Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
1.
Article in English | MEDLINE | ID: mdl-35182256

ABSTRACT

To evaluate the association between circulating levels of inflammatory cytokines and the occurrence of post-percutaneous coronary intervention (PCI) coronary slow flow (CSF) in patients with non-ST segment elevation acute coronary syndrome (NSTE-ACS). CSF after PCI commonly occurs and implies poor outcomes, while the determinants of post-PCI CSF in patients with NSTE-ACS remain controversial. In this multicenter case control study, 176 patients diagnosed with NSTE-ACS and with post-PCI CSF occurred composed of CSF group, while 352 matched NSTE-ACS patients composed control group. Corrected thrombolysis in myocardial infarction frame count (cTFC), circulating levels of inflammatory cytokines and PCI related parameters were analyzed using Logistic regression models. Among 528 patients with median age of 67 (59-76) and male proportion of 65.5%, 176 (35.0%) patients had occurrence of post-PCI CSF defined as cTFC ≥ 24. Patients with CSF presented more intense inflammatory activity revealed by higher levels of white blood cell, high-sensitivity C-reactive protein (hs-CRP), interleukin-1ß (IL-1ß), soluble IL-2 receptor (sIL-2R), IL-6, IL-8, IL-10 and tumor necrosis factor-α (TNF-α), while PCI related parameters were comparable. Correlation analysis showed cTFC was positively correlated with those inflammatory cytokines. Logistic regression model indicates that hs-CRP (odds ratio (OR) = 3.038, 95% confidence interval (CI) 1.545-5.975), sIL-2R (OR = 2.103, 95% CI 1.959-4.026) and TNF-α (OR = 3.708, 95% CI 1.426-9.641) were valuable predictors for CSF occurrence. Elevated circulating levels of inflammatory cytokine including hs-CRP, sIL-2R and TNF-α rather than PCI related parameters could predict post-PCI CSF in patients with NSTE-ACS.

2.
J Cell Mol Med ; 24(21): 12799-12812, 2020 11.
Article in English | MEDLINE | ID: mdl-32945126

ABSTRACT

Endothelial-mesenchymal transition (EndMT) is a major source of transformed cardiac fibroblasts, which is reported to play a key role in cardiac fibrosis (CF), a pathogenesis of cardiovascular diseases such as heart failure, myocardial infarction and atrial fibrillation. Nonetheless, the specific mechanism underlying the progression of EndMT to CF is still largely unknown. In this study, we aimed to investigate the role of milk fat globule-EGF factor 8 (MFGE8), a kind of soluble glycoprotein, in TGF-ß1-induced EndMT. In animal experiments, the expression of MFGE8 was found down-regulated in the left ventricle and aorta of rats after transverse aortic constriction (TAC) compared with the sham group, especially in endothelial cells (ECs). In in vitro cultured ECs, silencing MFGE8 with small interfering RNA (siRNA) was found to promote the process of TGF-ß1-induced EndMT, whereas administration of recombinant human MFGE8 (rh-MFGE8) attenuated the process. Moreover, activated Smad2/3 signalling pathway after TGF-ß1 treatment and EndMT-related transcription factors, such as Snail, Twist and Slug, was potentiated by MFGE8 knock-down but inhibited by rh-MFGE8. In conclusion, our experiments indicate that MFGE8 might play a protective role in TGF-ß1-induced EndMT and might be a potential therapeutic target for cardiac fibrosis.


Subject(s)
Antigens, Surface/genetics , Down-Regulation/genetics , Human Umbilical Vein Endothelial Cells/metabolism , Mesoderm/metabolism , Milk Proteins/genetics , Myocardium/pathology , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Snail Family Transcription Factors/metabolism , Animals , Antigens, Surface/metabolism , Cell Movement/drug effects , Cells, Cultured , Down-Regulation/drug effects , Fibrosis , Gene Silencing/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Male , Milk Proteins/metabolism , Neovascularization, Physiologic/drug effects , RNA, Small Interfering/metabolism , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , Signal Transduction , Transcription Factors/metabolism , Transforming Growth Factor beta1/metabolism
3.
Biochem Biophys Res Commun ; 528(3): 545-553, 2020 07 30.
Article in English | MEDLINE | ID: mdl-32505342

ABSTRACT

Diabetic cardiomyopathy (DCM) is one of the main causes of heart failure in patients with diabetes. Cardiac fibrosis caused by endothelial mesenchymal transformation (EndMT) plays an important role in the pathogenesis of DCM. NLRC5 is a recently discovered immune and inflammatory regulatory molecule in the NOD-like receptor family, and is involved in organ fibrosis. In this study, we found that the expression of NLRC5 was up-regulated in endothelial cells (ECs) and cardiac fibroblasts (CFs) in diabetes models both in vivo and in vitro. NLRC5 knockdown significantly inhibited high glucose-induced EndMT. In addition, NLRC5 deficiency inhibited the expression of phosphorylated Smad2/3 and the activation of EndMT-related transcription factors in ECs induced by high glucose. However, the effect of NLRC5 deficiency on CFs was not obvious. In summary, our results suggest that NLRC5 deficiency ameliorates cardiac fibrosis in DCM by inhibiting EndMT through Smad2/3 signaling pathway and related transcription factors. NLRC5 is likely to be a biomarker and therapeutic target of cardiac fibrosis in diabetic cardiomyopathy.


Subject(s)
Diabetic Cardiomyopathies/metabolism , Diabetic Cardiomyopathies/pathology , Intracellular Signaling Peptides and Proteins/deficiency , Animals , Cell Transdifferentiation , Cells, Cultured , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetic Cardiomyopathies/prevention & control , Fibrosis , Gene Knockdown Techniques , Glucose/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Myocardium/metabolism , Myocardium/pathology , Signal Transduction , Smad Proteins, Receptor-Regulated/metabolism
4.
J Mol Cell Cardiol ; 139: 164-175, 2020 02.
Article in English | MEDLINE | ID: mdl-31958465

ABSTRACT

Atrial fibrillation (AF) is characterized by potentiated growth of atrial fibroblasts and excessive deposition of the extracellular matrix. Atrial fibrosis has emerged as a hallmark of atrial structural remodeling linked to AF. Nonetheless, the specific mechanism underlying the progression of atrial fibrosis to AF is still largely unknown. MFGE8 (milk fat globule-EGF factor 8) is a soluble glycoprotein associated with many human diseases. Recently, a number of studies revealed that MFGE8 plays a crucial role in heart disease. Yet, MFGE8 regulation and function in the process of atrial fibrosis and vulnerability to AF remain unexplored. In this study, we found that the expression of MFGE8 was downregulated in the atriums of patients with AF compared with individuals without AF. In addition, the expression of MFGE8 was lower in atriums of angiotensin II (Ang-II)-stimulated rats as compared with the sham group. In vitro, silencing of MFGE8 by small interfering RNA significantly increased Ang-II-induced atrial fibrosis, whereas administration of recombinant human MFGE8 (rhMFGE8) attenuated the atrial fibrosis. Moreover, we found that the activated TGF-ß1/Smad2/3 pathway after Ang-II treatment was significantly potentiated by the MFGE8 knockdown but inhibited by rhMFGE8 in vitro. Inhibition of integrin ß3 which is the receptor for MFGE8, suppressed the TGF-ß1/Smad2/3 activating effects of the MFGE8 knockdown in Ang-II-treated rat atrial fibroblasts. Finally, we administered rhMFGE8 to rats; it attenuated atrial fibrosis and remodeling and further reduced AF vulnerability induced by Ang-II, indicating that MFGE8 might have the potential both as a novel biomarker and as a therapeutic target in atrial fibrosis and AF.


Subject(s)
Antigens, Surface/metabolism , Atrial Fibrillation/metabolism , Atrial Fibrillation/pathology , Heart Atria/pathology , Milk Proteins/metabolism , Signal Transduction , Smad Proteins/metabolism , Transforming Growth Factor beta1/metabolism , Angiotensin II , Animals , Antigens, Surface/genetics , Atrial Fibrillation/genetics , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis , Gene Expression Regulation/drug effects , Humans , Integrin beta3/metabolism , Male , Milk Proteins/genetics , Models, Biological , Protein Binding/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...