence of cardiorenal benefits. MF should be avoided in the AM phenotype due to their weight loss property. Although weight loss is not desired in AM phenotype, SGLT-2i may still be preferred with close follow-up in certain individuals demonstrating high cardiovascular disease (CVD) risk. Of note, SGLT-2i should be considered earlier in the diabetes treatment in both groups due to their multiple benefits, i.e., organ protective effects, the potential to reduce polypharmacy, and improve frailty status. The concept of different metabolic phenotypes in frail older adults with diabetes once again shows "one size fits all" cannot be applied in geriatric medicine, and a tailored, individualized approach should be adopted to get the highest benefit from treatments.
Collapse
Affiliation(s)
- Gulistan Bahat
- Division of Geriatrics, Department of Internal Medicine, Istanbul Medical School, Istanbul University, Capa, 34390, Istanbul, Türkiye.
| | - Serdar Ozkok
- Division of Geriatrics, Department of Internal Medicine, Hatay Training and Research Hospital, 31040, Hatay, Türkiye
| | - Mirko Petrovic
- Section of Geriatrics, Department of Internal Medicine and Paediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| |
Collapse
|
12
|
Nabil-Adam A, E. Elnosary M, L. Ashour M, M. Abd El-Moneam N, A. Shreadah M. Flavonoids Biosynthesis in Plants as a Defense Mechanism: Role and Function Concerning Pharmacodynamics and Pharmacokinetic Properties. FLAVONOID METABOLISM - RECENT ADVANCES AND APPLICATIONS IN CROP BREEDING 2023. [DOI: 10.5772/intechopen.108637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
Flavonoids are a major class of secondary metabolites that comprises more than 6000 compounds that have been identified. They are biosynthesized via the phenylpropanoid metabolic pathway that involves groups of enzymes such as isomerases, hydroxylases, and reductases that greatly affect the determination of the flavonoid skeleton. For example, transferase enzymes responsible for the modification of sugar result in changes in the physiological activity of the flavonoids and changes in their physical properties, such as solubility, reactivity, and interaction with cellular target molecules, which affect their pharmacodynamics and pharmacokinetic properties. In addition, flavonoids have diverse biological activities such as antioxidants, anticancer, and antiviral in managing Alzheimer’s disease. However, most marine flavonoids are still incompletely discovered because marine flavonoid biosynthesis is produced and possesses unique substitutions that are not commonly found in terrestrial bioactive compounds. The current chapter will illustrate the importance of flavonoids’ role in metabolism and the main difference between marine and terrestrial flavonoids.
Collapse
|
13
|
Dong M, Chen H, Wen S, Yuan Y, Yang L, Li Y, Yuan X, Xu D, Zhou L. The Neuronal and Non-Neuronal Pathways of Sodium-Glucose Cotransporter-2 Inhibitor on Body Weight-Loss and Insulin Resistance. Diabetes Metab Syndr Obes 2023; 16:425-435. [PMID: 36820270 PMCID: PMC9938665 DOI: 10.2147/dmso.s399367] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 02/08/2023] [Indexed: 02/16/2023] Open
Abstract
With the emergence of sodium-glucose cotransporter 2 inhibitors (SGLT2i), the treatment of type 2 diabetes mellitus (T2DM) has achieved a new milestone, of which the insulin-independent mechanism could produce weight loss, improve insulin resistance (IR) and exert other protective effects. Besides the well-acknowledged biochemical processes, the dysregulated balance between sympathetic and parasympathetic activity may play a significant role in IR and obesity. Weight loss caused by SGLT-2i could be achieved via activating the liver-brain-adipose neural axis in adipocytes. We previously demonstrated that SGLT-2 are widely expressed in central nervous system (CNS) tissues, and SGLT-2i could inhibit central areas associated with autonomic control through unidentified pathways, indicating that the role of the central sympathetic inhibition of SGLT-2i on blood pressure and weight loss. However, the exact pathway of SGLT2i related to these effects and to what extent it depends on the neural system are not fully understood. The evidence of how SGLT-2i interacts with the nervous system is worth exploring. Therefore, in this review, we will illustrate the potential neurological processes by which SGLT2i improves IR in skeletal muscle, liver, adipose tissue, and other insulin-target organs via the CNS and sympathetic nervous system/parasympathetic nervous system (SNS/PNS).
Collapse
Affiliation(s)
- Meiyuan Dong
- Graduate School of Hebei Medical University, Shijiazhuang, People’s Republic of China
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Huiling Chen
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Song Wen
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Yue Yuan
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Liling Yang
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Yanyan Li
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Xinlu Yuan
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Dongxiang Xu
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Ligang Zhou
- Graduate School of Hebei Medical University, Shijiazhuang, People’s Republic of China
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling, Shanghai Pudong Hospital, Shanghai, People’s Republic of China
- Correspondence: Ligang Zhou, Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, People’s Republic of China, Tel +8613611927616, Email
| |
Collapse
|
14
|
Dong M, Chen H, Wen S, Yuan Y, Yang L, Xu D, Zhou L. The Mechanism of Sodium-Glucose Cotransporter-2 Inhibitors in Reducing Uric Acid in Type 2 Diabetes Mellitus. Diabetes Metab Syndr Obes 2023; 16:437-445. [PMID: 36820272 PMCID: PMC9938669 DOI: 10.2147/dmso.s399343] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 02/08/2023] [Indexed: 02/16/2023] Open
Abstract
Hyperuricemia is a common comorbidity in patients with type 2 diabetes mellitus (T2DM), as insulin resistance (IR) or hyperinsulinemia is associated with higher serum uric acid (SUA) levels due to decreased uric acid (UA) secretion, and SUA vice versa is an important risk factor that promotes the occurrence and progression of T2DM and its complications. Growing evidence suggests that sodium-glucose cotransporter 2 inhibitors (SGLT-2i), a novel anti-diabetic drug initially developed to treat T2DM, may exert favorable effects in reducing SUA. Currently, one of the possible mechanisms is that SGLT2i increases urinary glucose excretion, probably inhibiting glucose transport 9 (GLUT9)-mediated uric acid reabsorption in the collecting duct, resulting in increased uric acid excretion in exchange for glucose reabsorption. Regardless of this possible mechanism, the underlying comprehensive mechanisms remain poorly elucidated. Therefore, in the present review, a variety of other potential mechanisms will be covered to identify the therapeutic role of SGLT-2i in hyperuricemia.
Collapse
Affiliation(s)
- Meiyuan Dong
- Graduate School of Hebei Medical University, Shijiazhuang, People’s Republic of China
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Huiling Chen
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Song Wen
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Yue Yuan
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Liling Yang
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Dongxiang Xu
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Ligang Zhou
- Graduate School of Hebei Medical University, Shijiazhuang, People’s Republic of China
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling, Shanghai Pudong Hospital, Fudan University, Shanghai, People’s Republic of China
- Correspondence: Ligang Zhou, Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, People’s Republic of China, Tel +8613611927616, Email
| |
Collapse
|
15
|
Lin L, Tan W, Pan X, Tian E, Wu Z, Yang J. Metabolic Syndrome-Related Kidney Injury: A Review and Update. Front Endocrinol (Lausanne) 2022; 13:904001. [PMID: 35813613 PMCID: PMC9261267 DOI: 10.3389/fendo.2022.904001] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 05/09/2022] [Indexed: 11/15/2022] Open
Abstract
Metabolic syndrome (MetS) includes visceral obesity, hyperglycemia, dyslipidemia, and hypertension. The prevalence of MetS is 20-25%, which is an important risk factor for chronic kidney disease (CKD). MetS causes effects on renal pathophysiology, including glomerular hyperfiltration, RAAS, microalbuminuria, profibrotic factors and podocyte injury. This review compares several criteria of MetS and analyzes their differences. MetS and the pathogenesis of CKD includes insulin resistance, obesity, dyslipidemia, inflammation, oxidative stress, and endothelial dysfunction. The intervention of MetS-related renal damage is the focus of this article and includes controlling body weight, hypertension, hyperglycemia, and hyperlipidemia, requiring all components to meet the criteria. In addition, interventions such as endoplasmic reticulum stress, oxidative stress, gut microbiota, body metabolism, appetite inhibition, podocyte apoptosis, and mesenchymal stem cells are reviewed.
Collapse
Affiliation(s)
- Lirong Lin
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University (Gener Hospital), Chongqing, China
| | - Wei Tan
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University (Gener Hospital), Chongqing, China
| | - Xianfeng Pan
- Department of Nephrology, Chongqing Kaizhou District People’s Hospital of Chongqing, Chongqing, China
| | - En Tian
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University (Gener Hospital), Chongqing, China
| | - Zhifeng Wu
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University (Gener Hospital), Chongqing, China
| | - Jurong Yang
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University (Gener Hospital), Chongqing, China
| |
Collapse
|
16
|
Irion CI, Williams M, Capcha JC, Eisenberg T, Lambert G, Takeuchi LM, Seo G, Yousefi K, Kanashiro-Takeuchi R, Webster KA, Young KC, Hare JM, Shehadeh LA. Col4a3-/- Mice on Balb/C Background Have Less Severe Cardiorespiratory Phenotype and SGLT2 Over-Expression Compared to 129x1/SvJ and C57Bl/6 Backgrounds. Int J Mol Sci 2022; 23:6674. [PMID: 35743114 PMCID: PMC9223785 DOI: 10.3390/ijms23126674] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/12/2022] [Accepted: 06/13/2022] [Indexed: 01/27/2023] Open
Abstract
Alport syndrome (AS) is a hereditary renal disorder with no etiological therapy. In the preclinical Col4a3-/- model of AS, disease progression and severity vary depending on mouse strain. The sodium-glucose cotransporter 2 (SGLT2) is emerging as an attractive therapeutic target in cardiac/renal pathologies, but its application to AS remains untested. This study investigates cardiorespiratory function and SGLT2 renal expression in Col4a3-/- mice from three different genetic backgrounds, 129x1/SvJ, C57Bl/6 and Balb/C. male Col4a3-/- 129x1/SvJ mice displayed alterations consistent with heart failure with preserved ejection fraction (HFpEF). Female, but not male, C57Bl/6 and Balb/C Col4a3-/- mice exhibited mild changes in systolic and diastolic function of the heart by echocardiography. Male C57Bl/6 Col4a3-/- mice presented systolic dysfunction by invasive hemodynamic analysis. All strains except Balb/C males demonstrated alterations in respiratory function. SGLT2 expression was significantly increased in AS compared to WT mice from all strains. However, cardiorespiratory abnormalities and SGLT2 over-expression were significantly less in AS Balb/C mice compared to the other two strains. Systolic blood pressure was significantly elevated only in mutant 129x1/SvJ mice. The results provide further evidence for strain-dependent cardiorespiratory and hypertensive phenotype variations in mouse AS models, corroborated by renal SGLT2 expression, and support ongoing initiatives to develop SGLT2 inhibitors for the treatment of AS.
Collapse
Affiliation(s)
- Camila I. Irion
- Department of Medicine, Division of Cardiology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (C.I.I.); (M.W.); (J.C.C.); (T.E.); (G.L.); (J.M.H.)
- Leonard M. Miller School of Medicine, Interdisciplinary Stem Cell Institute, University of Miami, Miami, FL 33136, USA; (L.M.T.); (K.Y.); (R.K.-T.)
| | - Monique Williams
- Department of Medicine, Division of Cardiology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (C.I.I.); (M.W.); (J.C.C.); (T.E.); (G.L.); (J.M.H.)
- Leonard M. Miller School of Medicine, Interdisciplinary Stem Cell Institute, University of Miami, Miami, FL 33136, USA; (L.M.T.); (K.Y.); (R.K.-T.)
| | - Jose Condor Capcha
- Department of Medicine, Division of Cardiology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (C.I.I.); (M.W.); (J.C.C.); (T.E.); (G.L.); (J.M.H.)
- Leonard M. Miller School of Medicine, Interdisciplinary Stem Cell Institute, University of Miami, Miami, FL 33136, USA; (L.M.T.); (K.Y.); (R.K.-T.)
| | - Trevor Eisenberg
- Department of Medicine, Division of Cardiology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (C.I.I.); (M.W.); (J.C.C.); (T.E.); (G.L.); (J.M.H.)
| | - Guerline Lambert
- Department of Medicine, Division of Cardiology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (C.I.I.); (M.W.); (J.C.C.); (T.E.); (G.L.); (J.M.H.)
- Leonard M. Miller School of Medicine, Interdisciplinary Stem Cell Institute, University of Miami, Miami, FL 33136, USA; (L.M.T.); (K.Y.); (R.K.-T.)
| | - Lauro M. Takeuchi
- Leonard M. Miller School of Medicine, Interdisciplinary Stem Cell Institute, University of Miami, Miami, FL 33136, USA; (L.M.T.); (K.Y.); (R.K.-T.)
| | - Grace Seo
- Department of Medical Education, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
| | - Keyvan Yousefi
- Leonard M. Miller School of Medicine, Interdisciplinary Stem Cell Institute, University of Miami, Miami, FL 33136, USA; (L.M.T.); (K.Y.); (R.K.-T.)
- Department of Molecular and Cellular Pharmacology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Rosemeire Kanashiro-Takeuchi
- Leonard M. Miller School of Medicine, Interdisciplinary Stem Cell Institute, University of Miami, Miami, FL 33136, USA; (L.M.T.); (K.Y.); (R.K.-T.)
- Department of Molecular and Cellular Pharmacology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Keith A. Webster
- Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston, TX 77030, USA;
- Department of Ophthalmology, Vascular Biology Institute, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Karen C. Young
- Department of Pediatrics, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
| | - Joshua M. Hare
- Department of Medicine, Division of Cardiology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (C.I.I.); (M.W.); (J.C.C.); (T.E.); (G.L.); (J.M.H.)
- Leonard M. Miller School of Medicine, Interdisciplinary Stem Cell Institute, University of Miami, Miami, FL 33136, USA; (L.M.T.); (K.Y.); (R.K.-T.)
| | - Lina A. Shehadeh
- Department of Medicine, Division of Cardiology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (C.I.I.); (M.W.); (J.C.C.); (T.E.); (G.L.); (J.M.H.)
- Leonard M. Miller School of Medicine, Interdisciplinary Stem Cell Institute, University of Miami, Miami, FL 33136, USA; (L.M.T.); (K.Y.); (R.K.-T.)
| |
Collapse
|