Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.898
Filter
1.
Am J Case Rep ; 25: e943945, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38985686

ABSTRACT

BACKGROUND Sodium-glucose cotransporter 2 (SGLT2) inhibitors are used to improve the prognosis of patients with diabetes, heart failure, or chronic kidney disease. The use of SGLT2 inhibitors in patients without diabetes is expected to increase. Diabetic ketoacidosis is a severe complication of SGLT2 inhibitors in patients with diabetes. People without diabetes are thought to be less likely to develop ketoacidosis, and reports of SGLT2 inhibitor-induced ketoacidosis are uncommon in people without diabetes. CASE REPORT Herein, we describe a case of ketoacidosis in an 83-year-old Japanese woman without diabetes who was administered SGLT2 inhibitors for heart failure (ejection fraction: approximately 30%). Two weeks prior to admission, she had suffered a vertebral fracture and rib fracture due to a fall, which was followed by anorexia, but she continued to take SGLT2 inhibitors. On admission, blood test results revealed a blood glucose level of 124 mg/dL, hemoglobin A1C level of 5.9%, pH of 7.329, HCO3⁻ concentration of 14.3 mmol/L, and a ß-hydroxybutyrate concentration of 5150 µmol/L, leading to a diagnosis of euglycemic ketoacidosis. The patient's C-peptide level was consistent with the blood glucose levels on admission, indicating that she had adequate insulin secretion. The patient was treated only with glucose administration without insulin and was discharged after discontinuation of the SGLT2 inhibitor. CONCLUSIONS This case illustrates that patients with or without diabetes may develop SGLT2 inhibitor-related ketoacidosis after several days of inadequate food intake; therefore, patients should be informed of this risk.


Subject(s)
Heart Failure , Sodium-Glucose Transporter 2 Inhibitors , Humans , Female , Heart Failure/chemically induced , Sodium-Glucose Transporter 2 Inhibitors/adverse effects , Aged, 80 and over , Ketosis/chemically induced , Glucosides/adverse effects , Diabetic Ketoacidosis/chemically induced
2.
J Cardiothorac Surg ; 19(1): 464, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39044225

ABSTRACT

BACKGROUND: Cardiac dysfunction, including arrhythmias, may be one of the main clinical manifestations of Becker muscular dystrophy (BMD). Amiodarone is widely used to treat arrhythmia. However, multi-systemic toxicity caused by amiodarone, especially hepatotoxicity, should not be neglected. Here, we introduce a novel case of multi-systemic amiodarone toxicity involving the liver, renal and coagulation in BDM patient with ABCB4 gene mutation. CASE PRESENTATION: We present a case of a 16-year-old boy admitted with heart failure and atrial fibrillation (AF). He was diagnosed with Becker muscular dystrophy (BMD) and gene testing showed comorbid mutations in gene DMD, ABCB4 and DSC2. Amiodarone was prescribed to control the paroxysmal atrial fibrillation intravenously. However, his liver enzyme levels were sharply elevated, along with cardiac shock, renal failure and coagulation disorders. After bedside continuous renal replacement therapy, the patient's liver function and clinical status rehabilitated. CONCLUSIONS: ABCB4 gene mutation might be involved in amiodarone-induced hepatotoxicity. Studies in a cohort might help to prove this hypothesis in the future.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B , Amiodarone , Anti-Arrhythmia Agents , Heart Failure , Muscular Dystrophy, Duchenne , Mutation , Humans , Amiodarone/adverse effects , Amiodarone/administration & dosage , Male , Adolescent , Heart Failure/chemically induced , ATP Binding Cassette Transporter, Subfamily B/genetics , Anti-Arrhythmia Agents/adverse effects , Anti-Arrhythmia Agents/therapeutic use , Anti-Arrhythmia Agents/administration & dosage , Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/complications , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/etiology , Atrial Fibrillation/drug therapy
3.
BMC Cardiovasc Disord ; 24(1): 350, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38987722

ABSTRACT

BACKGROUND: Antineoplastic medications, including doxorubicin, idarubicin, and epirubicin, have been found to adversely affect the heart due to oxidative stress - mitochondrial dysfunction - ferroptosis (ORMFs), which act as contributing attributes to anthracycline-induced cardiotoxicity. To better understand this phenomenon, the time-resolved measurements of ORMFS genes were analyzed in this study. METHODS: The effect of three anthracycline drugs on ORMFs genes was studied using a human 3D cardiac microtissue cell model. Transcriptome data was collected over 14 days at two doses (therapeutic and toxic). WGCNA identified key module-related genes, and functional enrichment analysis investigated the biological processes quantified by ssGSEA, such as immune cell infiltration and angiogenesis. Biopsies were collected from heart failure patients and control subjects. GSE59672 and GSE2965 were collected for validation. Molecular docking was used to identify anthracyclines's interaction with key genes. RESULTS: The ORMFs genes were screened in vivo or in vitro. Using WGCNA, six co-expressed gene modules were grouped, with MEblue emerging as the most significant module. Eight key genes intersecting the blue module with the dynamic response genes were obtained: CD36, CDH5, CHI3L1, HBA2, HSD11B1, OGN, RPL8, and VWF. Compared with control samples, all key genes except RPL8 were down-regulated in vitro ANT treatment settings, and their expression levels varied over time. According to functional analyses, the key module-related genes were engaged in angiogenesis and the immune system pathways. In all ANT-treated settings, ssGSEA demonstrated a significant down-regulation of angiogenesis score and immune cell activity, including Activated CD4 T cell, Immature B cell, Memory B cell, Natural killer cell, Type 1 T helper cell, and Type 2 T helper cell. Molecular docking revealed that RPL8 and CHI3L1 show significant binding affinity for anthracyclines. CONCLUSION: This study focuses on the dynamic characteristics of ORMFs genes in both human cardiac microtissues and cardiac biopsies from ANT-treated patients. It has been highlighted that ORMFs genes may contribute to immune infiltration and angiogenesis in cases of anthracycline-induced cardiotoxicity. A thorough understanding of these genes could potentially lead to improved diagnosis and treatment of the disease.


Subject(s)
Cardiotoxicity , Ferroptosis , Molecular Docking Simulation , Oxidative Stress , Humans , Oxidative Stress/drug effects , Ferroptosis/drug effects , Ferroptosis/genetics , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Mitochondria, Heart/genetics , Gene Regulatory Networks , Time Factors , Transcriptome , Epirubicin/adverse effects , Doxorubicin , Antibiotics, Antineoplastic/adverse effects , Case-Control Studies , Idarubicin , Heart Failure/chemically induced , Heart Failure/genetics , Heart Failure/metabolism , Heart Failure/physiopathology , Gene Expression Profiling , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Longitudinal Studies , Anthracyclines/adverse effects , Gene Expression Regulation , Signal Transduction
4.
J Cardiovasc Pharmacol ; 84(1): 110-117, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38922579

ABSTRACT

ABSTRACT: Hypercatecholaminergic conditions are known to cause heart failure and cardiac fibrosis when severe. Although previous investigations have studied the effects of beta-blockade in experimental models of catecholaminergic states, the detailed benefits of beta-blockade in more realistic models of hyper-adrenergic states were less clear. In this study, we examined acute cardiac changes in rats with hyperacute catecholamine-induced heart failure with and without propranolol treatment. Male Sprague-Dawley rats (n = 12) underwent a 6-hour infusion of epinephrine and norepinephrine alone, with an additional propranolol bolus (1 mg/kg) at hour 1 (n = 6). Cardiac tissues were examined after 6 hours. Cardiac immunohistochemistry revealed significantly decreased expression of phosphorylated p-38 (left ventricle, P = 0.021; right ventricle, P = 0.021), with upregulation of reactive oxidative species and other profibrosis proteins, after catecholamine infusion alone. After 1 propranolol 1 mg/kg bolus, the levels of phosphorylated-p38 returned to levels comparable with sham (left ventricle, P = 0.021; right ventricle, P = 0.043), with additional findings including downregulation of the apoptotic pathway and profibrotic proteins. We conclude that catecholamine-induced heart failure exerts damage through the p-38 mitogen-activated protein kinase pathway and demonstrates profibrotic changes mediated by matrix metalloproteinase 9, alpha-smooth muscle actin, and fibroblast growth factor 23. Changes in these pathways attenuated acute catecholamine-induced heart failure after propranolol bolus 1 mg/kg. We conclude that propranolol bolus at 1 mg/kg is able to mediate the effects of catecholamine excess through the p-38 mitogen-activated protein kinase pathway, profibrosis, and extrinsic apoptosis pathway.


Subject(s)
Adrenergic beta-Antagonists , Fibrosis , Heart Failure , Norepinephrine , Propranolol , Rats, Sprague-Dawley , p38 Mitogen-Activated Protein Kinases , Animals , Male , Propranolol/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Rats , Adrenergic beta-Antagonists/pharmacology , Adrenergic beta-Antagonists/administration & dosage , Heart Failure/drug therapy , Heart Failure/metabolism , Heart Failure/pathology , Heart Failure/physiopathology , Heart Failure/chemically induced , Norepinephrine/metabolism , Epinephrine/toxicity , Epinephrine/administration & dosage , Phosphorylation , Apoptosis/drug effects , Disease Models, Animal , Myocardium/pathology , Myocardium/metabolism , Myocardium/enzymology , Catecholamines/metabolism , Reactive Oxygen Species/metabolism
5.
J Am Heart Assoc ; 13(12): e033733, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38860414

ABSTRACT

BACKGROUND: Chronic sympathetic stimulation drives desensitization and downregulation of ß1 adrenergic receptor (ß1AR) in heart failure. We aim to explore the differential downregulation subcellular pools of ß1AR signaling in the heart. METHODS AND RESULTS: We applied chronic infusion of isoproterenol to induced cardiomyopathy in male C57BL/6J mice. We applied confocal and proximity ligation assay to examine ß1AR association with L-type calcium channel, ryanodine receptor 2, and SERCA2a ((Sarco)endoplasmic reticulum calcium ATPase 2a) and Förster resonance energy transfer-based biosensors to probe subcellular ß1AR-PKA (protein kinase A) signaling in ventricular myocytes. Chronic infusion of isoproterenol led to reduced ß1AR protein levels, receptor association with L-type calcium channel and ryanodine receptor 2 measured by proximity ligation (puncta/cell, 29.65 saline versus 14.17 isoproterenol, P<0.05), and receptor-induced PKA signaling at the plasma membrane (Förster resonance energy transfer, 28.9% saline versus 1.9% isoproterenol, P<0.05) and ryanodine receptor 2 complex (Förster resonance energy transfer, 30.2% saline versus 10.6% isoproterenol, P<0.05). However, the ß1AR association with SERCA2a was enhanced (puncta/cell, 51.4 saline versus 87.5 isoproterenol, P<0.05), and the receptor signal was minimally affected. The isoproterenol-infused hearts displayed decreased PDE4D (phosphodiesterase 4D) and PDE3A and increased PDE2A, PDE4A, and PDE4B protein levels. We observed a reduced role of PDE4 and enhanced roles of PDE2 and PDE3 on the ß1AR-PKA activity at the ryanodine receptor 2 complexes and myocyte shortening. Despite the enhanced ß1AR association with SERCA2a, the endogenous norepinephrine-induced signaling was reduced at the SERCA2a complexes. Inhibiting monoamine oxidase A rescued the norepinephrine-induced PKA signaling at the SERCA2a and myocyte shortening. CONCLUSIONS: This study reveals distinct mechanisms for the downregulation of subcellular ß1AR signaling in the heart under chronic adrenergic stimulation.


Subject(s)
Calcium Channels, L-Type , Cyclic AMP-Dependent Protein Kinases , Down-Regulation , Isoproterenol , Mice, Inbred C57BL , Myocytes, Cardiac , Receptors, Adrenergic, beta-1 , Ryanodine Receptor Calcium Release Channel , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Signal Transduction , Animals , Receptors, Adrenergic, beta-1/metabolism , Male , Ryanodine Receptor Calcium Release Channel/metabolism , Isoproterenol/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Calcium Channels, L-Type/metabolism , Calcium Channels, L-Type/drug effects , Disease Models, Animal , Mice , Heart Failure/metabolism , Heart Failure/chemically induced , Heart Failure/physiopathology , Cardiomyopathies/metabolism , Cardiomyopathies/chemically induced , Fluorescence Resonance Energy Transfer
6.
J Am Heart Assoc ; 13(9): e033700, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38700005

ABSTRACT

BACKGROUND: The only clinically approved drug that reduces doxorubicin cardiotoxicity is dexrazoxane, but its application is limited due to the risk of secondary malignancies. So, exploring alternative effective molecules to attenuate its cardiotoxicity is crucial. Colchicine is a safe and well-tolerated drug that helps reduce the production of reactive oxygen species. High doses of colchicine have been reported to block the fusion of autophagosomes and lysosomes in cancer cells. However, the impact of colchicine on the autophagy activity within cardiomyocytes remains inadequately elucidated. Recent studies have highlighted the beneficial effects of colchicine on patients with pericarditis, postprocedural atrial fibrillation, and coronary artery disease. It remains ambiguous how colchicine regulates autophagic flux in doxorubicin-induced heart failure. METHODS AND RESULTS: Doxorubicin was administered to establish models of heart failure both in vivo and in vitro. Prior studies have reported that doxorubicin impeded the breakdown of autophagic vacuoles, resulting in damaged mitochondria and the accumulation of reactive oxygen species. Following the administration of a low dose of colchicine (0.1 mg/kg, daily), significant improvements were observed in heart function (left ventricular ejection fraction: doxorubicin group versus treatment group=43.75%±3.614% versus 57.07%±2.968%, P=0.0373). In terms of mechanism, a low dose of colchicine facilitated the degradation of autolysosomes, thereby mitigating doxorubicin-induced cardiotoxicity. CONCLUSIONS: Our research has shown that a low dose of colchicine is pivotal in restoring the autophagy activity, thereby attenuating the cardiotoxicity induced by doxorubicin. Consequently, colchicine emerges as a promising therapeutic candidate to improve doxorubicin cardiotoxicity.


Subject(s)
Autophagy , Cardiotoxicity , Colchicine , Doxorubicin , Lysosomes , Myocytes, Cardiac , Colchicine/toxicity , Colchicine/pharmacology , Doxorubicin/toxicity , Cardiotoxicity/prevention & control , Autophagy/drug effects , Lysosomes/drug effects , Lysosomes/metabolism , Animals , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Disease Models, Animal , Male , Heart Failure/chemically induced , Heart Failure/drug therapy , Heart Failure/metabolism , Antibiotics, Antineoplastic/toxicity , Reactive Oxygen Species/metabolism , Mice , Mice, Inbred C57BL , Ventricular Function, Left/drug effects
7.
Am J Cardiol ; 222: 175-182, 2024 07 01.
Article in English | MEDLINE | ID: mdl-38692401

ABSTRACT

Anthracyclines are pivotal in cancer treatment, yet their clinical utility is hindered by the risk of cardiotoxicity. Preclinical studies highlight the effectiveness of sodium-glucose cotransporter-2 inhibitors (SGLT2i) in mitigating anthracycline-induced cardiotoxicity. Nonetheless, the translation of these findings to clinical practice remains uncertain. This study aims to evaluate the safety and potential of SGLT2i for preventing cardiotoxicity in patients with cancer, without preexisting heart failure (HF), receiving anthracyclines therapy. Using the TriNetX Global Research Network, patients with cancer, without previous HF diagnosis, receiving anthracycline therapy were identified and classified into 2 groups based on SGLT2i usage. A 1:1 propensity score matching was used to control for baseline characteristics between the 2 groups. Patients were followed for 2 years. The primary end point was new-onset HF, and the secondary end points were HF exacerbation, new-onset arrhythmia, myocardial infarction, all-cause mortality, and all-cause hospitalization. Safety outcomes included acute renal failure and creatinine levels. A total of 79,074 patients were identified, and 1,412 were included post-matching (706 in each group). They comprised 53% females, 62% White, with a mean age of 62.5 ± 11.4 years. Over the 2-year follow-up period, patients on SGLT2i had lower rates of new-onset HF (hazard ratio 0.147, 95% confidence interval 0.073 to 0.294) and arrhythmia (hazard ratio 0.397, 95% confidence interval 0.227 to 0.692) compared with those not on SGLT2i. The incidence of all-cause mortality, myocardial infarction, all-cause hospitalization, and safety outcomes were similar between both groups. In conclusion, among patients with cancer receiving anthracycline therapy without preexisting HF, SGLT2i use demonstrates both safety and effectiveness in reducing anthracycline-induced cardiotoxicity, with a decreased incidence of new-onset HF, HF exacerbation, and arrhythmias.


Subject(s)
Anthracyclines , Cardiotoxicity , Heart Failure , Neoplasms , Sodium-Glucose Transporter 2 Inhibitors , Humans , Female , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Male , Anthracyclines/therapeutic use , Anthracyclines/adverse effects , Middle Aged , Cardiotoxicity/prevention & control , Cardiotoxicity/etiology , Neoplasms/drug therapy , Aged , Heart Failure/chemically induced , Propensity Score , Hospitalization/statistics & numerical data , Myocardial Infarction/epidemiology , Myocardial Infarction/prevention & control
8.
Cancer Med ; 13(10): e7233, 2024 May.
Article in English | MEDLINE | ID: mdl-38752474

ABSTRACT

BACKGROUND: Over the past decade, immune checkpoint inhibitors (ICIs) have significantly transformed cancer treatment. However, ICIs inevitably may cause a spectrum of immune-related adverse events, among which cardiovascular toxicity, particularly myocarditis, while infrequent, has garnered increasing attention due to its high fatality rate. METHODS: We conducted a multicenter retrospective study to characterize ICI-associated cardiovascular adverse events. Logistic regression was performed to explore the risk factors for the development of myocarditis and severe myocarditis. Receiver operating characteristic curves were conducted to assess the diagnostic abilities of cardiac biomarkers to distinguish different cardiovascular toxicities, and the performance and calibration were evaluated using Hosmer-Lemeshow test. RESULTS: Forty-four patients were identified, including thirty-five myocarditis, five heart failure, three arrhythmias, and one myocardial infarction. Compared with other patients, myocarditis patients had higher cardiac troponin-I (cTnI) levels (p < 0.001), higher creatine kinase levels (p = 0.003), higher creatine kinase isoenzyme-MB (CK-MB) levels (p = 0.013), and shorter time to the incidence of adverse cardiovascular events (p = 0.022) after ICI treatment. Twenty-one patients (60%) were classified as severe myocarditis, and they presented higher cardiac troponin I (cTnI) levels (p = 0.013), higher N-terminal pro-B-type natriuretic peptide levels (p = 0.031), higher creatine kinase levels (p = 0.018), higher CK-MB levels (p = 0.026), and higher neutrophil to lymphocyte ratio (NLR) levels (p = 0.016) compared to non-severe myocarditis patients after ICI treatment. Multivariate logistic regression showed that CK-MB (adjusted odds ratio [OR]: 1.775, 95% confidence interval [CI]: 1.055-2.984, p = 0.031) was the independent risk factor of the development of ICI-associated myocarditis, and cTnI (adjusted OR: 1.021, 95% CI: 1.002-1.039, p = 0.03) and NLR (adjusted OR: 1.890, 95% CI: 1.026-3.483, p = 0.041) were the independent risk factors of ICI-associated severe myocarditis. The receiver operating characteristic curve showed an area under curve of 0.785 (95% CI: 0.642 to 0.928, p = 0.013) for CK-MB, 0.765 (95% CI: 0.601 to 0.929, p = 0.013) for cTnI, and 0.773 for NLR (95% CI: 0.597 to 0.948, p = 0.016). CONCLUSIONS: Elevated CK-MB after ICI treatment is the independent risk factor for the incidence of ICI-associated myocarditis, and elevated cTnI and NLR after ICI treatment are the independent risk factors for the development of ICI-associated severe myocarditis. CK-MB, cTnI, and NLR demonstrated a promising predictive utility for the identification of ICI-associated myocarditis and severe myocarditis.


Subject(s)
Immune Checkpoint Inhibitors , Myocarditis , Humans , Male , Retrospective Studies , Female , Immune Checkpoint Inhibitors/adverse effects , Myocarditis/chemically induced , Myocarditis/epidemiology , Myocarditis/diagnosis , Middle Aged , Aged , Risk Factors , Biomarkers/blood , Neoplasms/drug therapy , Troponin I/blood , ROC Curve , Cardiovascular Diseases/chemically induced , Cardiovascular Diseases/epidemiology , Creatine Kinase, MB Form/blood , Natriuretic Peptide, Brain/blood , Heart Failure/chemically induced
9.
Toxicol Appl Pharmacol ; 487: 116957, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38735590

ABSTRACT

Heart failure is associated with histone deacetylase (HDAC) regulation of gene expression, the inhibition of which is thought to be beneficial for heart failure therapy. Here, we explored the cardioprotective effects and underlying mechanism of a novel selenium-containing HDAC inhibitor, Se-SAHA, on isoproterenol (ISO)-induced heart failure. We found that pretreatment with Se-SAHA attenuated ISO-induced cardiac hypertrophy and fibrosis in neonatal rat ventricular myocytes (NRVMs). Se-SAHA significantly attenuated the generation of ISO-induced reactive oxygen species (ROS) and restored the expression levels of superoxide dismutase 2 (SOD2) and heme oxygenase-1 (HO-1) in vitro. Furthermore, Se-SAHA pretreatment prevented the accumulation of autophagosomes. Se-SAHA reversed the high expression of HDAC1 and HDAC6 induced by ISO incubation. However, after the addition of the HDAC agonist, the effect of Se-SAHA on blocking autophagy was inhibited. Using ISO-induced mouse models, cardiac ventricular contractile dysfunction, hypertrophy, and fibrosis was reduced treated by Se-SAHA. In addition, Se-SAHA inhibited HDAC1 and HDAC6 overexpression in ISO-treated mice. Se-SAHA treatment significantly increased the activity of SOD2 and improved the ability to eliminate free radicals. Se-SAHA hindered the excessive levels of the microtubule-associated protein 1 light chain 3 (LC3)-II and Beclin-1 in heart failure mice. Collectively, our results indicate that Se-SAHA exerts cardio-protection against ISO-induced heart failure via antioxidative stress and autophagy inhibition.


Subject(s)
Autophagy , Heart Failure , Histone Deacetylase Inhibitors , Isoproterenol , Mice, Inbred C57BL , Myocytes, Cardiac , Oxidative Stress , Rats, Sprague-Dawley , Animals , Isoproterenol/toxicity , Heart Failure/chemically induced , Heart Failure/prevention & control , Heart Failure/pathology , Heart Failure/drug therapy , Autophagy/drug effects , Histone Deacetylase Inhibitors/pharmacology , Oxidative Stress/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Myocytes, Cardiac/metabolism , Male , Rats , Mice , Superoxide Dismutase/metabolism , Reactive Oxygen Species/metabolism , Antioxidants/pharmacology , Fibrosis , Cells, Cultured , Cardiomegaly/chemically induced , Cardiomegaly/prevention & control , Cardiomegaly/pathology
10.
Pak J Pharm Sci ; 37(2): 337-347, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38767101

ABSTRACT

Heart failure is a condition in which the heart's one or both ventricles are unable to either receive an adequate amount of blood or eject an adequate amount of blood. Diabetes is considered one of the major risk factors for cardiovascular diseases. The current research is designed to evaluate the cardioprotective effects of dapagliflozin in streptozotocin and isoproterenol-induced comorbid rats. The COX-2, TNF-α, NF-КB, NLRP3, PPAR-γ, CKMB, TROP-I, AR, GP and SGLT were docked against dapagliflozin, propranolol and metformin. Dapagliflozin restored adequate blood flow and halted myofibril damage. Moreover, it's evident from this study that dapagliflozin significantly decreased serum concentration of various blood markers, decreased relative growth rate and QT interval prolongation, as compared to the negative control group. However, it improved the ventricular ejection fraction in rats of the treatment group. The GST, GSH and CAT levels were increased, as compared to normal. On the contrary, a decrease in LPO concentrations was observed. Evaluation of the coronal section of heart tissues showed the anti-inflammatory expressions evaluated through H & E staining and immunohistochemical techniques and with ELISA and PCR. In a nutshell, dapagliflozin reverses myocardial necrosis and apoptosis.


Subject(s)
Benzhydryl Compounds , Glucosides , Heart Failure , Isoproterenol , NLR Family, Pyrin Domain-Containing 3 Protein , PPAR gamma , Signal Transduction , Streptozocin , Animals , Glucosides/pharmacology , Isoproterenol/toxicity , Heart Failure/chemically induced , Heart Failure/drug therapy , Heart Failure/metabolism , Benzhydryl Compounds/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , PPAR gamma/metabolism , Rats , Signal Transduction/drug effects , Male , Rats, Wistar , Diabetes Mellitus, Experimental/drug therapy , Cardiotonic Agents/pharmacology , Apoptosis/drug effects , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Myocardium/metabolism , Myocardium/pathology
11.
BMJ ; 385: e076268, 2024 04 17.
Article in English | MEDLINE | ID: mdl-38631737

ABSTRACT

OBJECTIVE: To investigate risks of multiple adverse outcomes associated with use of antipsychotics in people with dementia. DESIGN: Population based matched cohort study. SETTING: Linked primary care, hospital and mortality data from Clinical Practice Research Datalink (CPRD), England. POPULATION: Adults (≥50 years) with a diagnosis of dementia between 1 January 1998 and 31 May 2018 (n=173 910, 63.0% women). Each new antipsychotic user (n=35 339, 62.5% women) was matched with up to 15 non-users using incidence density sampling. MAIN OUTCOME MEASURES: The main outcomes were stroke, venous thromboembolism, myocardial infarction, heart failure, ventricular arrhythmia, fracture, pneumonia, and acute kidney injury, stratified by periods of antipsychotic use, with absolute risks calculated using cumulative incidence in antipsychotic users versus matched comparators. An unrelated (negative control) outcome of appendicitis and cholecystitis combined was also investigated to detect potential unmeasured confounding. RESULTS: Compared with non-use, any antipsychotic use was associated with increased risks of all outcomes, except ventricular arrhythmia. Current use (90 days after a prescription) was associated with elevated risks of pneumonia (hazard ratio 2.19, 95% confidence interval (CI) 2.10 to 2.28), acute kidney injury (1.72, 1.61 to 1.84), venous thromboembolism (1.62, 1.46 to 1.80), stroke (1.61, 1.52 to 1.71), fracture (1.43, 1.35 to 1.52), myocardial infarction (1.28, 1.15 to 1.42), and heart failure (1.27, 1.18 to 1.37). No increased risks were observed for the negative control outcome (appendicitis and cholecystitis). In the 90 days after drug initiation, the cumulative incidence of pneumonia among antipsychotic users was 4.48% (4.26% to 4.71%) versus 1.49% (1.45% to 1.53%) in the matched cohort of non-users (difference 2.99%, 95% CI 2.77% to 3.22%). CONCLUSIONS: Antipsychotic use compared with non-use in adults with dementia was associated with increased risks of stroke, venous thromboembolism, myocardial infarction, heart failure, fracture, pneumonia, and acute kidney injury, but not ventricular arrhythmia. The range of adverse outcomes was wider than previously highlighted in regulatory alerts, with the highest risks soon after initiation of treatment.


Subject(s)
Acute Kidney Injury , Antipsychotic Agents , Appendicitis , Cholecystitis , Dementia , Heart Failure , Myocardial Infarction , Pneumonia , Stroke , Venous Thromboembolism , Adult , Humans , Female , Male , Antipsychotic Agents/therapeutic use , Cohort Studies , Venous Thromboembolism/epidemiology , Appendicitis/complications , Stroke/epidemiology , Myocardial Infarction/epidemiology , Arrhythmias, Cardiac/complications , Heart Failure/chemically induced , Dementia/drug therapy , Pneumonia/drug therapy , Acute Kidney Injury/chemically induced
12.
Br J Pharmacol ; 181(16): 2964-2990, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38679957

ABSTRACT

BACKGROUND AND PURPOSE: Cancer therapy-related cardiovascular adverse events (CAEs) in presence of comorbidities, are in the spotlight of the cardio-oncology guidelines. Carfilzomib (Cfz), indicated for relapsed/refractory multiple myeloma (MM), presents with serious CAEs. MM is often accompanied with co-existing comorbidities. However, Cfz use in MM patients with cardiometabolic syndrome (CMS) or in heart failure with reduced ejection fraction (HFrEF), is questionable. EXPERIMENTAL APPROACH: ApoE-/- and C57BL6/J male mice received 14 weeks Western Diet (WD) (CMS models). C57BL6/J male mice underwent permanent LAD ligation for 14 days (early-stage HFrEF model). CMS- and HFrEF-burdened mice received Cfz for two consecutive or six alternate days. Daily metformin and atorvastatin administrations were performed additionally to Cfz, as prophylactic interventions. Mice underwent echocardiography, while proteasome activity, biochemical and molecular analyses were conducted. KEY RESULTS: CMS did not exacerbate Cfz left ventricular (LV) dysfunction, whereas Cfz led to metabolic complications in both CMS models. Cfz induced autophagy and Ca2+ homeostasis dysregulation, whereas metformin and atorvastatin prevented Cfz-mediated LV dysfunction and molecular deficits in the CMS-burdened myocardium. Early-stage HFrEF led to depressed LV function and increased protein phosphatase 2A (PP2A) activity. Cfz further increased myocardial PP2A activity, inflammation and Ca2+-cycling dysregulation. Metformin co-administration exerted an anti-inflammatory potential on the myocardium without improving LV function. CONCLUSION AND IMPLICATIONS: CMS and HFrEF seem to exacerbate Cfz-induced CAEs, by presenting metabolism-related hidden toxicity and PP2A-related cardiac inflammation, respectively. Metformin retains its prophylactic potential in the presence of CMS, while mitigating inflammation and Ca2+ signalling dysregulation in the HFrEF myocardium.


Subject(s)
Cardiotoxicity , Heart Failure , Mice, Inbred C57BL , Oligopeptides , Animals , Male , Cardiotoxicity/prevention & control , Oligopeptides/pharmacology , Oligopeptides/administration & dosage , Heart Failure/chemically induced , Heart Failure/metabolism , Heart Failure/drug therapy , Heart Failure/prevention & control , Mice , Metabolic Syndrome/chemically induced , Metabolic Syndrome/drug therapy , Metabolic Syndrome/metabolism
13.
J Am Heart Assoc ; 13(8): e032397, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38591334

ABSTRACT

BACKGROUND: This study investigated whether initial SGLT2 (sodium-glucose cotransporter 2) inhibitor-based treatment is superior to metformin-based regimens as a primary prevention strategy among low-risk patients with diabetes. METHODS AND RESULTS: In this nationwide cohort study, a total of 38 496 patients with diabetes with low cardiovascular risk were identified (age 62.0±11.6 years, men 50%) from January 1 to December 31, 2016. Patients receiving SGLT2 inhibitors-based and metformin-based regimens were 1:2 matched by propensity score. Study outcomes included all-cause mortality, cardiovascular death, hospitalization for heart failure, stroke, and progression to end-stage renal disease. Compared with 1928 patients receiving metformin-based regimens, 964 patients receiving SGLT2 inhibitor-based regimens had similar all-cause mortality (hazard ratio [HR], 0.75 [95% CI, 0.51-1.12]), cardiovascular death (HR, 0.69 [95% CI, 0.25-1.89]), hospitalization for heart failure (HR, 1.06 [95% CI, 0.59-1.92]), stroke (HR, 0.78 [95% CI, 0.48-1.27]), and progression to end-stage renal disease (HR, 0.88 [95% CI, 0.32-2.39]). However, SGLT2 inhibitors were associated with a lower risk of all-cause mortality (HR, 0.47 [95% CI, 0.23-0.99]; P for interaction=0.008) and progression to end-stage renal disease (HR, 0.22 [95% CI, 0.06-0.82]; P for interaction=0.04) in patients under the age of 65. CONCLUSIONS: In comparison to metformin-based regimens, SGLT2 inhibitor-based regimens showed a similar risk of all-cause mortality and adverse cardiorenal events. SGLT2 inhibitors might be considered as first-line therapy in select low-risk patients, for example, younger patients with diabetes.


Subject(s)
Cardiovascular Diseases , Diabetes Mellitus, Type 2 , Heart Failure , Kidney Failure, Chronic , Metformin , Sodium-Glucose Transporter 2 Inhibitors , Stroke , Male , Humans , Middle Aged , Aged , Sodium-Glucose Transporter 2 Inhibitors/adverse effects , Metformin/therapeutic use , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/drug therapy , Cohort Studies , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/prevention & control , Cardiovascular Diseases/chemically induced , Risk Factors , Treatment Outcome , Heart Failure/epidemiology , Heart Failure/chemically induced , Heart Disease Risk Factors , Stroke/chemically induced , Glucose , Hypoglycemic Agents/therapeutic use
14.
Ann Rheum Dis ; 83(8): 1028-1033, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38594057

ABSTRACT

OBJECTIVE: To investigate the risk of cardiovascular disease (CVD) associated with increasing dose of a non-steroidal anti-inflammatory drug (NSAID) in patients with ankylosing spondylitis (AS). METHODS: Using the Korean National Health Insurance database, patients newly diagnosed with AS without prior CVD between 2010 and 2018 were included in this nationwide cohort study. The primary outcome was CVD, a composite outcome of ischaemic heart disease, stroke or congestive heart failure. Exposure to NSAIDs was evaluated using a time-varying approach. The dose of NSAIDs was considered in each exposure period. Cox proportional hazard regression was used to investigate the risk of CVD associated with NSAID use. RESULTS: Of the 19 775 patients (mean age, 36 years; 75% were male), 19 706 received NSAID treatment. During follow-up period of 98 290 person-years, 1663 cases of CVD occurred including 1157 cases of ischaemic heart disease, 301 cases of stroke and 613 cases of congestive heart failure. Increasing dose of NSAIDs was associated with incident CVD after adjusting for confounders (adjusted HR (aHR) 1.10; 95% CI 1.08 to 1.13). Specifically, increasing dose of NSAIDs was associated with incident ischaemic heart disease (aHR 1.08; 95% CI 1.05 to 1.11), stroke (aHR 1.09; 95% CI 1.04 to 1.15) and congestive heart failure (aHR 1.12; 95% CI 1.08 to 1.16). The association between NSAID dose and higher CVD risk was consistent in different subgroups. CONCLUSION: In a real-world AS cohort, higher dose of NSAID treatment was associated with a higher risk of CVD, including ischaemic heart disease, stroke and congestive heart failure.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal , Cardiovascular Diseases , Spondylitis, Ankylosing , Humans , Spondylitis, Ankylosing/drug therapy , Spondylitis, Ankylosing/complications , Spondylitis, Ankylosing/epidemiology , Male , Female , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Adult , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/chemically induced , Middle Aged , Republic of Korea/epidemiology , Heart Failure/epidemiology , Heart Failure/chemically induced , Dose-Response Relationship, Drug , Proportional Hazards Models , Cohort Studies , Stroke/epidemiology , Stroke/chemically induced , Risk Factors , Incidence
15.
Pharmacoepidemiol Drug Saf ; 33(4): e5790, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38575389

ABSTRACT

PURPOSE: The prevalent new user design extends the active comparator new user design to include patients switching to a treatment of interest from a comparator. We examined the impact of adding "switchers" to incident new users on the estimated hazard ratio (HR) of hospitalized heart failure. METHODS: Using MarketScan claims data (2000-2014), we estimated HRs of hospitalized heart failure between patients initiating GLP-1 receptor agonists (GLP-1 RA) and sulfonylureas (SU). We considered three estimands: (1) the effect of incident new use; (2) the effect of switching; and (3) the effect of incident new use or switching, combining the two population. We used time-conditional propensity scores (TCPS) and time-stratified standardized morbidity ratio (SMR) weighting to adjust for confounding. RESULTS: We identified 76 179 GLP-1 RA new users, of which 12% were direct switchers (within 30 days) from SU. Among incident new users, GLP-1 RA was protective against heart failure (adjHRSMR = 0.74 [0.69, 0.80]). Among switchers, GLP-1 RA was not protective (adjHRSMR = 0.99 [0.83, 1.18]). Results in the combined population were largely driven by the incident new users, with GLP-1 RA having a protective effect (adjHRSMR = 0.77 [0.72, 0.83]). Results using TCPS were consistent with those estimated using SMR weighting. CONCLUSIONS: When analyses were conducted only among incident new users, GLP-1 RA had a protective effect. However, among switchers from SU to GLP-1 RA, the effect estimates substantially shifted toward the null. Combining patients with varying treatment histories can result in poor confounding control and camouflage important heterogeneity.


Subject(s)
Diabetes Mellitus, Type 2 , Heart Failure , Humans , Diabetes Mellitus, Type 2/epidemiology , Sulfonylurea Compounds/therapeutic use , Risk Factors , Heart Failure/drug therapy , Heart Failure/epidemiology , Heart Failure/chemically induced , Glucagon-Like Peptide 1/agonists , Glucagon-Like Peptide-1 Receptor , Hypoglycemic Agents/therapeutic use
16.
Expert Opin Drug Deliv ; 21(3): 399-422, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38623735

ABSTRACT

INTRODUCTION: Doxorubicin (DOX) emerges as a cornerstone in the arsenal of potent chemotherapeutic agents. Yet, the clinical deployment of DOX is tarnished by its proclivity to induce severe cardiotoxic effects, culminating in heart failure and other consequential morbidities. In response, a panoply of strategies has undergone rigorous exploration over recent decades, all aimed at attenuating DOX's cardiotoxic impact. The advent of encapsulating DOX within lipidic or polymeric nanocarriers has yielded a dual triumph, augmenting DOX's therapeutic efficacy while mitigating its deleterious side effects. AREAS COVERED: Recent strides have spotlighted the emergence of DOX conjugates as particularly auspicious avenues for ameliorating DOX-induced cardiotoxicity. These conjugates entail the fusion of DOX through physical or chemical bonds with diminutive natural or synthetic moieties, polymers, biomolecules, and nanoparticles. This spectrum encompasses interventions that impinge upon DOX's cardiotoxic mechanism, modulate cellular uptake and localization, confer antioxidative properties, or refine cellular targeting. EXPERT OPINION: The endorsement of DOX conjugates as a compelling stratagem to mitigate DOX-induced cardiotoxicity resounds from this exegesis, amplifying safety margins and the therapeutic profile of this venerated chemotherapeutic agent. Within this ambit, DOX conjugates stand as a beacon of promise in the perpetual pursuit of refining chemotherapy-induced cardiac compromise.


Subject(s)
Antibiotics, Antineoplastic , Cardiotoxicity , Doxorubicin , Drug Carriers , Nanoparticles , Doxorubicin/adverse effects , Doxorubicin/administration & dosage , Cardiotoxicity/prevention & control , Cardiotoxicity/etiology , Humans , Animals , Antibiotics, Antineoplastic/adverse effects , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/chemistry , Drug Carriers/chemistry , Nanoparticles/chemistry , Drug Delivery Systems , Polymers/chemistry , Heart Failure/drug therapy , Heart Failure/chemically induced , Lipids/chemistry
17.
J Mol Cell Cardiol ; 191: 12-22, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38643934

ABSTRACT

Doxorubicin (DOX) is a widely used chemotherapeutic agent that can cause serious cardiotoxic side effects, leading to heart failure (HF). Impaired mitochondrial function is thought to be key factor driving progression into HF. We have previously shown in a rat model of DOX-HF that heart failure with reduced ejection fraction correlates with mitochondrial loss and dysfunction. Adenosine monophosphate-dependent kinase (AMPK) is a cellular energy sensor, regulating mitochondrial biogenesis and energy metabolism, including fatty acid oxidation. We hypothesised that AMPK activation could restore mitochondrial function and therefore be a novel cardioprotective strategy for the prevention of DOX-HF. Consequently, we set out to assess whether 5-aminoimidazole-4-carboxamide 1-ß-D-ribofuranoside (AICAR), an activator of AMPK, could prevent cardiac functional decline in this chronic intravenous rat model of DOX-HF. In line with our hypothesis, AICAR improved cardiac systolic function. AICAR furthermore improved cardiac mitochondrial fatty acid oxidation, independent of mitochondrial number, and in the absence of observable AMPK-activation. In addition, we found that AICAR prevented loss of myocardial mass. RNAseq analysis showed that this may be driven by normalisation of pathways associated with ribosome function and protein synthesis, which are impaired in DOX-treated rat hearts. AICAR furthermore prevented dyslipidemia and excessive body-weight loss in DOX-treated rats, which may contribute to preservation of myocardial mass. Though it is unclear whether AICAR exerted its cardioprotective effect through cardiac or extra-cardiac AMPK-activation or via an AMPK-independent effect, these results show promise for the use of AICAR as a cardioprotective agent in DOX-HF to both preserve cardiac function and mass.


Subject(s)
AMP-Activated Protein Kinases , Aminoimidazole Carboxamide , Cardiotonic Agents , Doxorubicin , Heart Failure , Ribonucleotides , Animals , Doxorubicin/adverse effects , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Heart Failure/chemically induced , Heart Failure/prevention & control , Heart Failure/metabolism , Heart Failure/pathology , Heart Failure/drug therapy , Ribonucleotides/pharmacology , Male , Cardiotonic Agents/pharmacology , Rats , AMP-Activated Protein Kinases/metabolism , Mitochondria, Heart/metabolism , Mitochondria, Heart/drug effects , Myocardium/metabolism , Myocardium/pathology , Fatty Acids/metabolism , Disease Models, Animal
19.
Anticancer Drugs ; 35(6): 556-558, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38453155

ABSTRACT

This case report features a 62-year-old male with stage IB lung adenocarcinoma harboring an epidermal growth factor receptor exon 19 deletion, who underwent treatment with osimertinib following a left upper lobectomy and lymph node dissection. Despite a history of smoking and well-managed type 2 diabetes, the patient developed heart failure 18 months post-initiation of osimertinib therapy, marking one of the latest occurrences of heart failure following osimertinib treatment documented in limited literature. Cardiac MRI revealed significant left ventricular enlargement, lateral wall myocardial thinning, and localized myocardial fibrosis without perfusion defects, a finding not previously reported in literature. This case underscores the severe and unusual cardiac effects of osimertinib in patients with latent risk factors, highlighting the importance of vigilant cardiac monitoring and a multidisciplinary management approach.


Subject(s)
Acrylamides , Aniline Compounds , Carcinoma, Non-Small-Cell Lung , Cardiotoxicity , Lung Neoplasms , Humans , Acrylamides/adverse effects , Acrylamides/therapeutic use , Aniline Compounds/adverse effects , Aniline Compounds/therapeutic use , Male , Lung Neoplasms/drug therapy , Middle Aged , Carcinoma, Non-Small-Cell Lung/drug therapy , Cardiotoxicity/etiology , Antineoplastic Agents/adverse effects , Heart Failure/chemically induced , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Indoles , Pyrimidines
20.
Cardiovasc Toxicol ; 24(5): 513-518, 2024 May.
Article in English | MEDLINE | ID: mdl-38530625

ABSTRACT

Acute high-output heart failure (HOHF) with pulmonary hypertension and liver injury caused by amlodipine poisoning is very rare. We report a 52-year-old woman who suffered from severe shock after an overdose of amlodipine. Hemodynamic monitoring showed that while her left ventricular systolic function and cardiac output were elevated, her systemic vascular resistance decreased significantly. At the same time, the size of her right heart, her central venous pressure, and the oxygen saturation of her central venous circulation all increased abnormally. The patient's circulatory function and right ventricular dysfunction gradually improved after large doses of vasopressors and detoxification measures. However, her bilirubin and transaminase levels increased significantly on hospital day 6, with a CT scan showing patchy, low-density areas in her liver along with ascites. After liver protective treatment and plasma exchange, the patient's liver function gradually recovered. A CT scan 4 months later showed all her liver abnormalities, including ascites, had resolved. The common etiologies of HOHF were excluded in this case, and significantly reduced systemic vascular resistance caused by amlodipine overdose was thought to be the primary pathophysiological basis of HOHF. The significant increase in venous return and pulmonary blood flow is considered to be the main mechanism of right ventricular dysfunction and pulmonary hypertension. Hypoxic hepatitis caused by a combination of hepatic congestion and distributive shock may be the most important factors causing liver injury in this patient. Whether amlodipine has other mechanisms leading to HOHF and pulmonary hypertension needs to be further studied. Considering the significant increase of right heart preload, aggressive fluid resuscitation should be done very cautiously in patients with HOHF and shock secondary to amlodipine overdose.


Subject(s)
Amlodipine , Chemical and Drug Induced Liver Injury , Drug Overdose , Heart Failure , Hypertension, Pulmonary , Humans , Female , Amlodipine/poisoning , Middle Aged , Hypertension, Pulmonary/physiopathology , Hypertension, Pulmonary/chemically induced , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/physiopathology , Drug Overdose/complications , Heart Failure/chemically induced , Heart Failure/physiopathology , Treatment Outcome , Cardiac Output, High/physiopathology , Cardiac Output, High/chemically induced , Antihypertensive Agents , Ventricular Function, Right/drug effects , Calcium Channel Blockers/poisoning , Severity of Illness Index , Hemodynamics/drug effects , Acute Disease
SELECTION OF CITATIONS
SEARCH DETAIL