structural remodeling
Recently Published Documents


TOTAL DOCUMENTS

485
(FIVE YEARS 114)

H-INDEX

52
(FIVE YEARS 7)

2021 ◽  
Vol 12 ◽  
Author(s):  
Yudi Zhang ◽  
Yuping Fu ◽  
Tiannan Jiang ◽  
Binghua Liu ◽  
Hongke Sun ◽  
...  

Atrial fibrillation (AF) is the most common sustained cardiac arrhythmia in clinical setting. Its pathogenesis was associated with metabolic disorder, especially defective fatty acids oxidation (FAO). However, whether promoting FAO could prevent AF occurrence and development remains elusive. In this study, we established a mouse model of obesity-related AF through high-fat diet (HFD) feeding, and used l-carnitine (LCA, 150 mg/kg⋅BW/d), an endogenous cofactor of carnitine palmitoyl-transferase-1B (CPT1B; the rate-limiting enzyme of FAO) to investigate whether FAO promotion can attenuate the AF susceptibility in obesity. All mice underwent electrophysiological assessment for atrial vulnerability, and echocardiography, histology and molecular evaluation for AF substrates and underlying mechanisms, which were further validated by pharmacological experiments in vitro. HFD-induced obese mice increased AF vulnerability and exhibited apparent atrial structural remodeling, including left atrial dilation, cardiomyocyte hypertrophy, connexin-43 remodeling and fibrosis. Pathologically, HFD apparently leads to defective cardiac FAO and subsequent lipotoxicity, thereby evoking a set of pathological reactions including oxidative stress, DNA damage, inflammation, and insulin resistance. Enhancing FAO via LCA attenuated lipotoxicity and lipotoxicity-induced pathological changes in the atria of obese mice, resulting in restored structural remodeling and ameliorated AF susceptibility. Mechanistically, LCA activated AMPK/PGC1α signaling both in vivo and in vitro, and pharmacological inhibition of AMPK via Compound C attenuated LCA-induced cardio-protection in palmitate-treated primary atrial cardiomyocytes. Taken together, our results demonstrated that FAO promotion via LCA attenuated obesity-mediated AF and structural remodeling by activating AMPK signaling and alleviating atrial lipotoxicity. Thus, enhancing FAO may be a potential therapeutic target for AF.


2021 ◽  
Vol 12 ◽  
Author(s):  
Martin Mollenhauer ◽  
Senai Bokredenghel ◽  
Simon Geißen ◽  
Anna Klinke ◽  
Tobias Morstadt ◽  
...  

The six-transmembrane protein of prostate 2 (Stamp2) acts as an anti-inflammatory protein in macrophages by protecting from overt inflammatory signaling and Stamp2 deficiency accelerates atherosclerosis in mice. Herein, we describe an unexpected role of Stamp2 in polymorphonuclear neutrophils (PMN) and characterize Stamp2’s protective effects in myocardial ischemic injury. In a murine model of ischemia and reperfusion (I/R), echocardiography and histological analyses revealed a pronounced impairment of cardiac function in hearts of Stamp2-deficient- (Stamp2-/-) mice as compared to wild-type (WT) animals. This difference was driven by aggravated cardiac fibrosis, as augmented fibroblast-to-myofibroblast transdifferentiation was observed which was mediated by activation of the redox-sensitive p38 mitogen-activated protein kinase (p38 MAPK). Furthermore, we observed increased production of reactive oxygen species (ROS) in Stamp2-/- hearts after I/R, which is the likely cause for p38 MAPK activation. Although myocardial macrophage numbers were not affected by Stamp2 deficiency after I/R, augmented myocardial infiltration by polymorphonuclear neutrophils (PMN) was observed, which coincided with enhanced myeloperoxidase (MPO) plasma levels. Primary PMN isolated from Stamp2-/- animals exhibited a proinflammatory phenotype characterized by enhanced nuclear factor (NF)-κB activity and MPO secretion. To prove the critical role of PMN for the observed phenotype after I/R, antibody-mediated PMN depletion was performed in Stamp2-/- mice which reduced deterioration of LV function and adverse structural remodeling to WT levels. These data indicate a novel role of Stamp2 as an anti-inflammatory regulator of PMN and fibroblast-to-myofibroblast transdifferentiation in myocardial I/R injury.


2021 ◽  
pp. JN-RM-0871-21
Author(s):  
Kristina D. Micheva ◽  
Marianna Kiraly ◽  
Marc M. Perez ◽  
Daniel V. Madison

PLoS ONE ◽  
2021 ◽  
Vol 16 (9) ◽  
pp. e0257435
Author(s):  
Miyako Nakano ◽  
Susana Sabido-Bozo ◽  
Kouta Okazaki ◽  
Auxiliadora Aguilera-Romero ◽  
Sofia Rodriguez-Gallardo ◽  
...  

Glycosylphosphatidylinositol (GPI) anchoring of proteins is an essential post-translational modification in all eukaryotes that occurs at the endoplasmic reticulum (ER) and serves to deliver GPI-anchored proteins (GPI-APs) to the cell surface where they play a wide variety of vital physiological roles. This paper describes a specialized method for purification and structural analysis of the GPI glycan of individual GPI-APs in yeast. The protocol involves the expression of a specific GPI-AP tagged with GFP, enzymatic release from the cellular membrane fraction, immunopurification, separation by electrophoresis and analysis of the peptides bearing GPI glycans by mass spectrometry after trypsin digestion. We used specifically this protocol to address the structural remodeling that undergoes the GPI glycan of a specific GPI-AP during its transport to the cell surface. This method can be also applied to investigate the GPI-AP biosynthetic pathway and to directly confirm predicted GPI-anchoring of individual proteins.


Author(s):  
Jing Zhang ◽  
Yan Liang ◽  
William H. Bradford ◽  
Farah Sheikh

AbstractDesmosomes are critical adhesion structures in cardiomyocytes, with mutation/loss linked to the heritable cardiac disease, arrhythmogenic right ventricular cardiomyopathy (ARVC). Early studies revealed the ability of desmosomal protein loss to trigger ARVC disease features including structural remodeling, arrhythmias, and inflammation; however, the precise mechanisms contributing to diverse disease presentations are not fully understood. Recent mechanistic studies demonstrated the protein degradation component CSN6 is a resident cardiac desmosomal protein which selectively restricts cardiomyocyte desmosomal degradation and disease. This suggests defects in protein degradation can trigger the structural remodeling underlying ARVC. Additionally, a subset of ARVC-related mutations show enhanced vulnerability to calpain-mediated degradation, further supporting the relevance of these mechanisms in disease. Desmosomal gene mutations/loss has been shown to impact arrhythmogenic pathways in the absence of structural disease within ARVC patients and model systems. Studies have shown the involvement of connexins, calcium handling machinery, and sodium channels as early drivers of arrhythmias, suggesting these may be distinct pathways regulating electrical function from the desmosome. Emerging evidence has suggested inflammation may be an early mechanism in disease pathogenesis, as clinical reports have shown an overlap between myocarditis and ARVC. Recent studies focus on the association between desmosomal mutations/loss and inflammatory processes including autoantibodies and signaling pathways as a way to understand the involvement of inflammation in ARVC pathogenesis. A specific focus will be to dissect ongoing fields of investigation to highlight diverse pathogenic pathways associated with desmosomal mutations/loss.


2021 ◽  
Vol 129 (Suppl_1) ◽  
Author(s):  
Arvind Sridhar ◽  
Liang Hong ◽  
Olivia T Ly ◽  
Hanna Chen ◽  
Srikanth Perike ◽  
...  

Background: Inducible atrial fibrillation (AF) in diet-induced obese (DIO) mice is mediated in part by a combined effect of ion channel remodeling and atrial fibrosis. NADPH oxidase 2 (NOX2), a major source of reactive oxygen species (ROS) production in human atria, is increased in DIO mice. Although a mitochondrial antioxidant (MitoTEMPO) reduced AF burden, and reversed ion channel and structural remodeling, the role of NOX2 in increased ROS production and atrial remodeling in obesity-induced AF remains unclear. Objective: To test the hypothesis that increased NOX2 modulates atrial remodeling in obesity-induced AF, we treated DIO mice with apocynin, NOX2 inhibitor, and fed a 60% high fat diet (HFD) to Nox2 -KO mice. Methods: Weight, BP, plasma glucose, trans-esophageal rapid (TE) pacing and F 2 -isoprostanes were measured in DIO mice and compared to controls. Echocardiography, electrophysiology (EP), immunohistochemistry, Western blotting, cellular patch clamping and optical mapping studies were performed. Results: The average weight of DIO mice treated with apocynin (DIO-A) and vehicle control mice was 38.4 ± 3.8 g versus 44.0 ± 7.5 g versus 31.7 ± 1.19 g respectively (P≤0.0001). Both groups of DIO mice displayed progressive increase in weight over 10 weeks of HFD + drug treatment compared to controls (P≤0.0001). After TE pacing, DIO mice treated with apocynin showed significantly reduced pacing-induced AF burden when compared to DIO mice treated with vehicle. DIO mice treated with apocynin displayed 20.2 ± 26.1 sec versus 162.3 ± 133.7 sec and 18.3 ± 18.1 sec in DIO mice treated with vehicle and control mice respectively (P≤0.0001). Western blotting experiments showed that potassium channels, Kv7.1 and Kv1.5 protein expression is restored in Nox2 -KO HFD mice compared to DIO mice thus indicating restoration of ion channel remodeling upon Nox2 inhibition. Conclusions: We showed that genetic and pharmacological inhibition of NOX2 abrogates ion channel remodeling and reverses obesity-induced AF burden. Our findings may have important implications for the management of obesity-mediated AF in patients.


2021 ◽  
Vol 158 ◽  
pp. 72-81
Author(s):  
Jiajie Yan ◽  
Cheryl Killingsworth ◽  
Greg Walcott ◽  
Yujie Zhu ◽  
Silvio Litovsky ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document