1
|
Gervas-Arruga J, Barba-Romero MÁ, Fernández-Martín JJ, Gómez-Cerezo JF, Segú-Vergés C, Ronzoni G, Cebolla JJ. In Silico Modeling of Fabry Disease Pathophysiology for the Identification of Early Cellular Damage Biomarker Candidates. Int J Mol Sci 2024; 25:10329. [PMID: 39408658 PMCID: PMC11477023 DOI: 10.3390/ijms251910329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 09/19/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Fabry disease (FD) is an X-linked lysosomal disease whose ultimate consequences are the accumulation of sphingolipids and subsequent inflammatory events, mainly at the endothelial level. The outcomes include different nervous system manifestations as well as multiple organ damage. Despite the availability of known biomarkers, early detection of FD remains a medical need. This study aimed to develop an in silico model based on machine learning to identify candidate vascular and nervous system proteins for early FD damage detection at the cellular level. A combined systems biology and machine learning approach was carried out considering molecular characteristics of FD to create a computational model of vascular and nervous system disease. A data science strategy was applied to identify risk classifiers by using 10 K-fold cross-validation. Further biological and clinical criteria were used to prioritize the most promising candidates, resulting in the identification of 36 biomarker candidates with classifier abilities, which are easily measurable in body fluids. Among them, we propose four candidates, CAMK2A, ILK, LMNA, and KHSRP, which have high classification capabilities according to our models (cross-validated accuracy ≥ 90%) and are related to the vascular and nervous systems. These biomarkers show promise as high-risk cellular and tissue damage indicators that are potentially applicable in clinical settings, although in vivo validation is still needed.
Collapse
Affiliation(s)
| | - Miguel Ángel Barba-Romero
- Department of Internal Medicine, Albacete University Hospital, 02006 Albacete, Spain;
- Albacete Medical School, Castilla-La Mancha University, 02006 Albacete, Spain
| | | | - Jorge Francisco Gómez-Cerezo
- Department of Internal Medicine, Infanta Sofía University Hospital, 28702 Madrid, Spain;
- Faculty of Medicine, European University of Madrid, 28670 Madrid, Spain
| | | | | | | |
Collapse
|
2
|
Cebolla JJ, Giraldo P, Gómez J, Montoto C, Gervas-Arruga J. Machine Learning-Driven Biomarker Discovery for Skeletal Complications in Type 1 Gaucher Disease Patients. Int J Mol Sci 2024; 25:8586. [PMID: 39201273 PMCID: PMC11354847 DOI: 10.3390/ijms25168586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/01/2024] [Accepted: 08/05/2024] [Indexed: 09/02/2024] Open
Abstract
Type 1 Gaucher disease (GD1) is a rare, autosomal recessive disorder caused by glucocerebrosidase deficiency. Skeletal manifestations represent one of the most debilitating and potentially irreversible complications of GD1. Although imaging studies are the gold standard, early diagnostic/prognostic tools, such as molecular biomarkers, are needed for the rapid management of skeletal complications. This study aimed to identify potential protein biomarkers capable of predicting the early diagnosis of bone skeletal complications in GD1 patients using artificial intelligence. An in silico study was performed using the novel Therapeutic Performance Mapping System methodology to construct mathematical models of GD1-associated complications at the protein level. Pathophysiological characterization was performed before modeling, and a data science strategy was applied to the predicted protein activity for each protein in the models to identify classifiers. Statistical criteria were used to prioritize the most promising candidates, and 18 candidates were identified. Among them, PDGFB, IL1R2, PTH and CCL3 (MIP-1α) were highlighted due to their ease of measurement in blood. This study proposes a validated novel tool to discover new protein biomarkers to support clinician decision-making in an area where medical needs have not yet been met. However, confirming the results using in vitro and/or in vivo studies is necessary.
Collapse
Affiliation(s)
| | - Pilar Giraldo
- FEETEG, 50006 Zaragoza, Spain;
- Hospital QuirónSalud Zaragoza, 50012 Zaragoza, Spain
| | | | | | | |
Collapse
|
3
|
Palazzuoli A, Severino P, D’Amato A, Myftari V, Tricarico L, Correale M, Dattilo G, Fioretti F, Nodari S. Distinct Profiles and New Pharmacological Targets for Heart Failure with Preserved Ejection Fraction. Rev Cardiovasc Med 2024; 25:270. [PMID: 39139408 PMCID: PMC11317327 DOI: 10.31083/j.rcm2507270] [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: 02/10/2024] [Revised: 04/15/2024] [Accepted: 04/19/2024] [Indexed: 08/15/2024] Open
Abstract
Background Heart failure with preserved ejection fraction (HFpEF) is a multifactorial condition with a variety of pathophysiological causes and morphological manifestations. The inclusion criteria and patient classification have become overly simplistic due to the customary differentiation regarding the ejection fraction (EF) cutoff. EF is considered a measure of systolic function; nevertheless, it only represents a portion of the true contractile state and has been shown to have certain limits due to methodological and hemodynamic irregularities. Methods As a result, broader randomized clinical trials have yet to incorporate the most recent criteria for HFpEF diagnosis, leading to a lack of data consistency and confusion in interpreting the results. The primary variations between the bigger clinical trials published in this context concerning patient selection and echocardiographic characteristics were analyzed. For all these reasons, we aim to clarify the main features and clinical impact of HFpEF in a study combining imaging, bio-humoral analysis, and clinical history to identify the specific subgroups that respond better to tailored treatment. Results Disparate clinical characteristics and a lack of uniform diagnostic standards may cause suboptimal therapeutic feedback. To optimize treatment, we suggest shifting the paradigm from the straightforward EF measurement to a more comprehensive model that considers additional information, such as structural traits, related disorders, and biological and environmental data. Therefore, by evaluating certain echocardiographic and clinical factors, a stepwise diagnostic procedure may be useful in identifying patients at high risk, subjects with early HFpEF, and those with evident HFpEF. Conclusions The present assessment underscores the significance of the precision medicine approach in guaranteeing optimal patient outcomes by providing the best care according to each distinct profile.
Collapse
Affiliation(s)
- Alberto Palazzuoli
- Cardiovascular Diseases Unit, Cardio Thoracic and Vascular Department Le Scotte Hospital University of Siena, 53100 Siena, Italy
| | - Paolo Severino
- Department of Clinical, Internal Anesthesiological and Cardiovascular Sciences University La Sapienza Rome, 00161 Roma, Italy
| | - Andrea D’Amato
- Department of Clinical, Internal Anesthesiological and Cardiovascular Sciences University La Sapienza Rome, 00161 Roma, Italy
| | - Vincenzo Myftari
- Department of Clinical, Internal Anesthesiological and Cardiovascular Sciences University La Sapienza Rome, 00161 Roma, Italy
| | - Lucia Tricarico
- UO Cardiologia Universitaria – Intensive Coronary Unit Policlinico Riuniti Foggia, 71122 Foggia, Italy
| | - Michele Correale
- UO Cardiologia Universitaria – Intensive Coronary Unit Policlinico Riuniti Foggia, 71122 Foggia, Italy
| | - Giuseppe Dattilo
- Cardiology Unit, Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
| | - Francesco Fioretti
- Cardiology Section, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, Spedali Civili Hospital and University of Brescia, 25123 Brescia, Italy
| | - Savina Nodari
- Cardiology Section, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, Spedali Civili Hospital and University of Brescia, 25123 Brescia, Italy
| |
Collapse
|
4
|
Li Y, Zhang Z, Zhang Z, Zheng N, Ding X. Empagliflozin, a sodium-glucose cotransporter inhibitor enhancing mitochondrial action and cardioprotection in metabolic syndrome. J Cell Physiol 2024; 239:e31264. [PMID: 38764242 DOI: 10.1002/jcp.31264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 03/13/2024] [Accepted: 03/18/2024] [Indexed: 05/21/2024]
Abstract
Metabolic syndrome (MetS) has a large clinical population nowadays, usually due to excessive energy intake and lack of exercise. During MetS, excess nutrients stress the mitochondria, resulting in relative hypoxia in tissues and organs, even when blood supply is not interrupted or reduced, making mitochondrial dysfunction a central pathogenesis of cardiovascular disease in the MetS. Sodium-glucose cotransporter 2 inhibitors were designed as a hyperglycemic drug that acts on the renal tubules to block sugar reabsorption in primary urine. Recently they have been shown to have anti-inflammatory and other protective effects on cardiomyocytes in MetS, and have also been recommended in the latest heart failure guidelines as a routine therapy. Among these inhibitors, empagliflozin shows better clinical promise due to less influence from glomerular filtration rate. This review focuses on the mitochondrial mechanisms of empagliflozin, which underlie the anti-inflammatory and recover cellular functions in MetS cardiomyocytes, including stabilizing calcium concentration, mediating metabolic reprogramming, maintaining homeostasis of mitochondrial quantity and quality, stable mitochondrial DNA copy number, and repairing damaged mitochondrial DNA.
Collapse
Affiliation(s)
- Yunhao Li
- Graduate School, China Medical University, Shenyang, China
- Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Zhanming Zhang
- Faculty of Science, The University of Hong Kong, Hong Kong, China
| | - Zheming Zhang
- Graduate School, China Medical University, Shenyang, China
- Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Ningning Zheng
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Xudong Ding
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
5
|
Mashayekhi M, Safa BI, Gonzalez MSC, Kim SF, Echouffo-Tcheugui JB. Systemic and organ-specific anti-inflammatory effects of sodium-glucose cotransporter-2 inhibitors. Trends Endocrinol Metab 2024; 35:425-438. [PMID: 38423898 PMCID: PMC11096060 DOI: 10.1016/j.tem.2024.02.003] [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: 12/03/2023] [Revised: 02/01/2024] [Accepted: 02/02/2024] [Indexed: 03/02/2024]
Abstract
Inflammation plays an essential role and is a common feature in the pathogenesis of many chronic diseases. The exact mechanisms through which sodium-glucose cotransporter-2 (SGLT2) inhibitors achieve their much-acclaimed clinical benefits largely remain unknown. In this review, we detail the systemic and tissue- or organ-specific anti-inflammatory effects of SGLT2 inhibitors using evidence from animal and human studies. We discuss the potential pathways through which SGLT2 inhibitors exert their anti-inflammatory effects, including oxidative stress, mitochondrial, and inflammasome pathways. Finally, we highlight the need for further investigation of the extent of the contribution of the anti-inflammatory effects of SGLT2 inhibition to improvements in cardiometabolic and renal outcomes in clinical studies.
Collapse
Affiliation(s)
- Mona Mashayekhi
- Vanderbilt University Medical Center, Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Nashville, TN, USA
| | - Bilgunay Ilkin Safa
- Vanderbilt University Medical Center, Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Nashville, TN, USA
| | - Matthew S C Gonzalez
- Vanderbilt University Medical Center, Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Nashville, TN, USA
| | - Sangwon F Kim
- Johns Hopkins University School of Medicine, Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Baltimore, MD, USA
| | - Justin B Echouffo-Tcheugui
- Johns Hopkins University School of Medicine, Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Baltimore, MD, USA.
| |
Collapse
|
6
|
Blanot M, Casaroli-Marano RP, Mondéjar-Medrano J, Sallén T, Ramírez E, Segú-Vergés C, Artigas L. Aflibercept Off-Target Effects in Diabetic Macular Edema: An In Silico Modeling Approach. Int J Mol Sci 2024; 25:3621. [PMID: 38612432 PMCID: PMC11011561 DOI: 10.3390/ijms25073621] [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: 01/03/2024] [Revised: 03/08/2024] [Accepted: 03/13/2024] [Indexed: 04/14/2024] Open
Abstract
Intravitreal aflibercept injection (IAI) is a treatment for diabetic macular edema (DME), but its mechanism of action (MoA) has not been completely elucidated. Here, we aimed to explore IAI's MoA and its multi-target nature in DME pathophysiology with an in silico (computer simulation) disease model. We used the Therapeutic Performance Mapping System (Anaxomics Biotech property) to generate mathematical models based on the available scientific knowledge at the time of the study, describing the relationship between the modulation of vascular endothelial growth factor receptors (VEGFRs) by IAI and DME pathophysiological processes. We also undertook an enrichment analysis to explore the processes modulated by IAI, visualized the effectors' predicted protein activity, and specifically evaluated the role of VEGFR1 pathway inhibition on DME treatment. The models simulated the potential pathophysiology of DME and the likely IAI's MoA by inhibiting VEGFR1 and VEGFR2 signaling. The action of IAI through both signaling pathways modulated the identified pathophysiological processes associated with DME, with the strongest effects in angiogenesis, blood-retinal barrier alteration and permeability, and inflammation. VEGFR1 inhibition was essential to modulate inflammatory protein effectors. Given the role of VEGFR1 signaling on the modulation of inflammatory-related pathways, IAI may offer therapeutic advantages for DME through sustained VEGFR1 pathway inhibition.
Collapse
Affiliation(s)
- Morgane Blanot
- Anaxomics Biotech S.L., 08007 Barcelona, Spain; (M.B.); (E.R.); (C.S.-V.); (L.A.)
| | - Ricardo Pedro Casaroli-Marano
- Department of Surgery (FMCS), Universitat de Barcelona, 08007 Barcelona, Spain
- Hospital Clínic de Barcelona (IDIBAPS), Universitat de Barcelona, 08007 Barcelona, Spain
| | | | - Thaïs Sallén
- Bayer Hispania S.L., 08970 Sant Joan Despí, Spain; (J.M.-M.); (T.S.)
| | - Esther Ramírez
- Anaxomics Biotech S.L., 08007 Barcelona, Spain; (M.B.); (E.R.); (C.S.-V.); (L.A.)
| | - Cristina Segú-Vergés
- Anaxomics Biotech S.L., 08007 Barcelona, Spain; (M.B.); (E.R.); (C.S.-V.); (L.A.)
- Research Programme on Biomedical Informatics (GRIB), Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, 08002 Barcelona, Spain
| | - Laura Artigas
- Anaxomics Biotech S.L., 08007 Barcelona, Spain; (M.B.); (E.R.); (C.S.-V.); (L.A.)
| |
Collapse
|
7
|
Nan Tie E, Nanayakkara S, Vizi D, Mariani J, Kaye DM. The Impact of Diabetes on Haemodynamic and Cardiometabolic Responses in Heart Failure With Preserved Ejection Fraction. Heart Lung Circ 2024; 33:376-383. [PMID: 38336542 DOI: 10.1016/j.hlc.2023.12.008] [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: 07/10/2023] [Revised: 11/26/2023] [Accepted: 12/07/2023] [Indexed: 02/12/2024]
Abstract
AIMS Heart failure with preserved ejection (HFpEF) and diabetes mellitus (DM) commonly co-exist. However, it is unclear if DM modifies the haemodynamic and cardiometabolic phenotype in patients with HFpEF. We aimed to interrogate the haemodynamic and cardiometabolic effects of DM in HFpEF. METHODS We compared the haemodynamic and metabolic profiles of non-DM patients and patients with DM-HFpEF at rest and during exercise using right heart catheterisation and mixed venous blood gas analysis. RESULTS Of 181 patients with HFpEF, 37 (20%) had DM. Patients with DM displayed a more adverse exercise haemodynamic response vs HFpEF alone (mean pulmonary arterial pressure: 47 mmHg [interquartile range {IQR} 42-55] vs 42 [38-47], p<0.001; workload indexed pulmonary capillary wedge pressure indexed: 0.80 mmHg/W [0.44-1.23] vs 0.57 [0.43-1.01], p=0.047). HFpEF-DM patients had a lower mixed venous oxygen saturation at rest (70% [IQR 66-73] vs 72 [69-75], p=0.003) and were unable to enhance O2 extraction to the same extent (Δ-28% [-33 to -15] vs -29 [-36 to -21], p=0.029), this occurred at a 22% lower median workload. Resting mixed venous lactate levels were higher in those with DM (1.5 mmol/L [IQR 1.1-1.9] vs 1 [0.9-1.3], p<0.001), and during exercise indexed to workload (0.09 mmol/L/W [0.06-0.13] vs 0.08 [0.05-0.11], p=0.018). CONCLUSION Concurrent diabetes and HFpEF was associated with greater metabolic responses at rest, with enhanced wedge driven pulmonary hypertension and relative lactataemia during exercise without appropriate augmentation of oxygen consumption.
Collapse
Affiliation(s)
- Emilia Nan Tie
- Department of Cardiology, Alfred Hospital, Melbourne, Vic, Australia. http://www.twitter.com/EmiliaNanTie
| | - Shane Nanayakkara
- Department of Cardiology, Alfred Hospital, Melbourne, Vic, Australia; Heart Failure Research Group, Baker Heart & Diabetes Institute, Melbourne, Vic, Australia
| | - Donna Vizi
- Department of Cardiology, Alfred Hospital, Melbourne, Vic, Australia
| | - Justin Mariani
- Department of Cardiology, Alfred Hospital, Melbourne, Vic, Australia; Heart Failure Research Group, Baker Heart & Diabetes Institute, Melbourne, Vic, Australia; Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Vic, Australia
| | - David M Kaye
- Department of Cardiology, Alfred Hospital, Melbourne, Vic, Australia; Heart Failure Research Group, Baker Heart & Diabetes Institute, Melbourne, Vic, Australia; Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Vic, Australia.
| |
Collapse
|
8
|
Sauer J, Marksteiner J, Lilliu E, Hackl B, Todt H, Kubista H, Dostal C, Podesser BK, Kiss A, Koenig X, Hilber K. Empagliflozin treatment rescues abnormally reduced Na + currents in ventricular cardiomyocytes from dystrophin-deficient mdx mice. Am J Physiol Heart Circ Physiol 2024; 326:H418-H425. [PMID: 38099845 PMCID: PMC11219046 DOI: 10.1152/ajpheart.00729.2023] [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: 11/17/2023] [Revised: 12/07/2023] [Accepted: 12/12/2023] [Indexed: 01/23/2024]
Abstract
Cardiac arrhythmias significantly contribute to mortality in Duchenne muscular dystrophy (DMD), a severe muscle illness caused by mutations in the gene encoding for the intracellular protein dystrophin. A major source for arrhythmia vulnerability in patients with DMD is impaired ventricular impulse conduction, which predisposes for ventricular asynchrony, decreased cardiac output, and the development of reentrant circuits. Using the dystrophin-deficient mdx mouse model for human DMD, we previously reported that the lack of dystrophin causes a significant loss of peak Na+ current (INa) in ventricular cardiomyocytes. This finding provided a mechanistic explanation for ventricular conduction defects and concomitant arrhythmias in the dystrophic heart. In the present study, we explored the hypothesis that empagliflozin (EMPA), an inhibitor of sodium/glucose cotransporter 2 in clinical use to treat type II diabetes and nondiabetic heart failure, rescues peak INa loss in dystrophin-deficient ventricular cardiomyocytes. We found that INa of cardiomyocytes derived from mdx mice, which had received clinically relevant doses of EMPA for 4 wk, was restored to wild-type level. Moreover, incubation of isolated mdx ventricular cardiomyocytes with 1 µM EMPA for 24 h significantly increased their peak INa. This effect was independent of Na+-H+ exchanger 1 inhibition by the drug. Our findings imply that EMPA treatment can rescue abnormally reduced peak INa of dystrophin-deficient ventricular cardiomyocytes. Long-term EMPA administration may diminish arrhythmia vulnerability in patients with DMD.NEW & NOTEWORTHY Dystrophin deficiency in cardiomyocytes leads to abnormally reduced Na+ currents. These can be rescued by long-term empagliflozin treatment.
Collapse
Affiliation(s)
- Jakob Sauer
- Department of Neurophysiology and Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Jessica Marksteiner
- Department of Neurophysiology and Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Elena Lilliu
- Department of Neurophysiology and Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Benjamin Hackl
- Department of Neurophysiology and Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Hannes Todt
- Department of Neurophysiology and Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Helmut Kubista
- Department of Neurophysiology and Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Christopher Dostal
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
| | - Bruno K Podesser
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
| | - Attila Kiss
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
| | - Xaver Koenig
- Department of Neurophysiology and Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Karlheinz Hilber
- Department of Neurophysiology and Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
9
|
Yadav J, Ahsan F, Panda P, Mahmood T, Ansari VA, Shamim A. Empagliflozin-A Sodium Glucose Co-transporter-2 Inhibitor: Overview ofits Chemistry, Pharmacology, and Toxicology. Curr Diabetes Rev 2024; 20:e230124226010. [PMID: 38265382 DOI: 10.2174/0115733998271026231127051545] [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: 07/30/2023] [Revised: 09/24/2023] [Accepted: 10/17/2023] [Indexed: 01/25/2024]
Abstract
BACKGROUND Empagliflozin is a sodium glucose co-transporter-2 (SGLT2) inhibitor that has gained significant attention in the treatment of type 2 diabetes mellitus. Understanding its chemistry, pharmacology, and toxicology is crucial for the safe and effective use of this medication. OBJECTIVE This review aims to provide a comprehensive overview of the chemistry, pharmacology, and toxicology of empagliflozin, synthesizing the available literature to present a concise summary of its properties and implications for clinical practice. METHODS A systematic search of relevant databases was conducted to identify studies and articles related to the chemistry, pharmacology, and toxicology of empagliflozin. Data from preclinical and clinical studies, as well as post-marketing surveillance reports, were reviewed to provide a comprehensive understanding of the topic. RESULTS Empagliflozin is a selective SGLT2 inhibitor that works by constraining glucose reabsorption in the kidneys, causing increased urinary glucose elimination. Its unique mechanism of action provides glycemic control, weight reduction, and blood pressure reduction. The drug's chemistry is characterized by its chemical structure, solubility, and stability. Pharmacologically, empagliflozin exhibits favorable pharmacokinetic properties with rapid absorption, extensive protein binding, and renal elimination. Clinical studies have demonstrated its efficacy in improving glycemic control, reducing cardiovascular risks, and preserving renal function. However, adverse effects, for instance, urinary tract infections, genital infections, and diabetic ketoacidosis have been reported. Toxicological studies indicate low potential for organ toxicity, mutagenicity, or carcinogenicity. CONCLUSION Empagliflozin is a promising SGLT2 inhibitor that offers an innovative approach to the treatment of type 2 diabetes mellitus. Its unique action mechanism and favorable pharmacokinetic profile contribute to its efficacy in improving glycemic control and reducing cardiovascular risks. While the drug's safety profile is generally favorable, clinicians should be aware of potential adverse effects and monitor patients closely. More study is required to determine the longterm safety and explore potential benefits in other patient populations. Overall, empagliflozin represents a valuable addition to the armamentarium of antidiabetic medications, offering significant benefits to patients suffering from type 2 diabetes mellitus. This study covers all aspects of empagliflozin, including its history, chemistry, pharmacology, and various clinical studies, case reports, and case series.
Collapse
Affiliation(s)
- Jyoti Yadav
- Department of Pharmacy, Integral University, Dasauli, Kursi Road, Lucknow (U.P.), 226026, India
| | - Farogh Ahsan
- Department of Pharmacy, Integral University, Dasauli, Kursi Road, Lucknow (U.P.), 226026, India
| | - Prabhudatta Panda
- Department of Pharmacy, Institute of Technology & Management, Gorakhpur (U.P.), 226026, India
| | - Tarique Mahmood
- Department of Pharmacy, Integral University, Dasauli, Kursi Road, Lucknow (U.P.), 226026, India
| | - Vaseem Ahamad Ansari
- Department of Pharmacy, Integral University, Dasauli, Kursi Road, Lucknow (U.P.), 226026, India
| | - Arshiya Shamim
- Department of Pharmacy, Integral University, Dasauli, Kursi Road, Lucknow (U.P.), 226026, India
| |
Collapse
|
10
|
Charaya KV, Schekochikhin DY, Nikiforova TV, Dimchishina AS, Soboleva TV, Shkliarov AM, Kusova ZR, Mesitskaya DF, Novikova NA, Andreev DA. The use of Dapagliflozin in Acute Decompensated Heart Failure: Results of the Randomized Study. KARDIOLOGIIA 2023; 63:11-18. [PMID: 37697958 DOI: 10.18087/cardio.2023.8.n2221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/12/2022] [Indexed: 09/13/2023]
Abstract
Aim To determine the effect of sodium-glucose cotransporter-2 inhibitors (SGLT2i) on kidney function in acute decompensated heart failure (ADHF).Material and methods A controlled randomized study on the dapagliflozin treatment in ADHF was performed. Patients were randomized to a main group (standard therapy supplemented with dapagliflozin) or a control group (standard therapy for ADHF). The primary endpoint was the development of acute kidney injury (AKI). 200 patients were included (mean age, 74±12 years; 51% men). 31% of patients had type 2 diabetes mellitus (DM2). Mean left ventricular ejection fraction (LV EF) was 47±14 %; in 44.5% of patients, LV EF was less than 45%. Median concentration of N-terminal pro-brain natriuretic peptide (NT-proBNP) was 5225 [3120; 9743] pg / ml, glomerular filtration rate (GFR) was 51 [38; 64] ml / min / 1.73 m2.Results In-hospital mortality was 6.5%. Analysis of the dynamics of body weight loss showed significant differences (4200 [2925; 6300] g vs. 3000 [1113; 4850] g; p=0.011) in favor of the dapagliflozin group. The requirement for increasing the daily dose of furosemide and adding an another class diuretic (thiazide or acetazolamide) did not differ between the groups. However, median furosemide dose during the stay in the hospital was lower in the dapagliflozin group (80 [67; 120] mg vs. 102 [43; 120] mg; p=0.016). At 48 hours after randomization, GFR significantly decreased in the dapagliflozin group (-5.5 [-11; 3] ml/min/ 1.73 m2) compared to the control group (-0.3 [-4; 5] ml / min/1.73 m2, р=0.012). Despite this, GFR did not differ between the groups at discharge (51 [41; 66] ml/min/1.73 m2 and 49 [38; 67] ml/min/1.73 m2, respectively; p = 0.84). In the dapagliflozin group, frequency of AKI episodes was not increased compared to the control group (13 and 9.4%, respectively; p = 0.45).Conclusion The dapagliflozin treatment in ADHF is associated with more pronounced body weight loss and lower average doses of loop diuretics during the period of stay in the hospital, with no associated clinically significant impairment of renal function.
Collapse
Affiliation(s)
- K V Charaya
- Sechenov First Moscow State Medical University, Moscow
| | | | | | | | - T V Soboleva
- Sechenov First Moscow State Medical University, Moscow
| | - A M Shkliarov
- Sechenov First Moscow State Medical University, Moscow
| | - Z R Kusova
- Sechenov First Moscow State Medical University, Moscow
| | | | - N A Novikova
- Sechenov First Moscow State Medical University, Moscow
| | - D A Andreev
- Sechenov First Moscow State Medical University, Moscow
| |
Collapse
|
11
|
Mele M, Imbrici P, Mele A, Togo MV, Dinoi G, Correale M, Brunetti ND, Nicolotti O, De Luca A, Altomare CD, Liantonio A, Amoroso N. Short-term anti-remodeling effects of gliflozins in diabetic patients with heart failure and reduced ejection fraction: an explainable artificial intelligence approach. Front Pharmacol 2023; 14:1175606. [PMID: 37361206 PMCID: PMC10289166 DOI: 10.3389/fphar.2023.1175606] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 05/26/2023] [Indexed: 06/28/2023] Open
Abstract
Introduction: Sodium-glucose cotransporter type 2 inhibitors (SGLT2i), gliflozins, play an emerging role for the treatment of heart failure with reduced left ventricular ejection fraction (HFrEF). Nevertheless, the effects of SGLT2i on ventricular remodeling and function have not been completely understood yet. Explainable artificial intelligence represents an unprecedented explorative option to clinical research in this field. Based on echocardiographic evaluations, we identified some key clinical responses to gliflozins by employing a machine learning approach. Methods: Seventy-eight consecutive diabetic outpatients followed for HFrEF were enrolled in the study. Using a random forests classification, a single subject analysis was performed to define the profile of patients treated with gliflozins. An explainability analysis using Shapley values was used to outline clinical parameters that mostly improved after gliflozin therapy and machine learning runs highlighted specific variables predictive of gliflozin response. Results: The five-fold cross-validation analyses showed that gliflozins patients can be identified with a 0.70 ± 0.03% accuracy. The most relevant parameters distinguishing gliflozins patients were Right Ventricular S'-Velocity, Left Ventricular End Systolic Diameter and E/e' ratio. In addition, low Tricuspid Annular Plane Systolic Excursion values along with high Left Ventricular End Systolic Diameter and End Diastolic Volume values were associated to lower gliflozin efficacy in terms of anti-remodeling effects. Discussion: In conclusion, a machine learning analysis on a population of diabetic patients with HFrEF showed that SGLT2i treatment improved left ventricular remodeling, left ventricular diastolic and biventricular systolic function. This cardiovascular response may be predicted by routine echocardiographic parameters, with an explainable artificial intelligence approach, suggesting a lower efficacy in case of advanced stages of cardiac remodeling.
Collapse
Affiliation(s)
- Marco Mele
- Department of Pharmacy-Drug Sciences, University of Bari, Bari, Italy
- University Hospital Policlinico Riuniti, Foggia, Italy
| | - Paola Imbrici
- Department of Pharmacy-Drug Sciences, University of Bari, Bari, Italy
| | - Antonietta Mele
- Department of Pharmacy-Drug Sciences, University of Bari, Bari, Italy
| | | | - Giorgia Dinoi
- Department of Pharmacy-Drug Sciences, University of Bari, Bari, Italy
| | | | | | - Orazio Nicolotti
- Department of Pharmacy-Drug Sciences, University of Bari, Bari, Italy
| | - Annamaria De Luca
- Department of Pharmacy-Drug Sciences, University of Bari, Bari, Italy
| | | | | | - Nicola Amoroso
- Department of Pharmacy-Drug Sciences, University of Bari, Bari, Italy
- National Institute of Nuclear Physics, Section of Bari, Bari, Italy
| |
Collapse
|
12
|
Gómez J, Artigas L, Valls R, Gervas-Arruga J. An in silico approach to identify early damage biomarker candidates in metachromatic leukodystrophy. Mol Genet Metab Rep 2023; 35:100974. [PMID: 37275681 PMCID: PMC10233284 DOI: 10.1016/j.ymgmr.2023.100974] [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: 10/06/2022] [Revised: 04/06/2023] [Accepted: 04/07/2023] [Indexed: 06/07/2023] Open
Abstract
Metachromatic leukodystrophy (MLD) is a rare, autosomal recessive lysosomal storage disease. Deficient activity of arylsulfatase A causes sulfatides to accumulate in cells of different tissues, including those in the central and peripheral nervous systems, leading to progressive demyelination and neurodegeneration. Although there is some association between specific arylsulfatase A alleles and disease severity, genotype-phenotype correlations are not fully understood. We aimed to identify biomarker candidates of early tissue damage in MLD using a modeling approach based on systems biology. A review of the literature was performed in an initial disease characterization step, allowing identification of pathophysiological processes involved in MLD and proteins relating to these processes. Three mathematical models were generated to simulate different stages of MLD at the molecular level: an early pro-inflammatory stage model (including only processes considered to be active in the early stages of disease), a pre-demyelination stage model (including additional processes that are active after some disease progression), and a demyelination stage model (in which all pathophysiological processes are active). The models evaluated 3457 proteins of interest, individually and by pairs through data mining techniques, applying five filters to prioritize biomarkers that could differentiate between the models. Sixteen potential biomarkers were identified, including effectors relating to mitochondrial dysfunction, remyelination, and neurodegeneration. The findings were corroborated in a gene expression data set from T lymphocytes of patients with MLD; all candidates formed combinations that were able to distinguish patients with MLD from controls, and all but one candidate distinguished late-infantile MLD from juvenile MLD as part of a combinatorial biomarker pair. In particular, pro-neuregulin-1 appeared as differential on all comparisons (patients with MLD vs controls and within clinical subtypes); casein kinase II subunit alpha was detected as a potential individual marker within clinical subtypes. These findings provide a panel of biomarker candidates suitable for experimental validation and highlight the utility of mathematical models to identify biomarker candidates of early tissue damage in MLD with a high degree of accuracy and sensitivity.
Collapse
|
13
|
Fender AC, Dobrev D. Evolving insights into the pleiotropic cardioprotective mechanisms of SGLT2 inhibitors. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:589-592. [PMID: 36943455 PMCID: PMC10042952 DOI: 10.1007/s00210-023-02459-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Affiliation(s)
- Anke C Fender
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany.
| |
Collapse
|
14
|
Is Dapagliflozin a Cardiac Anti-Aging Drug? JACC. HEART FAILURE 2023; 11:305-306. [PMID: 36647928 DOI: 10.1016/j.jchf.2022.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 10/11/2022] [Indexed: 12/12/2022]
|
15
|
Pugliese NR, Pellicori P, Filidei F, De Biase N, Maffia P, Guzik TJ, Masi S, Taddei S, Cleland JGF. Inflammatory pathways in heart failure with preserved left ventricular ejection fraction: implications for future interventions. Cardiovasc Res 2023; 118:3536-3555. [PMID: 36004819 PMCID: PMC9897694 DOI: 10.1093/cvr/cvac133] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/26/2022] [Accepted: 08/10/2022] [Indexed: 02/07/2023] Open
Abstract
Many patients with symptoms and signs of heart failure have a left ventricular ejection fraction ≥50%, termed heart failure with preserved ejection fraction (HFpEF). HFpEF is a heterogeneous syndrome mainly affecting older people who have many other cardiac and non-cardiac conditions that often cast doubt on the origin of symptoms, such as breathlessness, or signs, such as peripheral oedema, rendering them neither sensitive nor specific to the diagnosis of HFpEF. Currently, management of HFpEF is mainly directed at controlling symptoms and treating comorbid conditions such as hypertension, atrial fibrillation, anaemia, and coronary artery disease. HFpEF is also characterized by a persistent increase in inflammatory biomarkers. Inflammation may be a key driver of the development and progression of HFpEF and many of its associated comorbidities. Detailed characterization of specific inflammatory pathways may provide insights into the pathophysiology of HFpEF and guide its future management. There is growing interest in novel therapies specifically designed to target deregulated inflammation in many therapeutic areas, including cardiovascular disease. However, large-scale clinical trials investigating the effectiveness of anti-inflammatory treatments in HFpEF are still lacking. In this manuscript, we review the role of inflammation in HFpEF and the possible implications for future trials.
Collapse
Affiliation(s)
| | - Pierpaolo Pellicori
- Robertson Institute of Biostatistics and Clinical Trials Unit, University of Glasgow, Glasgow G12 8QQ, UK
| | - Francesco Filidei
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56126, Italy
| | - Nicolò De Biase
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56126, Italy
| | - Pasquale Maffia
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples 80138, Italy
| | - Tomasz J Guzik
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
- Department of Internal and Agricultural Medicine, Jagiellonian University, Collegium Medicum, Krakow 31-008, Poland
| | - Stefano Masi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56126, Italy
| | - Stefano Taddei
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56126, Italy
| | - John G F Cleland
- Robertson Institute of Biostatistics and Clinical Trials Unit, University of Glasgow, Glasgow G12 8QQ, UK
| |
Collapse
|
16
|
Hasan MM, Islam MU, Sadeq MJ, Fung WK, Uddin J. Review on the Evaluation and Development of Artificial Intelligence for COVID-19 Containment. SENSORS (BASEL, SWITZERLAND) 2023; 23:527. [PMID: 36617124 PMCID: PMC9824505 DOI: 10.3390/s23010527] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/23/2022] [Accepted: 12/29/2022] [Indexed: 06/17/2023]
Abstract
Artificial intelligence has significantly enhanced the research paradigm and spectrum with a substantiated promise of continuous applicability in the real world domain. Artificial intelligence, the driving force of the current technological revolution, has been used in many frontiers, including education, security, gaming, finance, robotics, autonomous systems, entertainment, and most importantly the healthcare sector. With the rise of the COVID-19 pandemic, several prediction and detection methods using artificial intelligence have been employed to understand, forecast, handle, and curtail the ensuing threats. In this study, the most recent related publications, methodologies and medical reports were investigated with the purpose of studying artificial intelligence's role in the pandemic. This study presents a comprehensive review of artificial intelligence with specific attention to machine learning, deep learning, image processing, object detection, image segmentation, and few-shot learning studies that were utilized in several tasks related to COVID-19. In particular, genetic analysis, medical image analysis, clinical data analysis, sound analysis, biomedical data classification, socio-demographic data analysis, anomaly detection, health monitoring, personal protective equipment (PPE) observation, social control, and COVID-19 patients' mortality risk approaches were used in this study to forecast the threatening factors of COVID-19. This study demonstrates that artificial-intelligence-based algorithms integrated into Internet of Things wearable devices were quite effective and efficient in COVID-19 detection and forecasting insights which were actionable through wide usage. The results produced by the study prove that artificial intelligence is a promising arena of research that can be applied for disease prognosis, disease forecasting, drug discovery, and to the development of the healthcare sector on a global scale. We prove that artificial intelligence indeed played a significantly important role in helping to fight against COVID-19, and the insightful knowledge provided here could be extremely beneficial for practitioners and research experts in the healthcare domain to implement the artificial-intelligence-based systems in curbing the next pandemic or healthcare disaster.
Collapse
Affiliation(s)
- Md. Mahadi Hasan
- Department of Computer Science and Engineering, Asian University of Bangladesh, Ashulia 1349, Bangladesh
| | - Muhammad Usama Islam
- School of Computing and Informatics, University of Louisiana at Lafayette, Lafayette, LA 70504, USA
| | - Muhammad Jafar Sadeq
- Department of Computer Science and Engineering, Asian University of Bangladesh, Ashulia 1349, Bangladesh
| | - Wai-Keung Fung
- Department of Applied Computing and Engineering, Cardiff School of Technologies, Cardiff Metropolitan University, Cardiff CF5 2YB, UK
| | - Jasim Uddin
- Department of Applied Computing and Engineering, Cardiff School of Technologies, Cardiff Metropolitan University, Cardiff CF5 2YB, UK
| |
Collapse
|
17
|
Aguiar-Neves I, Santos-Ferreira D, Fontes-Carvalho R. SGLT2 Inhibition in Heart Failure with Preserved Ejection Fraction - The New Frontier. Rev Cardiovasc Med 2023; 24:1. [PMID: 39076855 PMCID: PMC11270412 DOI: 10.31083/j.rcm2401001] [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: 09/29/2022] [Revised: 11/27/2022] [Accepted: 12/02/2022] [Indexed: 07/31/2024] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a complex clinical syndrome with high morbidity and increasing socio-economic burden, compounded by the lack of effective treatment options available to treat this disease. Sodium-glucose cotransporter-2 (SGLT2) inhibitors have previously been shown to improve cardiovascular and renal outcomes in patients with type 2 diabetes and patients with heart failure with reduced ejection fraction (HFrEF). Recent major clinical trials with SGLT2 inhibitors, both empagliflozin and dapagliflozin, have now demonstrated improved cardiovascular outcomes in patients with HFpEF and a significant reduction in heart failure hospitalization. Current evidence shows a potential for cardiovascular benefits with SGLT2 inhibition that is consistent across the spectrum of ejection fraction, age, New York Heart Association (NYHA) functional class, natriuretic peptide levels and diabetes status. Although the cardioprotective mechanisms behind SGLT2 inhibition remain unclear, ongoing clinical studies aim to clarify the role of SGLT2 inhibitors on biomarkers of cardiac metabolism, diastolic function and exercise capacity in HFpEF. This article analyzes current clinical evidence from randomized controlled trials and meta-analyses and explores the potential cardioprotective mechanisms of SGLT2 inhibitors, while also looking towards the future of SGLT2 inhibition in HFpEF.
Collapse
Affiliation(s)
- Inês Aguiar-Neves
- Cardiology Department, Centro Hospitalar de Vila Nova de Gaia/Espinho, 4434-502 Vila Nova de Gaia, Portugal
| | - Diogo Santos-Ferreira
- Cardiology Department, Centro Hospitalar de Vila Nova de Gaia/Espinho, 4434-502 Vila Nova de Gaia, Portugal
- Cardiovascular R&D Centre – UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, 4200-450 Porto, Portugal
| | - Ricardo Fontes-Carvalho
- Cardiology Department, Centro Hospitalar de Vila Nova de Gaia/Espinho, 4434-502 Vila Nova de Gaia, Portugal
- Cardiovascular R&D Centre – UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, 4200-450 Porto, Portugal
| |
Collapse
|
18
|
Frampton JE. Empagliflozin: A Review in Symptomatic Chronic Heart Failure. Drugs 2022; 82:1591-1602. [DOI: 10.1007/s40265-022-01778-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2022] [Indexed: 11/16/2022]
|
19
|
Lin K, Yang N, Luo W, Qian JF, Zhu WW, Ye SJ, Yuan CX, Xu DY, Liang G, Huang WJ, Shan PR. Direct cardio-protection of Dapagliflozin against obesity-related cardiomyopathy via NHE1/MAPK signaling. Acta Pharmacol Sin 2022; 43:2624-2635. [PMID: 35217813 PMCID: PMC9525284 DOI: 10.1038/s41401-022-00885-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 02/06/2022] [Indexed: 12/22/2022] Open
Abstract
Obesity is an important independent risk factor for cardiovascular diseases, remaining an important health concern worldwide. Evidence shows that saturated fatty acid-induced inflammation in cardiomyocytes contributes to obesity-related cardiomyopathy. Dapagliflozin (Dapa), a selective SGLT2 inhibitor, exerts a favorable preventive activity in heart failure. In this study, we investigated the protective effect of Dapa against cardiomyopathy caused by high fat diet-induced obesity in vitro and in vivo. Cultured rat cardiomyocyte H9c2 cells were pretreated with Dapa (1, 2.5 μM) for 1.5 h, followed by treatment with palmitic acid (PA, 200 μM) for 24 h. We showed that Dapa pretreatment concentration-dependently attenuated PA-induced cell hypertrophy, fibrosis and apoptosis. Transcriptome analysis revealed that inhibition of PA-activated MAPK/AP-1 pathway contributed to the protective effect of Dapa in H9c2 cells, and this was confirmed by anti-p-cJUN fluorescence staining assay. Using surface plasmon resonance analysis we found the direct binding of Dapa with NHE1. Gain and loss of function experiments further demonstrated the role of NHE1 in the protection of Dapa. In vivo experiments were conducted in mice fed a high fat diet for 5 months. The mice were administered Dapa (1 mg·kg-1·d-1, i.g.) in the last 2 months. Dapa administration significantly reduced the body weight and improved the serum lipid profiles. Dapa administration also alleviated HFD-induced cardiac dysfunction and cardiac aberrant remodeling via inhibiting MAPK/AP-1 pathway and ameliorating cardiac inflammation. In conclusion, Dapa exerts a direct protective effect against saturated fatty acid-induced cardiomyocyte injury in addition to the lowering effect on serum lipids. The protective effect results from negative regulating MAPK/AP-1 pathway in a NHE1-dependent way. The current study highlights the potential of clinical use of Dapa in the prevention of obesity-related cardiac dysfunction.
Collapse
Affiliation(s)
- Ke Lin
- Department of Cardiology, The Key Lab of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Na Yang
- Department of Cardiology, The Key Lab of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Wu Luo
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Jin-Fu Qian
- Department of Cardiology, The Key Lab of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Wei-Wei Zhu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Shi-Ju Ye
- Department of Cardiology, The Key Lab of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Chen-Xin Yuan
- Department of Cardiology, The Key Lab of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Di-Yun Xu
- Department of Cardiology, The Key Lab of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Wei-Jian Huang
- Department of Cardiology, The Key Lab of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China.
| | - Pei-Ren Shan
- Department of Cardiology, The Key Lab of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
20
|
Jiang Y, Xie J. Cost-effectiveness of adding empagliflozin to the standard therapy for Heart Failure with Preserved Ejection Fraction from the perspective of healthcare systems in China. Front Cardiovasc Med 2022; 9:946399. [PMID: 36119747 PMCID: PMC9479072 DOI: 10.3389/fcvm.2022.946399] [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: 05/17/2022] [Accepted: 08/11/2022] [Indexed: 11/13/2022] Open
Abstract
Background The Empagliflozin Outcome Trial in Patients with Chronic Heart Failure with Preserved Ejection Fraction (EMPEROR-Preserved) is the first randomized controlled trial to provide promising evidence on the efficacy of adding empagliflozin to the standard therapy in patients with Heart Failure with Preserved Ejection Fraction (HFpEF), but the cost-effectiveness of add-on empagliflozin treatment remains unclear. Method A Markov model using data from the EMPEROR-Preserved trial and national database was constructed to assess lifetime costs and utility from a China healthcare system perspective. The time horizon was 10 years and a 5% discount rate was applied. Incremental cost-effectiveness ratio (ICER) against willingness to pay (WTP) threshold was performed to evaluate the cost-effectiveness. A series of sensitivity analyses was applied to ensure the robustness of the results. Results Compared to standard therapy, the increased cost of adding empagliflozin from $4,645.23 to $5,916.50 was associated with a quality-adjusted life years (QALYs) gain from 4.70 to 4.81, projecting an ICER of $11,292.06, which was lower than a WTP threshold of $12,652.5. Univariate sensitivity analysis revealed that the parameters with the largest impact on ICER were cardiovascular mortality in both groups, followed by the cost of empagliflozin and the cost of hospitalization for heart failure. Probabilistic sensitivity analysis indicated that when the WTP threshold was $12,652.5 and $37,957.5, the probability of being cost-effective for adding empagliflozin was 52.7% and 67.6%, respectively. Scenario analysis demonstrated that the cost of empagliflozin, the cost of hospitalization for heart failure, NYHA functional classes, and time horizon had a greater impact on the ICER. Conclusion At a WTP threshold of $12,652.5, the add-on empagliflozin treatment for HFpEF was cost-effective in healthcare systems in China, which promoted the rational use of empagliflozin for HFpEF.
Collapse
Affiliation(s)
| | - Jun Xie
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
21
|
Karmazyn M, Pierce GN, Fliegel L. The Remaining Conundrum of the Role of the Na +/H + Exchanger Isoform 1 (NHE1) in Cardiac Physiology and Pathology: Can It Be Rectified? Rev Cardiovasc Med 2022; 23:284. [PMID: 39076631 PMCID: PMC11266974 DOI: 10.31083/j.rcm2308284] [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: 06/08/2022] [Revised: 06/29/2022] [Accepted: 07/08/2022] [Indexed: 07/31/2024] Open
Abstract
The mammalian Na + /H + exchanger (NHE) is a family of ubiquitous membrane proteins present in humans. Isoform one (NHE1) is present on the plasma membrane and regulates intracellular pH by removal of one intracellular proton in exchange for one extracellular sodium thus functioning as an electroneutral process. Human NHE1 has a 500 amino acid membrane domain plus a C-terminal 315 amino acid, regulatory cytosolic tail. It is regulated through a cytosolic regulatory C-terminal tail which is subject to phosphorylation and is modulated by proteins and lipids. Substantial evidence has implicated NHE1 activity in both myocardial ischemia and reperfusion damage and myocardial remodeling resulting in heart failure. Experimental data show excellent cardioprotection with NHE1 inhibitors although results from clinical results have been mixed. In cardiac surgery patients receiving the NHE1 inhibitor cariporide, subgroups showed beneficial effects of treatment. However, in one trial this was associated with a significantly increased incidence of ischemic strokes. This likely reflected both inappropriate dosing regimens as well as overly high drug doses. We suggest that further progress towards NHE1 inhibition as a treatment for cardiovascular disease is warranted through the development of novel compounds to inhibit NHE1 that are structurally different than those previously used in compromised clinical trials. Some novel pyrazinoyl guanidine inhibitors of NHE1 are already in development and the recent elucidation of the three-dimensional structure of the NHE1 protein and identity of the inhibitor binding site may facilitate development. An alternative approach may also be to control the endogenous regulation of activity of NHE1, which is activated in disease.
Collapse
Affiliation(s)
- Morris Karmazyn
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Grant N. Pierce
- Institute of Cardiovascular Sciences, Albrechtsen Research Centre, St. Boniface Hospital, and Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R2H 2A6, Canada
| | - Larry Fliegel
- Department of Biochemistry, University Alberta, Edmonton, AB T6G 2H7, Canada
| |
Collapse
|
22
|
Díaz-Beyá M, García-Fortes M, Valls R, Artigas L, Gómez-Casares MT, Montesinos P, Sánchez-Guijo F, Coma M, Vendranes M, Martínez-López J. A Systems Biology- and Machine Learning-Based Study to Unravel Potential Therapeutic Mechanisms of Midostaurin as a Multitarget Therapy on FLT3-Mutated AML. BIOMEDINFORMATICS 2022; 2:375-397. [DOI: 10.3390/biomedinformatics2030024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Acute myeloid leukemia (AML), a hematologic malignancy that results in bone marrow failure, is the most common acute leukemia in adults. The presence of FMS-related tyrosine kinase 3 (FLT3) mutations is associated with a poor prognosis, making the evaluation of FLT3-inhibitors an imperative goal in clinical trials. Midostaurin was the first FLT3-inhibitor approved by the FDA and EMA for the treatment of FLT3-mutated AML, and it showed a significant improvement in overall survival for newly diagnosed patients treated with midostaurin, in combination with standard chemotherapy (RATIFY study). The main interest of midostaurin has been the FLT3-specific inhibition, but little is known about its role as a multikinase inhibitor and whether it may be used in relapse and maintenance therapy. Here, we used systems biology- and machine learning-based approaches to deepen the potential benefits of the multitarget activity of midostaurin and to better understand its anti-leukemic effect on FLT3-mutated AML. The resulting in silico study revealed that the multikinase activity of midostaurin may play a role in the treatment’s efficacy. Additionally, we propose a series of molecular mechanisms that support a potential benefit of midostaurin as a maintenance therapy in FLT3-mutated AML, by regulating the microenvironment. The obtained results are backed up using independent gene expression data.
Collapse
Affiliation(s)
- Marina Díaz-Beyá
- Department of Hematology, Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Myeloid Neoplasms Group, Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Spain
| | - María García-Fortes
- Hematology Department, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain
| | - Raquel Valls
- Molecular Health Department, Anaxomics Biotech SL, 08007 Barcelona, Spain
| | - Laura Artigas
- Molecular Health Department, Anaxomics Biotech SL, 08007 Barcelona, Spain
| | - Mª Teresa Gómez-Casares
- Hematology Service, Hospital Universitario Insular Materno-Infantil, 35016 Las Palmas de Gran Canaria, Spain
| | - Pau Montesinos
- Departament of Hematology, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain
| | - Fermín Sánchez-Guijo
- Cancer Research Center (CIC), Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, Universidad de Salamanca, 37007 Salamanca, Spain
| | - Mireia Coma
- Molecular Health Department, Anaxomics Biotech SL, 08007 Barcelona, Spain
| | | | - Joaquín Martínez-López
- Hospital 12 de Octubre. Universidad Complutense. CNIO. Centro de Investigación Biomédica en Red-Oncología (CIBERONC), Instituto Carlos III, 28041 Madrid, Spain
| |
Collapse
|
23
|
Bayes-Genis A, Aimo A, Lupón J. Empagliflozin in HFpEF/HFmrEF: Prognostic Benefit Confirmed with Different Endpoint Definitions. Eur J Heart Fail 2022; 24:1406-1409. [PMID: 35781908 DOI: 10.1002/ejhf.2597] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/19/2022] [Accepted: 06/21/2022] [Indexed: 11/05/2022] Open
Affiliation(s)
- Antoni Bayes-Genis
- Institut del Cor, Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, Spain.,Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain.,CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - Alberto Aimo
- Scuola Superiore Sant'Anna, Pisa, Italy.,Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Josep Lupón
- Institut del Cor, Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, Spain
| |
Collapse
|
24
|
Charaya K, Shchekochikhin D, Andreev D, Dyachuk I, Tarasenko S, Poltavskaya M, Mesitskaya D, Bogdanova A, Ananicheva N, Kuzub A. Impact of dapagliflozin treatment on renal function and diuretics use in acute heart failure: a pilot study. Open Heart 2022; 9:e001936. [PMID: 35609943 PMCID: PMC9131063 DOI: 10.1136/openhrt-2021-001936] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 05/02/2022] [Indexed: 11/04/2022] Open
Abstract
OBJECTIVE To determine the impact of sodium-dependent glucose type 2 cotransporter inhibitors on the renal function in acute heart failure. METHODS In a single-centre, controlled, randomised study, patients were prescribed dapagliflozin in addition to standard therapy, or were in receipt of standard therapy. The prespecified outcome was renal function deterioration; the secondary outcomes were the development of resistance to diuretics, weight loss, death during hospitalisation and the rehospitalisation or death for any reason within 30 days following discharge. RESULTS 102 patients were included (73.4±11.7 years, 57.8% men). The average left ventricular ejection fraction was 44.9%±14.7%, the average N-terminal prohormone of brain natriuretic peptide (NT-proBNP) was 4706 (1757; 11 244) pg/mL, the average estimated glomerular filtration rate (eGFR) was 51.6±19.5 mL/min. eGFR decreased 48 hours after randomisation in the dapagliflozin group (-4.2 (-11.03; 2.28) mL/min vs 0.3 (-6; 6) mL/min; p=0.04) but did not differ between the groups on discharge (54.71±19.18 mL/min and 58.92±24.65 mL/min; p=0.36). The incidence of worsening renal function did not differ (34.4% vs 15.2%; p=0.07). In the dapagliflozin group, there was less tendency to increase the dose of loop diuretics (14% vs 30%; p=0.048), lower average doses of loop diuretics (78.46±38.95 mg/day vs 102.82±31.26 mg/day; p=0.001) and more significant weight loss (4100 (2950; 5750) g vs 3000 (1380; 4650) g; p=0.02). In-hospital mortality was 7.8% (4(8%) in the dapagliflozin and 4 (7.7%) in the control group (p=0.95). The number of deaths within 30 days following discharge in the dapagliflozin group and in the control group was 9 (19%) and 12 (25%), p=0.55; the number of rehospitalisations was 14 (29%) and 17 (35%), respectively (p=0.51). CONCLUSION The use of dapagliflozin was associated with a more pronounced weight loss and less need to increase diuretic therapy without significant deterioration of the renal function. Dapagliflozin did not improve the in-hospital and 30-day prognosis after discharge. TRIAL REGISTRATION NUMBER N04778787.
Collapse
Affiliation(s)
- Kristina Charaya
- Department of Cardiology, Functional and Ultrasound Diagnostics, Sechenov University, Moskva, Moskva, Russian Federation
| | - Dmitry Shchekochikhin
- Department of Cardiology, Functional and Ultrasound Diagnostics, Sechenov University, Moskva, Moskva, Russian Federation
| | - Denis Andreev
- Department of Cardiology, Functional and Ultrasound Diagnostics, Sechenov University, Moskva, Moskva, Russian Federation
| | - Irina Dyachuk
- Department of Cardiology, Functional and Ultrasound Diagnostics, Sechenov University, Moskva, Moskva, Russian Federation
| | - Svetlana Tarasenko
- Department of Cardiology, Functional and Ultrasound Diagnostics, Sechenov University, Moskva, Moskva, Russian Federation
| | - Maria Poltavskaya
- Department of Cardiology, Functional and Ultrasound Diagnostics, Sechenov University, Moskva, Moskva, Russian Federation
| | - Dinara Mesitskaya
- Department of Cardiology, Functional and Ultrasound Diagnostics, Sechenov University, Moskva, Moskva, Russian Federation
| | - Alexandra Bogdanova
- Department of Cardiology, Functional and Ultrasound Diagnostics, Sechenov University, Moskva, Moskva, Russian Federation
| | - Natalia Ananicheva
- Department of Cardiology, Functional and Ultrasound Diagnostics, Sechenov University, Moskva, Moskva, Russian Federation
| | - Alina Kuzub
- Department of Cardiology, Functional and Ultrasound Diagnostics, Sechenov University, Moskva, Moskva, Russian Federation
| |
Collapse
|
25
|
Wichaiyo S, Saengklub N. Alterations of sodium-hydrogen exchanger 1 function in response to SGLT2 inhibitors: what is the evidence? Heart Fail Rev 2022; 27:1973-1990. [PMID: 35179683 DOI: 10.1007/s10741-022-10220-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/02/2022] [Indexed: 02/07/2023]
Abstract
This review summarizes and describes the current evidence addressing how sodium-glucose cotransporter 2 (SGLT2) inhibitors alter the function of sodium-hydrogen exchanger 1 (NHE-1), in association with their protective effects against adverse cardiovascular events. In the heart, SGLT2 inhibitors modulate the function of NHE-1 (either by direct inhibition or indirect attenuation of protein expression), which promotes cardiac contraction and an enhanced energy supply, in association with improved mitochondrial function, reduced inflammation/oxidative/endoplasmic reticulum stress, and attenuated fibrosis and apoptotic/autophagic cell death. The vasodilating effect of SGLT2 inhibitors has also been proposed due to NHE-1 inhibition. Moreover, platelet-expressed NHE-1 might serve as a target for SGLT2 inhibitors, since these drugs and selective NHE-1 inhibitors produce comparable activity against adenosine diphosphate-stimulated platelet activation. Overall, it is promising that the modulation of the functions of NHE-1 on the heart, blood vessels, and platelets may act as a contributing pathway for the cardiovascular benefits of SGLT2 inhibitors in diabetes and heart failure.
Collapse
Affiliation(s)
- Surasak Wichaiyo
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, 447 Sri-Ayuthaya Road, Rajathevi, Bangkok, 10400, Thailand. .,Centre of Biopharmaceutical Science for Healthy Ageing, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand.
| | - Nakkawee Saengklub
- Centre of Biopharmaceutical Science for Healthy Ageing, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand.,Department of Physiology, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand
| |
Collapse
|
26
|
Kolesnik E, Scherr D, Rohrer U, Benedikt M, Manninger M, Sourij H, von Lewinski D. SGLT2 Inhibitors and Their Antiarrhythmic Properties. Int J Mol Sci 2022; 23:1678. [PMID: 35163599 PMCID: PMC8835896 DOI: 10.3390/ijms23031678] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 01/23/2022] [Accepted: 01/24/2022] [Indexed: 12/13/2022] Open
Abstract
Sodium-glucose cotransporter 2 (SGLT2) inhibitors are gaining ground as standard therapy for heart failure with a class-I recommendation in the recently updated heart failure guidelines from the European Society of Cardiology. Different gliflozins have shown impressive beneficial effects in patients with and without diabetes mellitus type 2, especially in reducing the rates for hospitalization for heart failure, yet little is known on their antiarrhythmic properties. Atrial and ventricular arrhythmias were reported by clinical outcome trials with SGLT2 inhibitors as adverse events, and SGLT2 inhibitors seemed to reduce the rate of arrhythmias compared to placebo treatment in those trials. Mechanistical links are mainly unrevealed, since hardly any experiments investigated their impact on arrhythmias. Prospective trials are currently ongoing, but no results have been published so far. Arrhythmias are common in the heart failure population, therefore the understanding of possible interactions with SGLT2 inhibitors is crucial. This review summarizes evidence from clinical data as well as the sparse experimental data of SGLT2 inhibitors and their effects on arrhythmias.
Collapse
Affiliation(s)
- Ewald Kolesnik
- Department of Cardiology, University Heart Centre Graz, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Daniel Scherr
- Department of Cardiology, University Heart Centre Graz, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Ursula Rohrer
- Department of Cardiology, University Heart Centre Graz, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Martin Benedikt
- Department of Cardiology, University Heart Centre Graz, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Martin Manninger
- Department of Cardiology, University Heart Centre Graz, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Harald Sourij
- Department of Endocrinology and Diabetology, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Dirk von Lewinski
- Department of Cardiology, University Heart Centre Graz, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| |
Collapse
|
27
|
Deschaine B, Verma S, Rayatzadeh H. Clinical Evidence and Proposed Mechanisms of Sodium-Glucose Cotransporter 2 Inhibitors in Heart Failure with Preserved Ejection Fraction: A Class Effect? Card Fail Rev 2022; 8:e23. [PMID: 35846984 PMCID: PMC9272408 DOI: 10.15420/cfr.2022.11] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 04/22/2022] [Indexed: 11/04/2022] Open
Abstract
Effective treatment for heart failure with preserved ejection fraction (HFpEF) is an unmet need in cardiovascular medicine. The pathophysiological drivers of HFpEF are complex, differing depending on phenotype, making a one-size-fits-all treatment approach unlikely. Remarkably, sodium-glucose cotransporter 2 inhibitors (SGLT2is) may be the first drug class to improve cardiovascular outcomes in HFpEF. Randomised controlled trials suggest a benefit in mortality, and demonstrate decreased hospitalisations and improvement in functional status. Limitations in trials exist, either due to small sample sizes, differing results between trials or decreased efficacy at higher ejection fractions. SGLT2is may provide a class effect by targeting various pathophysiological HFpEF mechanisms. Inhibition of SGLT2 and Na+/H+ exchanger 3 in the kidney promotes glycosuria, osmotic diuresis and natriuresis. The glucose deprivation activates sirtuins - protecting against oxidation and beneficially regulating metabolism. SGLT2is reduce excess epicardial adipose tissue and its deleterious adipokines. Na+/H+ exchanger 1 inhibition in the heart and lungs reduces sodium-induced calcium overload and pulmonary hypertension, respectively.
Collapse
Affiliation(s)
- Brent Deschaine
- University of Florida College of Medicine Gainesville, FL, US
| | - Sahil Verma
- Florida State University College of Medicine Tallahassee, FL, US
| | - Hussein Rayatzadeh
- Florida State University College of Medicine Tallahassee, FL, US.,Tallahassee Research Institute Tallahassee, FL, US.,Southern Medical Group Tallahassee, FL, US
| |
Collapse
|
28
|
Abdelhamid AM, Saber S, Youssef ME, Gaafar AGA, Eissa H, Abd-Eldayem MA, Alqarni M, Batiha GES, Obaidullah AJ, Shahien MA, El-Ahwany E, Amin NA, Etman MA, Kaddah MMY, Abd El-Fattah EE. Empagliflozin adjunct with metformin for the inhibition of hepatocellular carcinoma progression: Emerging approach for new application. Biomed Pharmacother 2021; 145:112455. [PMID: 34844106 DOI: 10.1016/j.biopha.2021.112455] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/05/2021] [Accepted: 11/16/2021] [Indexed: 02/08/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is on the rise worldwide, and its incidence in diabetic patients is two to three times that of non-diabetics. Current therapeutic options fail to provide considerable survival benefits to patients with HCC. There is a strong possibility that the FDA-approved antidiabetic combination of empagliflozin and metformin could show complementary effects to control HCC progression. However, their multitarget effects have not yet been studied on HCC development. Therefore, the present study aims to evaluate the antitumorigenic activity of this combination in non-diabetic mice with diethylnitrosamine-induced HCC. Empagliflozin/metformin combination prolonged survival and improved histological features of mice livers. Additionally, Empagliflozin/metformin showed anti-inflammatory potential and relieved oxidative stress. On the one hand these effects are likely attributed to the ability of metformin to inactivate NF-κB in an AMPK-dependent mechanism and on the other hand to the ability of the empagliflozin to inhibit the MAPKs, p38 and ERK1/2. Empagliflozin also showed a less robust effect on AMPK than that of metformin. Moreover, empagliflozin enhanced the autophagy inducing activity of metformin. Furthermore, empagliflozin/metformin exhibited increased apoptotic potential. Consequently, empagliflozin augmented the antitumorigenic function of metformin by exerting better control of angiogenesis, and metastasis. To conclude, our findings suggest empagliflozin as an ideal adjunct to metformin for the inhibition of HCC progression. In addition, since the incidence of hypoglycemia is minimal due to insulin-independent mechanism of action of both treatments, empagliflozin/metformin could be a promising therapeutic modality for the management of diabetic patients with HCC; and even non diabetic ones.
Collapse
Affiliation(s)
- Amir Mohamed Abdelhamid
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| | - Sameh Saber
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt.
| | - Mahmoud E Youssef
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| | - Ahmed Gaafar Ahmed Gaafar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Port Said University, Port Said, Egypt
| | - Hanan Eissa
- Department of Clinical Pharmacology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Marwa A Abd-Eldayem
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Horus University, New Damietta, Egypt
| | - Mohammed Alqarni
- Department of Pharmaceutical Chemistry, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, AlBeheira, Egypt
| | - Ahmad J Obaidullah
- Drug Exploration and Development Chair (DEDC), Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohamed Awad Shahien
- Department of Clinical Pharmacology, Faculty of Medicine, Damietta University, Damietta, Egypt
| | - Eman El-Ahwany
- Department of Immunology, Theodor Bilharz Research Institute, Giza, Egypt
| | - Noha A Amin
- Department of Hematology, Theodor Bilharz Research Institute, Giza, Egypt
| | - Mohamed Ali Etman
- Research and Development, Department of Drug Stability, Safe Pharma, Pharco Pharmaceuticals, Alexandria, Egypt
| | - Mohamed M Y Kaddah
- Pharmaceutical and Fermentation Industries Development Center, City of Scientific Research and Technological Applications, New Borg El-Arab 21934, Alexandria, Egypt
| | - Eslam E Abd El-Fattah
- Department of Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| |
Collapse
|