Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.433
Filter
1.
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
2.
Cell Mol Life Sci ; 81(1): 254, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38856931

ABSTRACT

The endogenous mitochondrial quality control (MQC) system serves to protect mitochondria against cellular stressors. Although mitochondrial dysfunction contributes to cardiac damage during many pathological conditions, the regulatory signals influencing MQC disruption during septic cardiomyopathy (SC) remain unclear. This study aimed to investigate the involvement of pyruvate kinase M2 (PKM2) and prohibitin 2 (PHB2) interaction followed by MQC impairment in the pathogenesis of SC. We utilized LPS-induced SC models in PKM2 transgenic (PKM2TG) mice, PHB2S91D-knockin mice, and PKM2-overexpressing HL-1 cardiomyocytes. After LPS-induced SC, cardiac PKM2 expression was significantly downregulated in wild-type mice, whereas PKM2 overexpression in vivo sustained heart function, suppressed myocardial inflammation, and attenuated cardiomyocyte death. PKM2 overexpression relieved sepsis-related mitochondrial damage via MQC normalization, evidenced by balanced mitochondrial fission/fusion, activated mitophagy, restored mitochondrial biogenesis, and inhibited mitochondrial unfolded protein response. Docking simulations, co-IP, and domain deletion mutant protein transfection experiments showed that PKM2 phosphorylates PHB2 at Ser91, preventing LPS-mediated PHB2 degradation. Additionally, the A domain of PKM2 and the PHB domain of PHB2 are required for PKM2-PHB2 binding and PHB2 phosphorylation. After LPS exposure, expression of a phosphorylation-defective PHB2S91A mutant negated the protective effects of PKM2 overexpression. Moreover, knockin mice expressing a phosphorylation-mimetic PHB2S91D mutant showed improved heart function, reduced inflammation, and preserved mitochondrial function following sepsis induction. Abundant PKM2 expression is a prerequisite to sustain PKM2-PHB2 interaction which is a key element for preservation of PHB2 phosphorylation and MQC, presenting novel interventive targets for the treatment of septic cardiomyopathy.


Subject(s)
Cardiomyopathies , Myocytes, Cardiac , Prohibitins , Pyruvate Kinase , Repressor Proteins , Sepsis , Animals , Phosphorylation , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Mice , Pyruvate Kinase/metabolism , Pyruvate Kinase/genetics , Sepsis/metabolism , Repressor Proteins/metabolism , Repressor Proteins/genetics , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Mitochondria, Heart/metabolism , Mice, Transgenic , Mice, Inbred C57BL , Male , Lipopolysaccharides , Humans , Mitophagy
3.
Circ Res ; 134(12): 1824-1840, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38843291

ABSTRACT

Immunometabolism is an emerging field at the intersection of immunology and metabolism. Immune cell activation plays a critical role in the pathogenesis of cardiovascular diseases and is integral for regeneration during cardiac injury. We currently possess a limited understanding of the processes governing metabolic interactions between immune cells and cardiomyocytes. The impact of this intercellular crosstalk can manifest as alterations to the steady state flux of metabolites and impact cardiac contractile function. Although much of our knowledge is derived from acute inflammatory response, recent work emphasizes heterogeneity and flexibility in metabolism between cardiomyocytes and immune cells during pathological states, including ischemic, cardiometabolic, and cancer-associated disease. Metabolic adaptation is crucial because it influences immune cell activation, cytokine release, and potential therapeutic vulnerabilities. This review describes current concepts about immunometabolic regulation in the heart, focusing on intercellular crosstalk and intrinsic factors driving cellular regulation. We discuss experimental approaches to measure the cardio-immunologic crosstalk, which are necessary to uncover unknown mechanisms underlying the immune and cardiac interface. Deeper insight into these axes holds promise for therapeutic strategies that optimize cardioimmunology crosstalk for cardiac health.


Subject(s)
Myocytes, Cardiac , Humans , Animals , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/immunology , Energy Metabolism , Cardiomyopathies/metabolism , Cardiomyopathies/immunology , Myocardium/metabolism , Myocardium/immunology , Myocardium/pathology
4.
Heart Fail Clin ; 20(3): 283-294, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38844299

ABSTRACT

Amyloidosis is a systemic condition characterized by multiple organs involvement. A multidisciplinary and multimodal approach in assessing patients is pivotal and recommended by the international scientific societies. Biomarkers represent an essential noninvasive tool to increase the suspicion of disease and orient further workup and clinical management of patients. This review provides an updated contemporary focus on the clinical use of biomarkers in cardiac amyloidosis, emphasizing their role in both the diagnostic and prognostic setting and discussing future perspective of emerging biomarkers.


Subject(s)
Amyloidosis , Biomarkers , Cardiomyopathies , Humans , Biomarkers/metabolism , Amyloidosis/diagnosis , Cardiomyopathies/diagnosis , Cardiomyopathies/metabolism , Prognosis
5.
Cells ; 13(11)2024 May 27.
Article in English | MEDLINE | ID: mdl-38891055

ABSTRACT

Intracellular cargo delivery via distinct transport routes relies on vesicle carriers. A key trafficking route distributes cargo taken up by clathrin-mediated endocytosis (CME) via early endosomes. The highly dynamic nature of the endosome network presents a challenge for its quantitative analysis, and theoretical modelling approaches can assist in elucidating the organization of the endosome trafficking system. Here, we introduce a new computational modelling approach for assessment of endosome distributions. We employed a model of induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) with inherited mutations causing dilated cardiomyopathy (DCM). In this model, vesicle distribution is defective due to impaired CME-dependent signaling, resulting in plasma membrane-localized early endosomes. We recapitulated this in iPSC-CMs carrying two different mutations, TPM1-L185F and TnT-R141W (MUT), using 3D confocal imaging as well as super-resolution STED microscopy. We computed scaled distance distributions of EEA1-positive vesicles based on a spherical approximation of the cell. Employing this approach, 3D spherical modelling identified a bi-modal segregation of early endosome populations in MUT iPSC-CMs, compared to WT controls. Moreover, spherical modelling confirmed reversion of the bi-modal vesicle localization in RhoA II-treated MUT iPSC-CMs. This reflects restored, homogeneous distribution of early endosomes within MUT iPSC-CMs following rescue of CME-dependent signaling via RhoA II-dependent RhoA activation. Overall, our approach enables assessment of early endosome distribution in cell-based disease models. This new method may provide further insight into the dynamics of endosome networks in different physiological scenarios.


Subject(s)
Endosomes , Induced Pluripotent Stem Cells , Myocytes, Cardiac , Humans , Induced Pluripotent Stem Cells/metabolism , Endosomes/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Endocytosis , Mutation/genetics , Computer Simulation , rhoA GTP-Binding Protein/metabolism , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Imaging, Three-Dimensional , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Models, Biological , Tropomyosin/metabolism , Tropomyosin/genetics
6.
Adv Exp Med Biol ; 1441: 991-1019, 2024.
Article in English | MEDLINE | ID: mdl-38884766

ABSTRACT

Cardiomyopathies are a heterogeneous group of disorders of the heart muscle that ultimately result in congestive heart failure. Rapid progress in genetics, molecular and cellular biology with breakthrough innovative genetic-engineering techniques, such as next-generation sequencing and multiomics platforms, stem cell reprogramming, as well as novel groundbreaking gene-editing systems over the past 25 years has greatly improved the understanding of pathogenic signaling pathways in inherited cardiomyopathies. This chapter will focus on intracellular and intercellular molecular signaling pathways that are activated by a genetic insult in cardiomyocytes to maintain tissue and organ level regulation and resultant cardiac remodeling in certain forms of cardiomyopathies. In addition, animal models of different clinical forms of human cardiomyopathies with their summaries of triggered key molecules and signaling pathways will be described.


Subject(s)
Cardiomyopathies , Disease Models, Animal , Myocytes, Cardiac , Signal Transduction , Animals , Humans , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Cardiomyopathies/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Signal Transduction/genetics
7.
Heart Fail Clin ; 20(3): 343-352, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38844305

ABSTRACT

Transthyretin amyloid cardiomyopathy (ATTR-CM) is a relatively prevalent cause of morbidity and mortality. Over the recent years, development of disease-modifying treatments has enabled stabilization of the circulating transthyretin tetramer and suppression of its hepatic production, resulting in a remarkable improvement in survival of patients with ATTR-CM. Second-generation drugs for silencing are currently under investigation in randomized clinical trials. In vivo gene editing of transthyretin has been achieving unanticipated suppression of hepatic production in ATTR-CM. Trials of antibodies inducing the active removal of transthyretin amyloid deposits in the heart are ongoing, and evidence has gathered for exceptional spontaneous regression of ATTR-CM.


Subject(s)
Amyloid Neuropathies, Familial , Benzoxazoles , Cardiomyopathies , Prealbumin , Humans , Amyloid Neuropathies, Familial/drug therapy , Amyloid Neuropathies, Familial/metabolism , Cardiomyopathies/drug therapy , Cardiomyopathies/metabolism , Benzoxazoles/therapeutic use , Prealbumin/metabolism , Prealbumin/genetics
8.
Heart Fail Clin ; 20(3): 261-270, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38844297

ABSTRACT

Amyloidosis refers to a heterogeneous group of disorders sharing common pathophysiological mechanisms characterized by the extracellular accumulation of fibrillar deposits consisting of the aggregation of misfolded proteins. Cardiac amyloidosis (CA), usually caused by deposition of misfolded transthyretin or immunoglobulin light chains, is an increasingly recognized cause of heart failure burdened by a poor prognosis. CA manifests with a restrictive cardiomyopathy which progressively leads to biventricular thickening, diastolic and then systolic dysfunction, arrhythmias, and valvular disease. The pathophysiology of CA is multifactorial and includes increased oxidative stress, mitochondrial damage, apoptosis, impaired metabolism, and modifications of intracellular calcium balance.


Subject(s)
Amyloidosis , Cardiomyopathies , Humans , Amyloidosis/physiopathology , Amyloidosis/metabolism , Cardiomyopathies/physiopathology , Cardiomyopathies/metabolism , Heart Failure/physiopathology , Heart Failure/metabolism , Oxidative Stress , Myocardium/pathology , Myocardium/metabolism
9.
Circ Res ; 134(12): 1808-1823, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38843289

ABSTRACT

Mounting experimental and clinical evidence has revealed that adaptive immune mechanisms targeting myocardial antigens are triggered by different forms of cardiac injury and impact disease progression. B and T lymphocytes recognize specific antigens via unique adaptive immune receptors generated through a somatic rearrangement process that generates a potential repertoire of 1019 unique receptors. While the adaptive immune receptor repertoire diversity provides the basis for immunologic specificity, making sense of it can be a challenging task. In the present review, we discuss key aspects underlying the generation of TCRs (T cell receptors) and emerging tools for their study in the context of myocardial diseases. Moreover, we outline how exploring TCR repertoires could lead to a deeper understanding of myocardial pathophysiological principles and potentially serve as diagnostic tools.


Subject(s)
Cardiomyopathies , Receptors, Antigen, T-Cell , Humans , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Animals , Cardiomyopathies/immunology , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Adaptive Immunity , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Myocardium/metabolism , Myocardium/immunology , Myocardium/pathology
10.
Heart Fail Clin ; 20(3): 307-316, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38844301

ABSTRACT

Cardiac amyloidosis (CA) is caused by the myocardial deposition of misfolded proteins, either amyloid transthyretin (ATTR) or immunoglobulin light chains (AL). The paradigm of this condition has transformed, since CA is increasingly recognized as a relatively prevalent cause of heart failure. Cardiac scintigraphy with bone tracers is the unique noninvasive technique able to confirm CA without performing tissue biopsy or advanced imaging tests. A moderate-to-intense myocardial uptake (Perugini grade ≥2) associated with the absence of a monoclonal component is greater than 99% specific for ATTR-CA, while AL-CA confirmation requires tissue biopsy.


Subject(s)
Amyloidosis , Cardiomyopathies , Radiopharmaceuticals , Humans , Cardiomyopathies/diagnostic imaging , Cardiomyopathies/metabolism , Amyloidosis/diagnostic imaging , Amyloidosis/metabolism , Amyloidosis/pathology , Radionuclide Imaging/methods , Bone and Bones/diagnostic imaging , Bone and Bones/metabolism , Bone and Bones/pathology , Myocardium/pathology , Myocardium/metabolism , Amyloid Neuropathies, Familial/diagnostic imaging , Amyloid Neuropathies, Familial/metabolism , Amyloid Neuropathies, Familial/pathology , Heart Failure/diagnostic imaging , Heart Failure/metabolism , Prealbumin/metabolism
11.
Heart Fail Clin ; 20(3): 333-341, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38844304

ABSTRACT

Transthyretin amyloid cardiomyopathy (ATTR-CM) is caused by the myocardial extracellular deposition of amyloid fibrils formed from the dissociation of TTR tetramer into monomers. The rate-limiting step in TTR amyloidogenesis is the dissociation of the TTR tetramer into monomers: Tafamidis is an effective TTR-stabilizer in its native homotetrameric structure. Tafamidis is a safe and effective drug in reducing symptoms, hospitalization and mortality in accurately selected patients affected by hereditary and wild-type transthyretin amyloid cardiomyopathy.


Subject(s)
Amyloid Neuropathies, Familial , Benzoxazoles , Cardiomyopathies , Humans , Benzoxazoles/therapeutic use , Benzoxazoles/pharmacology , Amyloid Neuropathies, Familial/drug therapy , Amyloid Neuropathies, Familial/complications , Amyloid Neuropathies, Familial/genetics , Cardiomyopathies/drug therapy , Cardiomyopathies/metabolism , Prealbumin/genetics , Prealbumin/metabolism
12.
PeerJ ; 12: e17414, 2024.
Article in English | MEDLINE | ID: mdl-38784400

ABSTRACT

Background: Sepsis-induced myocardial injury, as one of the important complications of sepsis, can significantly increase the mortality of septic patients. Our previous study found that nucleolin affected mitochondrial function in energy synthesis and had a protective effect on septic cardiomyopathy in mice. During sepsis, glucose metabolism disorders aggravated myocardial injury and had a negative effect on septic patients. Objectives: We investigated whether nucleolin could regulate glucose metabolism during endotoxemia-induced myocardial injury. Methods: The study tested whether the nucleolin cardiac-specific knockout in the mice could affect glucose metabolism through untargeted metabolomics, and the results of metabolomics were verified experimentally in H9C2 cells. The ATP content, lactate production, and oxygen consumption rate (OCR) were evaluated. Results: The metabolomics results suggested that glycolytic products were increased in endotoxemia-induced myocardial injury, and that nucleolin myocardial-specific knockout altered oxidative phosphorylation-related pathways. The experiment data showed that TNF-α combined with LPS stimulation could increase the lactate content and the OCR values by about 25%, and decrease the ATP content by about 25%. However, interference with nucleolin expression could further decrease ATP content and OCR values by about 10-20% and partially increase the lactate level in the presence of TNF-α and LPS. However, nucleolin overexpression had the opposite protective effect, which partially reversed the decrease in ATP content and the increase in lactate level. Conclusion: Down-regulation of nucleolin can exacerbate glucose metabolism disorders in endotoxemia-induced myocardial injury. Improving glucose metabolism by regulating nucleolin was expected to provide new therapeutic ideas for patients with septic cardiomyopathy.


Subject(s)
Endotoxemia , Glucose , Nucleolin , Phosphoproteins , RNA-Binding Proteins , Animals , Mice , Adenosine Triphosphate/metabolism , Cardiomyopathies/metabolism , Cardiomyopathies/genetics , Cardiomyopathies/etiology , Cell Line , Endotoxemia/metabolism , Glucose/metabolism , Lipopolysaccharides , Metabolomics , Mice, Knockout , Myocardium/metabolism , Myocardium/pathology , Oxidative Phosphorylation , Oxygen Consumption , Phosphoproteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/deficiency , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/genetics
13.
J Proteome Res ; 23(6): 1970-1982, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38718259

ABSTRACT

Lamin A/C (LMNA) is an important component of nuclear lamina. Mutations cause arrhythmia, heart failure, and sudden cardiac death. While LMNA-associated cardiomyopathy typically has an aggressive course that responds poorly to conventional heart failure therapies, there is variability in severity and age of penetrance between and even within specific mutations, which is poorly understood at the cellular level. Further, this heterogeneity has not previously been captured to mimic the heterozygous state, nor have the hundreds of clinical LMNA mutations been represented. Herein, we have overexpressed cardiopathic LMNA variants in HEK cells and utilized state-of-the-art quantitative proteomics to compare the global proteomic profiles of (1) aggregating Q353 K alone, (2) Q353 K coexpressed with WT, (3) aggregating N195 K coexpressed with WT, and (4) nonaggregating E317 K coexpressed with WT to help capture some of the heterogeneity between mutations. We analyzed each data set to obtain the differentially expressed proteins (DEPs) and applied gene ontology (GO) and KEGG pathway analyses. We found a range of 162 to 324 DEPs from over 6000 total protein IDs with differences in GO terms, KEGG pathways, and DEPs important in cardiac function, further highlighting the complexity of cardiac laminopathies. Pathways disrupted by LMNA mutations were validated with redox, autophagy, and apoptosis functional assays in both HEK 293 cells and in induced pluripotent stem cell derived cardiomyocytes (iPSC-CMs) for LMNA N195 K. These proteomic profiles expand our repertoire for mutation-specific downstream cellular effects that may become useful as druggable targets for personalized medicine approach for cardiac laminopathies.


Subject(s)
Lamin Type A , Mutation , Proteomics , Lamin Type A/genetics , Lamin Type A/metabolism , Humans , Proteomics/methods , HEK293 Cells , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Proteome/genetics , Proteome/metabolism , Gene Ontology
14.
Eur J Pharm Sci ; 199: 106794, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38788908

ABSTRACT

Myocardial fibrosis can induce cardiac dysfunction and remodeling. Great attention has been paid to traditional chinese medicine (TCM) 's effectiveness in treating MF. Radix Angelica sinensis (Oliv.) Diels and Radix Astragalus mongholicus Bunge ultrafiltration extract (RAS-RA), which is a key TCM compound preparation, have high efficacy in regulating inflammation. However, studies on its therapeutic effect on radiation-induced myocardial fibrosis (RIMF) are rare. In this study, RAS-RA had therapeutic efficacy in RIMF and elucidated its mechanism of action. First, we formulated the prediction network that described the relation of RAS-RA with RIMF according to data obtained in different databases. Then, we conducted functional enrichment to investigate the functions and pathways associated with potential RIMF targets for RAS-RA. In vivo experiments were also performed to verify these functions and pathways. Second, small animal ultrasound examinations, H&E staining, Masson staining, transmission electron microscopy, Enzyme-linked immunosorbent assay (ELISA), Western-blotting, Immunohistochemical method and biochemical assays were conducted to investigate the possible key anti-RIMF pathway in RAS-RA. In total, 440 targets were detected in those 21 effective components of RAS-RA; meanwhile, 1,646 RIMF-related disease targets were also discovered. After that, PPI network analysis was conducted to identify 20 key targets based on 215 overlap gene targets. As indicated by the gene ontology (GO) and kyoto encyclopedia of genes and genomes (KEGG) analysis results, inflammation and PI3K/AKT/mTOR pathways might have important effects on the therapeutic effects on RIMF. Molecular docking analysis revealed high binding of effective components to targets (affinity < -6 kcal/mol). Based on experimental verification results, RAS-RA greatly mitigated myocardial fibrosis while recovering the cardiac activity of rats caused by X-rays. According to relevant protein expression profiles, the PI3K/AKT/mTOR pathway was important for anti-fibrosis effect of RAS-RA. Experimental studies showed that RAS-RA improved cardiac function, decreased pathological damage and collagen fiber deposition in cardiac tissues, and improved the mitochondrial structure of the heart of rats. RAS-RA also downregulated TNF-α, IL-6, and IL-1ß levels. Additionally, RAS-RA improved the liver and kidney functions and pathological injury of rat kidney and liver tissues, enhanced liver and kidney functions, and protected the liver and kidneys. RAS-RA also increased PI3K, AKT and mTOR protein levels within cardiac tissues and downregulated α-SMA, Collagen I, and Collagen III. The findings of this study suggested that RAS-RA decreased RIMF by suppressing collagen deposition and inflammatory response by inhibiting the PI3K/AKT/mTOR pathway. Thus, RAS-RA was the potential therapeutic agent used to alleviate RIMF.


Subject(s)
Angelica sinensis , Drugs, Chinese Herbal , Fibrosis , Network Pharmacology , Rats, Sprague-Dawley , Animals , Angelica sinensis/chemistry , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/chemistry , Male , Rats , Astragalus Plant/chemistry , Myocardium/pathology , Myocardium/metabolism , Ultrafiltration/methods , Signal Transduction/drug effects , Cardiomyopathies/drug therapy , Cardiomyopathies/etiology , Cardiomyopathies/metabolism , TOR Serine-Threonine Kinases/metabolism
15.
J Am Heart Assoc ; 13(10): e030467, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38761081

ABSTRACT

BACKGROUND: Many cardiomyopathy-associated FLNC pathogenic variants are heterozygous truncations, and FLNC pathogenic variants are associated with arrhythmias. Arrhythmia triggers in filaminopathy are incompletely understood. METHODS AND RESULTS: We describe an individual with biallelic FLNC pathogenic variants, p.Arg650X and c.970-4A>G, with peripartum cardiomyopathy and ventricular arrhythmias. We also describe clinical findings in probands with FLNC variants including Val2715fs87X, Glu2458Serfs71X, Phe106Leu, and c.970-4A>G with hypertrophic and dilated cardiomyopathy, atrial fibrillation, and ventricular tachycardia. Induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) were generated. The FLNC truncation, Arg650X/c.970-4A>G, showed a marked reduction in filamin C protein consistent with biallelic loss of function mutations. To assess loss of filamin C, gene editing of a healthy control iPSC line was used to generate a homozygous FLNC disruption in the actin binding domain. Because filamin C has been linked to protein quality control, we assessed the necessity of filamin C in iPSC-CMs for response to the proteasome inhibitor bortezomib. After exposure to low-dose bortezomib, FLNC-null iPSC-CMs showed an increase in the chaperone proteins BAG3, HSP70 (heat shock protein 70), and HSPB8 (small heat shock protein B8) and in the autophagy marker LC3I/II. FLNC null iPSC-CMs had prolonged electric field potential, which was further prolonged in the presence of low-dose bortezomib. FLNC null engineered heart tissues had impaired function after low-dose bortezomib. CONCLUSIONS: FLNC pathogenic variants associate with a predisposition to arrhythmias, which can be modeled in iPSC-CMs. Reduction of filamin C prolonged field potential, a surrogate for action potential, and with bortezomib-induced proteasome inhibition, reduced filamin C led to greater arrhythmia potential and impaired function.


Subject(s)
Filamins , Proteostasis , Filamins/genetics , Filamins/metabolism , Humans , Female , Induced Pluripotent Stem Cells/metabolism , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/physiopathology , Arrhythmias, Cardiac/etiology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Cardiomyopathies/physiopathology , Male , Adult , Mutation , Bortezomib/pharmacology
16.
BMC Cardiovasc Disord ; 24(1): 286, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38816686

ABSTRACT

Septic cardiomyopathy is one of the most severe and common complications in patients with sepsis and poses a great threat to their prognosis. However, the potential mechanisms and effective therapeutic drugs need to be explored. The control of cardiac cell death by miRNAs has emerged as a prominent area of scientific interest in the diagnosis and treatment of heart disorders in recent times. In the present investigation, we discovered that overexpression of miR-31-5p prevented LPS-induced damage to H9C2 cells and that miR-31-5p could inhibit BAP1 production by binding to its 3'-UTR. BRCA1-Associated Protein 1 (BAP1) is a ubiquitin carboxy-terminal hydrolase. BAP1 upregulation blocked effect of miR-31-5p on H9C2 cell injury. Moreover, BAP1 inhibited the expression of solute carrier family 7 member 11 (SLC7A11) by deubiquitinating histone 2 A (H2Aub) on the promoter of SLC7A11. Furthermore, overexpression of miR-31-5p and downregulation of BAP1 inhibited SLC7A11 mediated ferroptosis. In addition, the downregulation of SLC7A11 reversed the inhibitory effect of miR-31-5p on the expression of myocardial injury and inflammatory factors, and cell apoptosis was reversed. In conclusion, these results indicate that miR-31-5p alleviates malignant development of LPS-induced H9C2 cell injury by targeting BAP1 and regulating SLC7A11 deubiquitination-mediated ferroptosis, which confirmed the protective effect of miR-31-5p on H9C2 cell injury and revealed potential mechanisms that may provide new targets for treatment of septic cardiomyopathy.


Subject(s)
Amino Acid Transport System y+ , Cardiomyopathies , Ferroptosis , MicroRNAs , Myocytes, Cardiac , Sepsis , Signal Transduction , Tumor Suppressor Proteins , Ubiquitin Thiolesterase , Ubiquitination , MicroRNAs/genetics , MicroRNAs/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myocytes, Cardiac/drug effects , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism , Cardiomyopathies/metabolism , Cardiomyopathies/genetics , Ferroptosis/drug effects , Ferroptosis/genetics , Animals , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Sepsis/genetics , Sepsis/metabolism , Cell Line , Amino Acid Transport System y+/genetics , Amino Acid Transport System y+/metabolism , Rats , Disease Models, Animal , Humans , Gene Expression Regulation , Lipopolysaccharides/pharmacology , Male
17.
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
18.
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
19.
Dis Model Mech ; 17(5)2024 May 01.
Article in English | MEDLINE | ID: mdl-38770680

ABSTRACT

Absence of dystrophin results in muscular weakness, chronic inflammation and cardiomyopathy in Duchenne muscular dystrophy (DMD). Pharmacological corticosteroids are the DMD standard of care; however, they have harsh side effects and unclear molecular benefits. It is uncertain whether signaling by physiological corticosteroids and their receptors plays a modifying role in the natural etiology of DMD. Here, we knocked out the glucocorticoid receptor (GR, encoded by Nr3c1) specifically in myofibers and cardiomyocytes within wild-type and mdx52 mice to dissect its role in muscular dystrophy. Double-knockout mice showed significantly worse phenotypes than mdx52 littermate controls in measures of grip strength, hang time, inflammatory pathology and gene expression. In the heart, GR deletion acted additively with dystrophin loss to exacerbate cardiomyopathy, resulting in enlarged hearts, pathological gene expression and systolic dysfunction, consistent with imbalanced mineralocorticoid signaling. The results show that physiological GR functions provide a protective role during muscular dystrophy, directly contrasting its degenerative role in other disease states. These data provide new insights into corticosteroids in disease pathophysiology and establish a new model to investigate cell-autonomous roles of nuclear receptors and mechanisms of pharmacological corticosteroids.


Subject(s)
Dystrophin , Mice, Inbred mdx , Mice, Knockout , Receptors, Glucocorticoid , Animals , Receptors, Glucocorticoid/metabolism , Dystrophin/metabolism , Dystrophin/genetics , Dystrophin/deficiency , Myocardium/pathology , Myocardium/metabolism , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Duchenne/metabolism , Muscle, Skeletal/pathology , Muscle, Skeletal/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myocytes, Cardiac/drug effects , Mice , Cardiomyopathies/pathology , Cardiomyopathies/metabolism , Mice, Inbred C57BL , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Animal/metabolism , Phenotype , Systole/drug effects
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...