Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.715
Filter
1.
Clin Cardiol ; 47(7): e24307, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38953367

ABSTRACT

BACKGROUND: We aim to provide a comprehensive review of the current state of knowledge of myocardial viability assessment in patients undergoing coronary artery bypass grafting (CABG), with a focus on the clinical markers of viability for each imaging modality. We also compare mortality between patients with viable myocardium and those without viability who undergo CABG. METHODS: A systematic database search with meta-analysis was conducted of comparative original articles (both observations and randomized controlled studies) of patients undergoing CABG with either viable or nonviable myocardium, in EMBASE, MEDLINE, Cochrane database, and Google Scholar, from inception to 2022. Imaging modalities included were dobutamine stress echocardiography (DSE), cardiac magnetic resonance (CMR), single-photon emission computed tomography (SPECT), and positron emission tomography (PET). RESULTS: A total of 17 studies incorporating a total of 2317 patients were included. Across all imaging modalities, the relative risk of death post-CABG was reduced in patients with versus without viability (random-effects model: odds ratio: 0.42; 95% confidence interval: 0.29-0.61; p < 0.001). Imaging for myocardial viability has significant clinical implications as it can affect the accuracy of the diagnosis, guide treatment decisions, and predict patient outcomes. Generally, based on local availability and expertise, either SPECT or DSE should be considered as the first step in evaluating viability, while PET or CMR would provide further evaluation of transmurality, perfusion metabolism, and extent of scar tissue. CONCLUSION: The assessment of myocardial viability is an essential component of preoperative evaluation in patients with ischemic heart disease undergoing surgical revascularization. Careful patient selection and individualized assessment of viability remain paramount.


Subject(s)
Coronary Artery Bypass , Myocardial Ischemia , Ventricular Function, Left , Humans , Cardiomyopathies/physiopathology , Cardiomyopathies/surgery , Cardiomyopathies/diagnosis , Cardiomyopathies/etiology , Coronary Artery Bypass/adverse effects , Coronary Artery Disease/surgery , Coronary Artery Disease/physiopathology , Coronary Artery Disease/diagnosis , Coronary Artery Disease/complications , Echocardiography, Stress/methods , Myocardial Ischemia/physiopathology , Myocardial Ischemia/surgery , Myocardial Ischemia/diagnosis , Myocardial Ischemia/complications , Myocardium/pathology , Tissue Survival , Tomography, Emission-Computed, Single-Photon , Ventricular Dysfunction, Left/physiopathology , Ventricular Dysfunction, Left/etiology , Ventricular Function, Left/physiology
2.
J Pregnancy ; 2024: 7713590, 2024.
Article in English | MEDLINE | ID: mdl-38957710

ABSTRACT

Preeclampsia and peripartum cardiomyopathy (PPCM) are significant obstetric problems that can arise during or after pregnancy. Both are known to be causes of maternal mortality and morbidity. Several recent studies have suggested a link between preeclampsia and the pathophysiology of PPCM. However, the common thread that connects the two has yet to be thoroughly and fully articulated. Here, we investigate the complex dynamics of preeclampsia and PPCM in this review. Our analysis focuses mainly on inflammatory and immunological responses, endothelial dysfunction as a shared pathway, and potential genetic predisposition to both diseases. To begin, we will look at how excessive inflammatory and immunological responses can lead to clinical symptoms of both illnesses, emphasizing the role of proinflammatory cytokines and immune cells in modifying vascular and tissue responses. Second, we consider endothelial dysfunction to be a crucial point at which endothelial damage and activation contribute to pathogenesis through increased vascular permeability, vascular dysfunction, and thrombus formation. Finally, we examine recent information suggesting genetic predispositions to preeclampsia and PPCM, such as genetic variants in genes involved in the management of blood pressure, the inflammatory response, and heart structural integrity. With this synergistic study, we seek to encourage more research and creative therapy solutions by emphasizing the need for an interdisciplinary approach to understanding and managing the connection between preeclampsia and PPCM.


Subject(s)
Cardiomyopathies , Peripartum Period , Pre-Eclampsia , Humans , Female , Pre-Eclampsia/physiopathology , Pre-Eclampsia/genetics , Pregnancy , Cardiomyopathies/etiology , Cardiomyopathies/genetics , Cardiomyopathies/physiopathology , Genetic Predisposition to Disease , Endothelium, Vascular/physiopathology , Pregnancy Complications, Cardiovascular/physiopathology , Pregnancy Complications, Cardiovascular/genetics
3.
Turk Kardiyol Dern Ars ; 52(5): 357-361, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38982822

ABSTRACT

Right ventricular pacing (RVP) is conventionally preferred in the treatment of patients with atrioventricular block. However, long-term RVP may lead to pacing-induced cardiomyopathy (PICM), characterized by new-onset or worsening ventricular functions due to dyssynchronous ventricular electrical activation, abnormal ventricular remodeling, and increased energy expenditure. Historically, biventricular pacing (BVP) and guideline-directed medical therapy were the only treatment option for PICM. Recently, conduction system pacing, including left bundle branch area pacing (LBBaP), has emerged as a physiological alternative to BVP, showing better results in electro-mechanical ventricular synchronization and hemodynamic parameters compared to BVP. We present a case involving a patient from whom the PICM was successfully recovered shortly after LBBaP.


Subject(s)
Cardiac Pacing, Artificial , Cardiomyopathies , Humans , Cardiomyopathies/therapy , Cardiomyopathies/etiology , Cardiac Pacing, Artificial/methods , Cardiac Pacing, Artificial/adverse effects , Electrocardiography , Male , Heart Ventricles/physiopathology
4.
Nutrients ; 16(13)2024 Jun 29.
Article in English | MEDLINE | ID: mdl-38999835

ABSTRACT

Arrhythmogenic cardiomyopathy (ACM) is a familial heart disease characterized by cardiac dysfunction, arrhythmias, and myocardial inflammation. Exercise and stress can influence the disease's progression. Thus, an investigation of whether a high-fat diet (HFD) contributes to ACM pathogenesis is warranted. In a robust ACM mouse model, 8-week-old Desmoglein-2 mutant (Dsg2mut/mut) mice were fed either an HFD or rodent chow for 8 weeks. Chow-fed wildtype (WT) mice served as controls. Echo- and electrocardiography images pre- and post-dietary intervention were obtained, and the lipid burden, inflammatory markers, and myocardial fibrosis were assessed at the study endpoint. HFD-fed Dsg2mut/mut mice showed numerous P-wave perturbations, reduced R-amplitude, left ventricle (LV) remodeling, and reduced ejection fraction (%LVEF). Notable elevations in plasma high-density lipoprotein (HDL) were observed, which correlated with the %LVEF. The myocardial inflammatory adipokines, adiponectin (AdipoQ) and fibroblast growth factor-1, were substantially elevated in HFD-fed Dsg2mut/mut mice, albeit no compounding effect was observed in cardiac fibrosis. The HFD not only potentiated cardiac dysfunction but additionally promoted adverse cardiac remodeling. Further investigation is warranted, particularly given elevated AdipoQ levels and the positive correlation of HDL with the %LVEF, which may suggest a protective effect. Altogether, the HFD worsened some, but not all, disease phenotypes in Dsg2mut/mut mice. Notwithstanding, diet may be a modifiable environmental factor in ACM disease progression.


Subject(s)
Diet, High-Fat , Animals , Diet, High-Fat/adverse effects , Mice , Disease Models, Animal , Myocardium/pathology , Myocardium/metabolism , Fibrosis , Male , Ventricular Remodeling , Desmoglein 2/genetics , Myocarditis/etiology , Myocarditis/physiopathology , Mice, Inbred C57BL , Arrhythmogenic Right Ventricular Dysplasia/etiology , Arrhythmogenic Right Ventricular Dysplasia/physiopathology , Adiponectin/blood , Inflammation , Cardiomyopathies/etiology , Cardiomyopathies/physiopathology
5.
Zhongguo Dang Dai Er Ke Za Zhi ; 26(7): 774-781, 2024 Jul 15.
Article in Chinese | MEDLINE | ID: mdl-39014956

ABSTRACT

Sepsis-induced myocardial depression (SIMD), a common complication of sepsis, is one of the main causes of death in patients with sepsis. The pathogenesis of SIMD is complicated, and the process of SIMD remains incompletely understood, with no single or definitive mechanism fully elucidated. Notably, pyroptosis, as a pro-inflammatory programmed cell death, is characterized by Gasdermin-mediated formation of pores on the cell membrane, cell swelling, and cell rupture accompanied by the release of large amounts of inflammatory factors and other cellular contents. Mechanistically, pyroptosis is mainly divided into the canonical pathway mediated by caspase-1 and the non-canonical pathway mediated by caspase-4/5/11. Pyroptosis has been confirmed to participate in various inflammation-associated diseases. In recent years, more and more studies have shown that pyroptosis is also involved in the occurrence and development of SIMD. This article reviews the molecular mechanisms of pyroptosis and its research progress in SIMD, aiming to provide novel strategies and targets for the treatment of SIMD.


Subject(s)
Pyroptosis , Sepsis , Humans , Sepsis/complications , Animals , Cardiomyopathies/etiology
6.
Int J Mol Sci ; 25(12)2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38928407

ABSTRACT

Radiotherapy (RT) may have a cardiotoxic effect on the heart and cardiovascular system. Postulated mechanisms mediating these complications include vascular endothelium damage and myocardial fibrosis. The aim of our study was to assess endothelial damage and myocardial fibrosis in the early period after RT on the basis of cardiac biomarkers and in relation to the radiation dose applied to individual heart structures in patients treated for non-small-cell lung cancer. This single-center prospective study included consecutive patients with lung cancer (LC) who were referred for treatment with radiochemotherapy (study group) or chemotherapy (control group). The study protocol included performing an echocardiographic examination, a standard ECG examination, and collecting blood samples for laboratory tests before starting treatment for lung cancer in the first week after completing RT (after four cycles of chemotherapy in the control group) and after 12 weeks from the end of treatment. The study included 23 patients in the study group and 20 patients in the control group. Compared to the baseline values, there was a significant increase in total cholesterol concentration in the study group immediately after the end of RT, which persisted for three months after the end of therapy. After taking into account the use of statins in the analysis, it was found that an increase in total cholesterol concentration after oncological treatment was observed only among patients who did not use statins. Taking into account the assessment of myocardial fibrosis markers, there were no significant changes in the concentration of matrix metallopeptidase 9 (MMP-9) and tissue inhibitors of metalloproteinases 1 (TIMP-1) in the study group. In patients treated with radiochemotherapy, there was a significant increase in the concentration of intercellular adhesion molecule 1 (ICAM-1) immediately after RT, when compared to the baseline. After taking into account the use of statins, an increase in ICAM-1 concentration immediately after RT was observed only in patients who did not use statins. There was also a significant correlation between the radiation dose received by the left anterior descending coronary artery (LAD) and left circumferential coronary artery, and vascular cell adhesion protein 1 (VCAM-1) concentration measured at three months after the end of RT. Immediately after completion of radiotherapy, a significant increase in the level of ICAM-1 is observed indicating endothelial damage. The radiation dose to coronary arteries should be minimized, as it correlates with the concentration of VCAM-1. The use of statins may prevent the increase in total cholesterol and ICAM-1 concentration after irradiation for lung cancer; however, further studies designed for this specific purpose are necessary to confirm the effectiveness of statins in this area.


Subject(s)
Fibrosis , Lung Neoplasms , Humans , Male , Female , Lung Neoplasms/radiotherapy , Lung Neoplasms/pathology , Middle Aged , Aged , Prospective Studies , Carcinoma, Non-Small-Cell Lung/radiotherapy , Carcinoma, Non-Small-Cell Lung/pathology , Endothelium, Vascular/radiation effects , Endothelium, Vascular/pathology , Endothelium, Vascular/metabolism , Endothelium, Vascular/drug effects , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/blood , Myocardium/pathology , Myocardium/metabolism , Radiotherapy/adverse effects , Tissue Inhibitor of Metalloproteinase-1/blood , Tissue Inhibitor of Metalloproteinase-1/metabolism , Cardiomyopathies/etiology , Cardiomyopathies/pathology , Cholesterol/blood , Biomarkers/blood
7.
G Ital Cardiol (Rome) ; 25(7): 483-490, 2024 Jul.
Article in Italian | MEDLINE | ID: mdl-38916463

ABSTRACT

Aortic valve stenosis and cardiac amyloidosis, particularly transthyretin-related, often coexist and share a common clinical and demographic profile. Several pathophysiological hypotheses have been proposed regarding the causes of this association, neither of which fully substantiated in practice. The key to detect the coexistence of cardiac amyloidosis and aortic valve stenosis lies in clinical suspicion. It is possible to hypothesize concurrent cardiac amyloidosis in patients with aortic valve stenosis with the aid of clinical, electrocardiographic, echocardiographic, and extracardiac "red flags". Subsequent non-invasive diagnostic steps are often sufficient to establish a definitive diagnosis. The early diagnosis of this condition is pivotal since the presence of dual pathology worsens patient's prognosis, especially without intervention. Available data on treatment show a better outcome in terms of survival and cardiovascular events in patients undergoing percutaneous correction of valvular heart disease rather than medical therapy alone, regardless of the presence of cardiac amyloidosis. Furthermore, it seems that cardiac amyloidosis does not impact survival after transcatheter aortic valve replacement, even if higher rates of rehospitalizations have been described. Indeed, percutaneous treatment of valvular heart disease is currently considered the primary therapeutic option. Subsequently a disease-modifying treatment for transthyretin amyloidosis may be considered in order to delay disease progression and improve outcomes, even if specific data are still lacking.


Subject(s)
Aortic Valve Stenosis , Humans , Aortic Valve Stenosis/etiology , Aortic Valve Stenosis/diagnosis , Aortic Valve Stenosis/surgery , Prognosis , Amyloidosis/diagnosis , Amyloidosis/therapy , Amyloidosis/complications , Cardiomyopathies/etiology , Cardiomyopathies/therapy , Cardiomyopathies/diagnosis , Amyloid Neuropathies, Familial/complications , Amyloid Neuropathies, Familial/diagnosis , Amyloid Neuropathies, Familial/therapy , Echocardiography
8.
Int J Mol Sci ; 25(11)2024 May 28.
Article in English | MEDLINE | ID: mdl-38892040

ABSTRACT

Cirrhotic cardiomyopathy (CCM) is defined as cardiac dysfunction associated with cirrhosis in the absence of pre-existing heart disease. CCM manifests as the enlargement of cardiac chambers, attenuated systolic and diastolic contractile responses to stress stimuli, and repolarization changes. CCM significantly contributes to mortality and morbidity in patients who undergo liver transplantation and contributes to the pathogenesis of hepatorenal syndrome/acute kidney injury. There is currently no specific treatment. The traditional management for non-cirrhotic cardiomyopathies, such as vasodilators or diuretics, is not applicable because an important feature of cirrhosis is decreased systemic vascular resistance; therefore, vasodilators further worsen the peripheral vasodilatation and hypotension. Long-term diuretic use may cause electrolyte imbalances and potentially renal injury. The heart of the cirrhotic patient is insensitive to cardiac glycosides. Therefore, these types of medications are not useful in patients with CCM. Exploring the therapeutic strategies of CCM is of the utmost importance. The present review summarizes the possible treatment of CCM. We detail the current status of non-selective beta-blockers (NSBBs) in the management of cirrhotic patients and discuss the controversies surrounding NSBBs in clinical practice. Other possible therapeutic agents include drugs with antioxidant, anti-inflammatory, and anti-apoptotic functions; such effects may have potential clinical application. These drugs currently are mainly based on animal studies and include statins, taurine, spermidine, galectin inhibitors, albumin, and direct antioxidants. We conclude with speculations on the future research directions in CCM treatment.


Subject(s)
Cardiomyopathies , Liver Cirrhosis , Humans , Liver Cirrhosis/complications , Liver Cirrhosis/drug therapy , Cardiomyopathies/therapy , Cardiomyopathies/etiology , Animals , Adrenergic beta-Antagonists/therapeutic use , Antioxidants/therapeutic use
9.
Heart Fail Clin ; 20(3): 325-331, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38844303

ABSTRACT

Amyloidosis is a systemic disease due to the accumulation of misfolded amyloid fibrils that damage the heart and worsen the prognosis. Heart failure (HF), a condition frequently linked with an advanced stage of this disease, is the most prevalent clinical manifestation that leads to its diagnosis. However, due to the growing awareness of the occurrence of cardiac amyloidosis (CA), it is now possible to perform an early diagnosis and have a positive impact on its natural course. This study aims to highlight the most compelling issues concerning patients' clinical management with HF and CA.


Subject(s)
Amyloidosis , Cardiomyopathies , Heart Failure , Humans , Amyloidosis/therapy , Heart Failure/etiology , Cardiomyopathies/therapy , Cardiomyopathies/etiology , Prognosis
10.
Clin Cardiol ; 47(6): e24298, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38873847

ABSTRACT

BACKGROUND: In patients with transthyretin cardiac amyloidosis (ATTR-CA), renal dysfunction is a poor prognostic indicator. Limited data are available on variables that portend worsening renal function (wRF) among ATTR-CA patients. OBJECTIVES: This study assesses which characteristics place patients at higher risk for the development of wRF (defined as a drop of ≥10% in glomerular filtration rate [GFR]) within the first year following diagnosis of ATTR-CA. METHODS: We included patients with ATTR-CA (n = 134) evaluated between 2/2016 and 12/2022 and followed for up to 1 year at our amyloid clinic. Patients were stratified into two groups: a group with maintained renal function (mRF) and a group with wRF and compared using appropriate testing. Significant variables in the univariate analysis were included in the multivariable logistic regression model to determine characteristics associated with wRF. RESULTS: Within a follow-up period of 326 ± 118 days, the median GFR% change measured -6% [-18%, +8]. About 41.8% (n = 56) had wRF, while the remainder had mRF. In addition, in patients with no prior history of chronic kidney disease (CKD), 25.5% developed de novo CKD. On multivariable logistic regression, only New York Heart Association (NYHA) class ≥III (odds ratio [OR]: 3.9, 95% confidence interval [CI]: [1.6-9.3]), history of ischemic heart disease (IHD) (OR: 0.3, 95% CI: [0.1-0.7]), and not receiving SGLT-2i (OR: 0.1, 95% CI: [0.02-0.5]) were significant predictors of wRF. CONCLUSION: Our study demonstrated that the development of de novo renal dysfunction or wRF is common following the diagnosis of ATTR-CA. Additionally, we identified worse NYHA class and no prior history of IHD as significant predictors associated with developing wRF, while receiving SGLT-2i therapy appeared to be protective in this population.


Subject(s)
Amyloid Neuropathies, Familial , Cardiomyopathies , Glomerular Filtration Rate , Humans , Male , Female , Amyloid Neuropathies, Familial/complications , Amyloid Neuropathies, Familial/diagnosis , Amyloid Neuropathies, Familial/physiopathology , Aged , Cardiomyopathies/diagnosis , Cardiomyopathies/physiopathology , Cardiomyopathies/etiology , Prognosis , Retrospective Studies , Risk Factors , Middle Aged , Follow-Up Studies , Renal Insufficiency, Chronic/diagnosis , Renal Insufficiency, Chronic/physiopathology , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/epidemiology , Disease Progression , Kidney/physiopathology , Time Factors , Incidence , Risk Assessment/methods
11.
Rev Fac Cien Med Univ Nac Cordoba ; 81(2): 432-452, 2024 06 28.
Article in Spanish | MEDLINE | ID: mdl-38941220

ABSTRACT

The diagnosis of Cirrhotic Cardiomyopathy is based on severe hepatic cirrosis with deterioration of cardiac function without previous cardiopathy, but this is subclinical during a long time. In this second part we review the non-invasive diagnostic methods and their prognostic value in patients with or without hepatic transplant, from ECG to cardiac images of magnetic resonance.


El diagnóstico de Cardiomiopatía Cirrótica está basado en la presencia de cirrosis hepática avanzada con alteraciones de la función cardíaca sin cardiopatía pre-existente, pero en gran parte de su evolución natural ésta es subclínica. Por ello son imprescindibles los estudios complementarios no invasivos para confirmar el diagnóstico y su rol pronóstico en pacientes con o sin trasplante hepático. En esta segunda parte revisamos los métodos de diagnóstico desde el ECG hasta las imágenes de resonancia magnética cardíaca.


Subject(s)
Cardiomyopathies , Liver Cirrhosis , Magnetic Resonance Imaging , Humans , Cardiomyopathies/etiology , Cardiomyopathies/physiopathology , Liver Cirrhosis/complications , Electrocardiography , Prognosis
12.
Medicine (Baltimore) ; 103(21): e38305, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38788008

ABSTRACT

RATIONALE: Thyrotoxic cardiomyopathy is a rare but severe complication of thyrotoxicosis, leading to episodes of acute heart failure. This case report highlights a rare presentation of thyrotoxic cardiomyopathy with low-output heart failure, emphasizing the importance of early diagnosis and comprehensive management. The report aims to increase awareness among clinicians about the potential reversibility of this condition and the effective strategies for managing such complex cases. PATIENT CONCERNS: This patient presented with dyspnea and chest constriction, without any antecedent predisposing factors. Subsequently, the patient abruptly manifested symptoms indicative of acute heart failure during outpatient consultation. Electrocardiography revealed rapid atrial fibrillation with type A preexcitation syndrome, whereas cardiac ultrasonography demonstrated global cardiac enlargement with a diminished ejection fraction (EF). DIAGNOSES: After a comprehensive evaluation, the patient was diagnosed with thyrotoxic cardiomyopathy, acute heart failure, and atrial fibrillation with preexcitation syndrome. INTERVENTIONS: Immediate interventions comprised diuretic administration, oxygen therapy, and antiarrhythmic agents, addressing acute heart failure concomitant with preexcitation syndrome. Following a fortnight of comprehensive therapeutic measures, the patient was discharged with a prescription for oral medications, notably methimazole. OUTCOMES: Following the intervention, the patient showed significant improvement with the resolution of heart failure symptoms and dyspnea, restoration of sinus rhythm, improved left ventricular ejection fraction (LVEF improved from 36% to 45%), and normalization of thyroid function. These outcomes underscore the efficacy of the intervention strategy and offer a hopeful prognosis for similar cases. LESSONS: Thyrotoxicosis may cause cardiomyopathy in patients with heart failure that manifests as dilated cardiac chambers. Clinicians should carefully screen patients for this reversible condition. Diagnosis requires a comprehensive assessment of various tests, and the therapeutic goal is to restore normal thyroid function.


Subject(s)
Cardiomyopathies , Heart Failure , Thyrotoxicosis , Humans , Heart Failure/etiology , Thyrotoxicosis/complications , Thyrotoxicosis/diagnosis , Cardiomyopathies/etiology , Cardiomyopathies/diagnosis , Cardiomyopathies/therapy , Atrial Fibrillation/etiology , Atrial Fibrillation/complications , Electrocardiography , Female , Male , Acute Disease
14.
J Am Coll Cardiol ; 83(22): 2214-2232, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38811098

ABSTRACT

Arrhythmias frequently accompany heart failure and left ventricular dysfunction. Tachycardias, atrial fibrillation, and premature ventricular contractions can induce a reversible form of dilated cardiomyopathy (CM) known as arrhythmia-induced CM (AiCM). The intriguing question is why certain individuals are more susceptible to AiCM, despite similar arrhythmia burdens. The primary challenge is determining the extent of arrhythmias' contribution to left ventricular systolic dysfunction. AiCM should be considered in patients with a mean heart rate of >100 beats/min, atrial fibrillation, or a PVC burden of >10%. Confirmation of AiCM occurs when CM reverses upon eliminating the responsible arrhythmia. Therapy choice depends on the specific arrhythmia, patient comorbidities, and preferences. After left ventricular function is restored, ongoing follow-up is essential if an abnormal myocardial substrate persists. Accurate diagnosis and treatment of AiCM have the potential to enhance patients' quality of life, improve clinical outcomes, and reduce hospital admissions and overall health care costs.


Subject(s)
Arrhythmias, Cardiac , Humans , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/therapy , Arrhythmias, Cardiac/physiopathology , Cardiomyopathies/etiology , Cardiomyopathies/therapy , Cardiomyopathies/physiopathology , Cardiomyopathies/diagnosis , Cardiomyopathy, Dilated/therapy , Cardiomyopathy, Dilated/physiopathology , Cardiomyopathy, Dilated/etiology
15.
Front Biosci (Landmark Ed) ; 29(5): 200, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38812311

ABSTRACT

AIMS: Changes in myocardial mitochondrial morphology and function in premature ventricular contractions (PVCs)-induced cardiomyopathy (PVCCM) remain poorly studied. Here, we investigated the effects of PVCs with different coupling intervals (CIs) on myocardial mitochondrial remodelling in a canine model of PVCCM. METHODS AND RESULTS: Twenty-one beagles underwent pacemaker implantation and were randomised into the sham (n = 7), short-coupled PVCs (SCP, n = 7), and long-coupled PVCs (LCP, n = 7) groups. Right ventricular (RV) apical bigeminy was produced for 12-week to induce PVCCM in the SCP (CI, 250 ms) and LCP (CI, 350 ms) groups. Echocardiography was performed at baseline and biweekly thereafter to evaluate cardiac function. Masson's trichrome staining measured ventricular interstitial fibrosis. The ultrastructural morphology of the myocardial mitochondria was analysed using transmission electron microscopy. Mitochondrial Ca2+ concentration, reactive oxygen species (ROS) levels, adenosine triphosphate (ATP) content, membrane potential, and electron transport chain (ETC) complex activity were measured to assess myocardial mitochondrial function. Twelve-week-PVCs led to left ventricular (LV) enlargement with systolic dysfunction, disrupted mitochondrial morphology, increased mitochondrial Ca2+ concentration and ROS levels, decreased mitochondrial ATP content and membrane potential, and impaired ETC complex activity in both the SCP and LCP groups (all p < 0.01 vs the sham group). Ventricular fibrosis was observed only in canines with LCP. Worse cardiac function and more pronounced abnormalities in mitochondrial morphology and function were observed in the LCP group than to the SCP group (all p < 0.05). CONCLUSION: We demonstrated myocardial mitochondrial abnormalities in dogs with PVCCM, characterised by abnormal mitochondrial morphology, mitochondrial Ca2+ overload, oxidative stress, and impaired mitochondrial energy metabolism. Compared to SCP, long-term LCP exposure resulted in more severe mitochondrial remodelling and cardiac dysfunction in dogs.


Subject(s)
Calcium , Cardiomyopathies , Disease Models, Animal , Mitochondria, Heart , Reactive Oxygen Species , Ventricular Premature Complexes , Animals , Dogs , Mitochondria, Heart/metabolism , Mitochondria, Heart/ultrastructure , Mitochondria, Heart/pathology , Cardiomyopathies/physiopathology , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Cardiomyopathies/etiology , Ventricular Premature Complexes/physiopathology , Ventricular Premature Complexes/metabolism , Reactive Oxygen Species/metabolism , Calcium/metabolism , Male , Adenosine Triphosphate/metabolism , Membrane Potential, Mitochondrial , Echocardiography
16.
Immun Inflamm Dis ; 12(5): e1229, 2024 May.
Article in English | MEDLINE | ID: mdl-38775678

ABSTRACT

BACKGROUND: Dioscin has many pharmacological effects; however, its role in sepsis-induced cardiomyopathy (SIC) is unknown. Accordingly, we concentrate on elucidating the mechanism of Dioscin in SIC rat model. METHODS: The SIC rat and H9c2 cell models were established by lipopolysaccharide (LPS) induction. The heart rate (HR), left ventricle ejection fraction (LVEF), mean arterial blood pressure (MAP), and heart weight index (HWI) of rats were evaluated. The myocardial tissue was observed by hematoxylin and eosin staining. 4-Hydroxy-2-nonenal (4-HNE) level in myocardial tissue was detected by immunohistochemistry. Superoxide dismutase (SOD), catalase (CAT), and glutathione (GSH) activities in serum samples of rats and H9c2 cells were determined by colorimetric assay. Bax, B-cell lymphoma-2 (Bcl-2), toll-like receptor 4 (TLR4), myeloid differentiation primary response 88 (MyD88), phosphorylated-p65 (p-p65), and p65 levels in myocardial tissues of rats and treated H9c2 cells were measured by quantitative real-time PCR and Western blot. Viability and reactive oxygen species (ROS) accumulation of treated H9c2 cells were assayed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide and dihydroethidium staining assays. RESULTS: Dioscin decreased HR and HWI, increased LVEF and MAP, alleviated the myocardial tissue damage, and reduced 4-HNE level in SIC rats. Dioscin reversed LPS-induced reduction on SOD, CAT, GSH, and Bcl-2 levels, and increment on Bax and TLR4 levels in rats and H9c2 cells. Overexpressed TLR4 attenuated the effects of Dioscin on promoting viability, as well as dwindling TLR4, ROS and MyD88 levels, and p-p65/p65 value in LPS-induced H9c2 cells. CONCLUSION: Protective effects of Dioscin against LPS-induced SIC are achieved via regulation of TLR4/MyD88/p65 signal pathway.


Subject(s)
Cardiomyopathies , Diosgenin , Myeloid Differentiation Factor 88 , Sepsis , Signal Transduction , Toll-Like Receptor 4 , Animals , Diosgenin/analogs & derivatives , Diosgenin/pharmacology , Diosgenin/therapeutic use , Toll-Like Receptor 4/metabolism , Rats , Myeloid Differentiation Factor 88/metabolism , Sepsis/complications , Sepsis/drug therapy , Sepsis/metabolism , Signal Transduction/drug effects , Male , Cardiomyopathies/drug therapy , Cardiomyopathies/etiology , Cardiomyopathies/metabolism , Cardiomyopathies/prevention & control , Cell Line , Rats, Sprague-Dawley , Transcription Factor RelA/metabolism , Oxidative Stress/drug effects , Lipopolysaccharides , Disease Models, Animal , Apoptosis/drug effects
17.
Int J Med Sci ; 21(6): 983-993, 2024.
Article in English | MEDLINE | ID: mdl-38774750

ABSTRACT

Previous studies have highlighted the protective effects of pyruvate kinase M2 (PKM2) overexpression in septic cardiomyopathy. In our study, we utilized cardiomyocyte-specific PKM2 knockout mice to further investigate the role of PKM2 in attenuating LPS-induced myocardial dysfunction, focusing on mitochondrial biogenesis and prohibitin 2 (PHB2). Our findings confirmed that the deletion of PKM2 in cardiomyocytes significantly exacerbated LPS-induced myocardial dysfunction, as evidenced by impaired contractile function and relaxation. Additionally, the deletion of PKM2 intensified LPS-induced myocardial inflammation. At the molecular level, LPS triggered mitochondrial dysfunction, characterized by reduced ATP production, compromised mitochondrial respiratory complex I/III activities, and increased ROS production. Intriguingly, the absence of PKM2 further worsened LPS-induced mitochondrial damage. Our molecular investigations revealed that LPS disrupted mitochondrial biogenesis in cardiomyocytes, a disruption that was exacerbated by the absence of PKM2. Given that PHB2 is known as a downstream effector of PKM2, we employed PHB2 adenovirus to restore PHB2 levels. The overexpression of PHB2 normalized mitochondrial biogenesis, restored mitochondrial integrity, and promoted mitochondrial function. Overall, our results underscore the critical role of PKM2 in regulating the progression of septic cardiomyopathy. PKM2 deficiency impeded mitochondrial biogenesis, leading to compromised mitochondrial integrity, increased myocardial inflammation, and impaired cardiac function. The overexpression of PHB2 mitigated the deleterious effects of PKM2 deletion. This discovery offers a novel insight into the molecular mechanisms underlying septic cardiomyopathy and suggests potential therapeutic targets for intervention.


Subject(s)
Cardiomyopathies , Mice, Knockout , Mitochondria, Heart , Myocytes, Cardiac , Prohibitins , Pyruvate Kinase , Sepsis , Animals , Cardiomyopathies/pathology , Cardiomyopathies/metabolism , Cardiomyopathies/genetics , Cardiomyopathies/etiology , Mice , Myocytes, Cardiac/pathology , Myocytes, Cardiac/metabolism , Sepsis/metabolism , Sepsis/pathology , Sepsis/genetics , Pyruvate Kinase/metabolism , Pyruvate Kinase/genetics , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Repressor Proteins/genetics , Repressor Proteins/metabolism , Humans , Organelle Biogenesis , Lipopolysaccharides/toxicity , Male , Disease Models, Animal
18.
Int Immunopharmacol ; 134: 112186, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38733824

ABSTRACT

BACKGROUND: Although the pathophysiological mechanism of septic cardiomyopathy has been continuously discovered, it is still a lack of effective treatment method. Cortistatin (CST), a neuroendocrine polypeptide of the somatostatin family, has emerged as a novel cardiovascular-protective peptide, but the specific mechanism has not been elucidated. PURPOSE: The aim of our study is to explore the role of CST in cardiomyocytes pyroptosis and myocardial injury in sepsis and whether CST inhibits cardiomyocytes pyroptosis through specific binding with somastatin receptor 2 (SSTR2) and activating AMPK/Drp1 signaling pathway. METHODS AND RESULTS: In this study, plasma CST levels were significantly high and were negatively correlated with N-terminal pro-B type natriuretic peptide (NT-proBNP), a biomarker for cardiac dysfunction, in patients with sepsis. Exogenous administration of CST significantly improved survival rate and cardiac function in mouse models of sepsis by inhibiting the activation of the NLRP3 inflammasome and pyroptosis of cardiomyocytes (decreased cleavage of caspase-1, IL-1ß and gasdermin D). Pharmacological inhibition and genetic ablation revealed that CST exerted anti-pyroptosis effects by specifically binding to somatostatin receptor subtype 2 (SSTR2), thus activating AMPK and inactivating Drp1 to inhibit mitochondrial fission in cardiomyocytes. CONCLUSIONS: This study is the first to report that CST attenuates septic cardiomyopathy by inhibiting cardiomyocyte pyroptosis through the SSTR2-AMPK-Drp1-NLRP3 pathway. Importantly, CST specifically binds to SSTR2, which promotes AMPK phosphorylation, inhibits Drp1-mediated mitochondrial fission, and reduces ROS levels, thereby inhibiting NLRP3 inflammasome activation-mediated pyroptosis and alleviating sepsis-induced myocardial injury.


Subject(s)
AMP-Activated Protein Kinases , Cardiomyopathies , Mice, Inbred C57BL , Myocytes, Cardiac , NLR Family, Pyrin Domain-Containing 3 Protein , Neuropeptides , Pyroptosis , Receptors, Somatostatin , Sepsis , Signal Transduction , Animals , Pyroptosis/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Receptors, Somatostatin/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Humans , Sepsis/drug therapy , Signal Transduction/drug effects , AMP-Activated Protein Kinases/metabolism , Neuropeptides/metabolism , Mice , Male , Cardiomyopathies/drug therapy , Cardiomyopathies/etiology , Cardiomyopathies/metabolism , Disease Models, Animal , Mice, Knockout
19.
Int J Mol Sci ; 25(9)2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38731929

ABSTRACT

Sepsis-induced cardiomyopathy (SICM) is one of the leading indicators for poor prognosis associated with sepsis. Despite its reversibility, prognosis varies widely among patients. Mitochondria play a key role in cellular energy production by generating adenosine triphosphate (ATP), which is vital for myocardial energy metabolism. Over recent years, mounting evidence suggests that severe sepsis not only triggers mitochondrial structural abnormalities such as apoptosis, incomplete autophagy, and mitophagy in cardiomyocytes but also compromises their function, leading to ATP depletion. This metabolic disruption is recognized as a significant contributor to SICM, yet effective treatment options remain elusive. Sepsis cannot be effectively treated with inotropic drugs in failing myocardium due to excessive inflammatory factors that blunt ß-adrenergic receptors. This review will share the recent knowledge on myocardial cell death in sepsis and its molecular mechanisms, focusing on the role of mitochondria as an important metabolic regulator of SICM, and discuss the potential for developing therapies for sepsis-induced myocardial injury.


Subject(s)
Cardiomyopathies , Sepsis , Sepsis/complications , Sepsis/metabolism , Humans , Cardiomyopathies/etiology , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Animals , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Mitophagy , Energy Metabolism , Mitochondria/metabolism , Mitochondria/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Apoptosis , Adenosine Triphosphate/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...