Cardiac fibroblasts support cardiac inflammation in heart failure

2014 ◽  
Vol 109 (5) ◽  
Author(s):  
Diana Lindner ◽  
Christin Zietsch ◽  
Juliane Tank ◽  
Samuel Sossalla ◽  
Nina Fluschnik ◽  
...  
2020 ◽  
Vol 41 (Supplement_2) ◽  
Author(s):  
T Thottakara ◽  
V.M Dhople ◽  
S Voss ◽  
J Schoen ◽  
U Voelker ◽  
...  

Abstract Introduction Cardiac inflammation with subsequent remodeling of the extracellular matrix (ECM) with fibrosis formation is an important precursor of heart failure. In myocardial inflammation release of cytokines, such as TNF-α was previously linked to the development of fibrosis. However, their exact role in fibroblasts activation, leading to their transformation into myofibroblasts and ultimately fibrosis is unknown. Therefore, the purpose of this work was to study the secretome of isolated cardiac fibroblasts of heart failure patients in vitro. Methods and results Human cardiac fibroblasts were obtained from endomyocardial biopsies of patients with reduced ejection fraction. Secretome- and gene expression analysis was performed on TNF-α (10 ng/ml) and TGF-β (5 ng/ml) treated and untreated fibroblasts. The analysis of the secretome in cell culture supernatant of these fibroblasts was performed by nano-LC-ESI tandem mass spectrometry. Subsequently, RNA was isolated from fibroblasts and expression analysis was performed using affymetrix gene chips and verified for the stimulation experiments using TaqMan. The secretome and gene expression studies are constistent regarding the most frequent ECM components. The proteins involved in the construction of the ECM accounted for 56% of the total protein intensity. The fibronectin represented the largest proportion of ECM proteins (50%), with collagens accounting for 29% and MMPs and TIMPs for approximately 5% and a small proportion of cytokines. In the secretome as well as in the gene expression, Il-6 and Il-8 showed the highest proportion of cytokines and type I as well as type IV (each approx. 40%) of collagens. MMP2, MMP1 and TIMP1 accounted for the largest proportion of MMPs and TIMPs. Stimulation with TGF-β and TNF-α led to changes in the composition of the cytokines and ECM proteins. After the stimulation with TNF-α, the proportion of cytokines increased from 3.1% to 6.6%, while the proportion of ECM proteins decreased slightly. Accordingly, after stimulation with TGF-β a well-known inductor of fibrosis, the proportion of collagens and other matrix proteins increased, while the proportion of cytokines decreased slightly. Conclusion In this study, we identified the most abundant proteins of the ECM and demonstrated their expansion caused by TGF-β stimulation of fibroblasts. Surprisingly, TNF-α led to an increase of cytokines secreted by the fibroblasts with a minimal reduction of ECM proteins. Here, the differentiated regulation of the fibroblast in the transformation of the ECM could be seen. TNF-α stimulation demonstrated to increase gene expression of the cytokines in fibroblasts, which in turn maintain inflammation with chemotactic effect. Hence, cardiac fibroblasts seem to be supporting cells for cardiac inflammation. There seemed to be no pro-fibrotic effect. In this work we could show the pro-inflammatory role of the cardiac fibroblast upon inflammatory stimulation. Funding Acknowledgement Type of funding source: None


2020 ◽  
Vol 134 (1) ◽  
pp. 71-72
Author(s):  
Naseer Ahmed ◽  
Masooma Naseem ◽  
Javeria Farooq

Abstract Recently, we have read with great interest the article published by Ibarrola et al. (Clin. Sci. (Lond.) (2018) 132, 1471–1485), which used proteomics and immunodetection methods to show that Galectin-3 (Gal-3) down-regulated the antioxidant peroxiredoxin-4 (Prx-4) in cardiac fibroblasts. Authors concluded that ‘antioxidant activity of Prx-4 had been identified as a protein down-regulated by Gal-3. Moreover, Gal-3 induced a decrease in total antioxidant capacity which resulted in a consequent increase in peroxide levels and oxidative stress markers in cardiac fibroblasts.’ We would like to point out some results stated in the article that need further investigation and more detailed discussion to clarify certain factors involved in the protective role of Prx-4 in heart failure.


Author(s):  
Amrita Sarkar ◽  
Khadija Rafiq

Cardiovascular Disease (CVD) is a class of diseases that involve disorders of heart and blood vessels, including hypertension, coronary heart disease, cerebrovascular disease, peripheral vascular disease, which finally lead to Heart Failure (HF). There are several treatments available all over the world, but still, CVD and heart failure became the number one problem causing death every year worldwide. Both experimental and clinical studies have shown a role for inflammation in the pathogenesis of heart failure. This seems related to an imbalance between pro-inflammatory and anti-inflammatory cytokines. Cardiac inflammation is a major pathophysiological mechanism operating in the failing heart, regardless of HF aetiology. Disturbances of the cellular and humoral immune system are frequently observed in heart failure. This review describes how B-cells play a specific role in the heart failure states. There is an urgent need to identify novel therapeutic targets and develop advanced therapeutic strategies to combat the syndrome of HF. Understanding and describing the elements of the humoral immunity function are essential and may suggest potential new treatment strategies.


Cells ◽  
2021 ◽  
Vol 10 (3) ◽  
pp. 557
Author(s):  
Stephanie D. Burr ◽  
James A. Stewart

Cardiovascular disease, specifically heart failure, is a common complication for individuals with type 2 diabetes mellitus. Heart failure can arise with stiffening of the left ventricle, which can be caused by “active” cardiac fibroblasts (i.e., myofibroblasts) remodeling the extracellular matrix (ECM). Differentiation of fibroblasts to myofibroblasts has been demonstrated to be an outcome of AGE/RAGE signaling. Hyperglycemia causes advanced glycated end products (AGEs) to accumulate within the body, and this process is greatly accelerated under chronic diabetic conditions. AGEs can bind and activate their receptor (RAGE) to trigger multiple downstream outcomes, such as altering ECM remodeling, inflammation, and oxidative stress. Previously, our lab has identified a small GTPase, Rap1a, that possibly overlaps the AGE/RAGE signaling cascade to affect the downstream outcomes. Rap1a acts as a molecular switch connecting extracellular signals to intracellular responses. Therefore, we hypothesized that Rap1a crosses the AGE/RAGE cascade to alter the expression of AGE/RAGE associated signaling proteins in cardiac fibroblasts in type 2 diabetic mice. To delineate this cascade, we used genetically different cardiac fibroblasts from non-diabetic, diabetic, non-diabetic RAGE knockout, diabetic RAGE knockout, and Rap1a knockout mice and treated them with pharmacological modifiers (exogenous AGEs, EPAC, Rap1a siRNA, and pseudosubstrate PKC-ζ). We examined changes in expression of proteins implicated as markers for myofibroblasts (α-SMA) and inflammation/oxidative stress (NF-κB and SOD-1). In addition, oxidative stress was also assessed by measuring hydrogen peroxide concentration. Our results indicated that Rap1a connects to the AGE/RAGE cascade to promote and maintain α-SMA expression in cardiac fibroblasts. Moreover, Rap1a, in conjunction with activation of the AGE/RAGE cascade, increased NF-κB expression as well as hydrogen peroxide concentration, indicating a possible oxidative stress response. Additionally, knocking down Rap1a expression resulted in an increase in SOD-1 expression suggesting that Rap1a can affect oxidative stress markers independently of the AGE/RAGE signaling cascade. These results demonstrated that Rap1a contributes to the myofibroblast population within the heart via AGE/RAGE signaling as well as promotes possible oxidative stress. This study offers a new potential therapeutic target that could possibly reduce the risk for developing diabetic cardiovascular complications attributed to AGE/RAGE signaling.


2021 ◽  
Vol 23 (7) ◽  
Author(s):  
Sally Yu Shi ◽  
Xin Luo ◽  
Tracy M. Yamawaki ◽  
Chi-Ming Li ◽  
Brandon Ason ◽  
...  

Abstract Purpose of Review Cardiac fibroblast activation contributes to fibrosis, maladaptive remodeling and heart failure progression. This review summarizes the latest findings on cardiac fibroblast activation dynamics derived from single-cell transcriptomic analyses and discusses how this information may aid the development of new multispecific medicines. Recent Findings Advances in single-cell gene expression technologies have led to the discovery of distinct fibroblast subsets, some of which are more prevalent in diseased tissue and exhibit temporal changes in response to injury. In parallel to the rapid development of single-cell platforms, the advent of multispecific therapeutics is beginning to transform the biopharmaceutical landscape, paving the way for the selective targeting of diseased fibroblast subpopulations. Summary Insights gained from single-cell technologies reveal critical cardiac fibroblast subsets that play a pathogenic role in the progression of heart failure. Combined with the development of multispecific therapeutic agents that have enabled access to previously “undruggable” targets, we are entering a new era of precision medicine.


2021 ◽  
Vol 2021 ◽  
pp. 1-8
Author(s):  
Lili Wang ◽  
Qianhui Zhang ◽  
Kexin Yuan ◽  
Jing Yuan

The incidence rate of cardiovascular disease (CVD) has been increasing year by year and has become the main cause for the increase of mortality. Mitochondrial DNA (mtDNA) plays a crucial role in the pathogenesis of CVD, especially in heart failure and ischemic heart diseases. With the deepening of research, more and more evidence showed that mtDNA is related to the occurrence and development of CVD. Current studies mainly focus on how mtDNA copy number, an indirect biomarker of mitochondrial function, contributes to CVD and its underlying mechanisms including mtDNA autophagy, the effect of mtDNA on cardiac inflammation, and related metabolic functions. However, no relevant studies have been conducted yet. In this paper, we combed the current research status of the mechanism related to the influence of mtDNA on the occurrence, development, and prognosis of CVD, so as to find whether these mechanisms have something in common, or is there a correlation between each mechanism for the development of CVD?


2019 ◽  
Author(s):  
Jiangbin Wu ◽  
Kadiam C Venkata Subbaiah ◽  
Li Huitong Xie ◽  
Feng Jiang ◽  
Deanne Mickelsen ◽  
...  

AbstractRationaleIncreased protein synthesis of pro-fibrotic genes is a common feature of cardiac fibrosis, a major manifestation of heart failure. Despite this important observation, critical factors and molecular mechanisms for translational control of pro-fibrotic genes during cardiac fibrosis remain unclear.ObjectiveThis study aimed to test the hypothesis that cardiac stress-induced expression of a bifunctional aminoacyl-tRNA synthetase (ARS), glutamyl-prolyl-tRNA synthetase (EPRS), is preferentially required for the translation of proline codon-rich (PRR) pro-fibrotic mRNAs in cardiac fibroblasts during cardiac fibrosis.Methods and ResultsBy analyses of multiple available unbiased large-scale screening datasets of human and mouse heart failure, we have discovered that EPRS acts as an integrated node among all the ARSs in various cardiac pathogenic processes. We confirmed that EPRS was induced at both mRNA and protein level (∼1.5-2.5 fold increase) in failing hearts compared with non-failing hearts using our cohort of human and mouse heart samples. Genetic knockout of one allele of Eprs globally (Eprs+/-) using CRISPR-Cas9 technology or in a myofibroblast-specific manner (Eprsflox/+; PostnMCM/+) strongly reduces cardiac fibrosis (∼50% reduction) in isoproterenol- and transverse aortic constriction-induced heart failure mouse models. Inhibition of EPRS by a prolyl-tRNA synthetase (PRS)-specific inhibitor, halofuginone (Halo), significantly decreased the translation efficiency of proline-rich collagens in cardiac fibroblasts. Furthermore, using transcriptome-wide RNA-Seq and polysome profiling-Seq in Halo-treated fibroblasts, we identified multiple novel Pro-rich genes in addition to collagens, such as Ltbp2 and Sulf1, which are translationally regulated by EPRS. As a major EPRS downstream effector, SULF1 is highly enriched in human and mouse myofibroblast. siRNA-mediated knockdown of SULF1 attenuates cardiac myofibroblast activation and collagen deposition.ConclusionsOur results indicate that EPRS preferentially controls the translational activation of proline codon-rich pro-fibrotic genes in cardiac fibroblasts and augments pathological cardiac remodeling.Novelty and SignificanceWhat is known?TGF-β and IL-11 increase synthesis of pro-fibrotic proteins during cardiac fibrosis.Many pro-fibrotic genes contain Pro genetic codon rich motifs such as collagens.EPRS is an essential house-keeping enzyme required for ligating Pro to tRNAPro for the synthesis of Pro-containing proteins.What New Information Does This Article Contribute?This study is a pioneering investigation of translational control mechanisms of pro-fibrotic gene expression in cardiac fibrosis.EPRS mRNA and protein expression are induced in failing human hearts and mouse hearts undergoing pathological cardiac remodeling.The first demonstration of the in vivo function of EPRS in cardiac remodeling. Heterozygous Eprs global knockout and myofibroblast-specific tamoxifen-inducible Eprs conditional knockout mice show reduced pathological cardiac fibrosis under stress, suggesting that the reduction of EPRS is cardioprotective.Identification of novel preferential translational target genes of EPRS. We found that EPRS regulates translation of Pro-rich (PRR) transcripts, which comprise most of the ECM and secretory signaling molecules. Among those targets, we identified multiple novel PRR genes such as LTBP2 and SULF1.SULF1 is validated as a myofibroblast marker protein in human and mouse heart failure and a potential anti-fibrosis target gene.In cardiac fibroblasts, the synthesis of pro-fibrotic proteins is upregulated by cardiac stressors to activate extracellular matrix deposition and impair cardiac function. In this study, we have discovered an EPRS-PRR gene axis that influences translational homeostasis of pro-fibrotic proteins and promotes pathological cardiac remodeling and fibrosis. EPRS is identified as a common node downstream of multiple cardiac stressors and a novel regulatory factor that facilitates pro-fibrotic mRNA translation in cardiac fibrosis. Global and myofibroblast-specific genetic ablation of EPRS can effectively reduce cardiac fibrosis. This study reveals a novel translational control mechanism that modulates cardiac fibrosis and heart function. Mild inhibition of PRR mRNA translation could be a general therapeutic strategy for the treatment of heart disease. These findings provide novel insights into the translational control mechanisms of cardiac fibrosis and will promote the development of novel therapeutics by inhibiting pro-fibrotic translation factors or their downstream effectors.


Circulation ◽  
2018 ◽  
Vol 138 (5) ◽  
pp. 513-526 ◽  
Author(s):  
Kevin J. Morine ◽  
Xiaoying Qiao ◽  
Sam York ◽  
Peter S. Natov ◽  
Vikram Paruchuri ◽  
...  

Background: Heart failure is a growing cause of morbidity and mortality worldwide. Transforming growth factor beta (TGF-β1) promotes cardiac fibrosis, but also activates counterregulatory pathways that serve to regulate TGF-β1 activity in heart failure. Bone morphogenetic protein 9 (BMP9) is a member of the TGFβ family of cytokines and signals via the downstream effector protein Smad1. Endoglin is a TGFβ coreceptor that promotes TGF-β1 signaling via Smad3 and binds BMP9 with high affinity. We hypothesized that BMP9 limits cardiac fibrosis by activating Smad1 and attenuating Smad3, and, furthermore, that neutralizing endoglin activity promotes BMP9 activity. Methods: We examined BMP9 expression and signaling in human cardiac fibroblasts and human subjects with heart failure. We used the transverse aortic constriction–induced model of heart failure to evaluate the functional effect of BMP9 signaling on cardiac remodeling. Results: BMP9 expression is increased in the circulation and left ventricle (LV) of human subjects with heart failure and is expressed by cardiac fibroblasts. Next, we observed that BMP9 attenuates type I collagen synthesis in human cardiac fibroblasts using recombinant human BMP9 and a small interfering RNA approach. In BMP9 –/– mice subjected to transverse aortic constriction, loss of BMP9 activity promotes cardiac fibrosis, impairs LV function, and increases LV levels of phosphorylated Smad3 (pSmad3), not pSmad1. In contrast, treatment of wild-type mice subjected to transverse aortic constriction with recombinant BMP9 limits progression of cardiac fibrosis, improves LV function, enhances myocardial capillary density, and increases LV levels of pSmad1, not pSmad3 in comparison with vehicle-treated controls. Because endoglin binds BMP9 with high affinity, we explored the effect of reduced endoglin activity on BMP9 activity. Neutralizing endoglin activity in human cardiac fibroblasts or in wild-type mice subjected to transverse aortic constriction–induced heart failure limits collagen production, increases BMP9 protein levels, and increases levels of pSmad1, not pSmad3. Conclusions: Our results identify a novel functional role for BMP9 as an endogenous inhibitor of cardiac fibrosis attributable to LV pressure overload and further show that treatment with either recombinant BMP9 or disruption of endoglin activity promotes BMP9 activity and limits cardiac fibrosis in heart failure, thereby providing potentially novel therapeutic approaches for patients with heart failure.


Sign in / Sign up

Export Citation Format

Share Document