Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Mol Cell Cardiol ; 165: 31-39, 2022 04.
Article in English | MEDLINE | ID: mdl-34968453

ABSTRACT

Pathological innate and adaptive immune response upon viral infection may lead to cardiac injury and dysfunction. Stabilin-1 is a scavenger receptor that regulates several aspects of the innate immunity. Whether stabilin-1 affects the inflammatory response during viral myocarditis (VM) is entirely unknown. Here, we assess the role of stabilin-1 in the pathogenesis of VM and its suitability as a therapeutic target. Genetic loss of stabilin-1 increased mortality and cardiac necrosis in a mouse model of human Coxsackievirus B3 (CVB3)-induced myocarditis. Absence of stabilin-1 significantly reduced monocyte recruitment and strongly reduced the number of alternatively activated anti-inflammatory macrophages in the heart, enhancing a pro-inflammatory cardiac niche with a detrimental T lymphocyte response during VM. Yeast two-hybrid screening, confirmed by affinity chromatography, identified fibronectin as a stabilin-1 interacting partner. Absence of stabilin-1 specifically decreased monocyte adhesion on extracellular fibronectin in vitro. Loss of Type III repeats Extra Domain A (EDA) of fibronectin during VM also increased the mortality and cardiac necrosis as in stabilin-1 knockout mice, with reduced monocytic cardiac recruitment and increased T lymphocyte response. Collectively, stabilin-1 has an immune-suppressive role of limiting myocardial damage during VM, regulating anti-inflammatory monocyte-recruitment to the site of inflammation.


Subject(s)
Coxsackievirus Infections , Myocarditis , Virus Diseases , Animals , Cell Adhesion Molecules, Neuronal , Disease Models, Animal , Enterovirus B, Human , Fibronectins , Macrophages , Mice , Monocytes/pathology , Necrosis
2.
J Cell Mol Med ; 25(2): 729-741, 2021 01.
Article in English | MEDLINE | ID: mdl-33295687

ABSTRACT

The metabolic syndrome (MetS) is an escalating problem worldwide, causing left ventricular stiffening, an early characteristic of diastolic dysfunction for which no treatment exists. As diastolic dysfunction and stiffening in MetS patients are associated with increased circulating dipeptidyl peptidase-4 (DPP-4) levels, we investigated whether the clinically approved DPP-4 inhibitor linagliptin reduces left ventricular stiffness in MetS-induced cardiac disease. Sixteen-week-old obese ZSF1 rats, displaying the MetS and left ventricular stiffness, received linagliptin-supplemented or placebo diet for four weeks. Linagliptin significantly reduced obesity, hyperlipidaemia, and hyperglycaemia and improved left ventricular relaxation. This improved relaxation was related to decreased cardiac fibrosis and cardiomyocyte passive stiffness (Fpassive ). The reduced Fpassive was the result of titin isoform switching from the stiff N2B to the more flexible N2BA and increased phosphorylation of total titin and specifically its N2Bus region (S4080 and S3391). Importantly, DPP-4 directly cleaved titin in vitro, resulting in an increased Fpassive , which was prevented by simultaneous administration of linagliptin. In conclusion, linagliptin improves left ventricular stiffness in obese ZSF1 rats by preventing direct DPP4-mediated titin cleavage, as well as by modulating both titin isoform levels and phosphorylation. Reducing left ventricular stiffness by administering linagliptin might prevent MetS-induced early diastolic dysfunction in human.


Subject(s)
Linagliptin/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Animals , Connectin/pharmacology , Heart Diseases/metabolism , Male , Mice, Obese , Myocardium/metabolism , Obesity/metabolism , Phosphorylation/drug effects , Protein Processing, Post-Translational , Rats
3.
Basic Res Cardiol ; 116(1): 10, 2021 02 09.
Article in English | MEDLINE | ID: mdl-33564961

ABSTRACT

We have previously demonstrated that systemic AMP-activated protein kinase α1 (AMPKα1) invalidation enhanced adverse LV remodelling by increasing fibroblast proliferation, while myodifferentiation and scar maturation were impaired. We thus hypothesised that fibroblastic AMPKα1 was a key signalling element in regulating fibrosis in the infarcted myocardium and an attractive target for therapeutic intervention. The present study investigates the effects of myofibroblast (MF)-specific deletion of AMPKα1 on left ventricular (LV) adaptation following myocardial infarction (MI), and the underlying molecular mechanisms. MF-restricted AMPKα1 conditional knockout (cKO) mice were subjected to permanent ligation of the left anterior descending coronary artery. cKO hearts exhibit exacerbated post-MI adverse LV remodelling and are characterised by exaggerated fibrotic response, compared to wild-type (WT) hearts. Cardiac fibroblast proliferation and MF content significantly increase in cKO infarcted hearts, coincident with a significant reduction of connexin 43 (Cx43) expression in MFs. Mechanistically, AMPKα1 influences Cx43 expression by both a transcriptional and a post-transcriptional mechanism involving miR-125b-5p. Collectively, our data demonstrate that MF-AMPKα1 functions as a master regulator of cardiac fibrosis and remodelling and might constitute a novel potential target for pharmacological anti-fibrotic applications.


Subject(s)
AMP-Activated Protein Kinases/deficiency , Connexin 43/metabolism , Myocardial Infarction/enzymology , Myocardium/enzymology , Myofibroblasts/enzymology , Ventricular Function, Left , Ventricular Remodeling , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Cell Proliferation , Connexin 43/genetics , Disease Models, Animal , Female , Fibrosis , Gene Deletion , HEK293 Cells , Humans , Male , Mice, Knockout , MicroRNAs/genetics , MicroRNAs/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/pathology , Myofibroblasts/pathology , Signal Transduction
4.
J Hum Nutr Diet ; 34(3): 616-628, 2021 06.
Article in English | MEDLINE | ID: mdl-33497494

ABSTRACT

BACKGROUND: The present study aimed to assess micronutrient intake among Greek adults and to identify the main food sources that contribute to it. METHODS: Food consumption data from 2389 participants in the Hellenic National Nutrition and Health Survey (HNNHS), collected with 24-h recalls, was used to calculate micronutrient intakes. Usual nutrient intake was estimated according to the National Cancer Institute method. Nutrient adequacy was estimated using the estimated average requirement (EAR) cut-point method, when available, or adequate intake otherwise. The probability approach was used to determine iron intake adequacy in females of reproductive age. Food group contribution for each nutrient assessed was derived to identify their main food sources. RESULTS: Almost all individuals had vitamin D intake below EAR, whereas vitamins A, E, K and C, as well as potassium intake, were also insufficient in a considerable percentage of the population (>70% in most age groups). Calcium intake was substantially below the EAR for females aged >50 years and males >70 years; the same for magnesium in males >70 years. Furthermore, 50% of females, including those of reproductive age, had intake of folate below EAR. More than 50% of the population (to 79%) exceeded the upper tolerable limit for sodium (2300 mg day-1 ). Food contribution analysis revealed that most vitamins were derived from low-quality foods (i.e. fast-food). CONCLUSIONS: A significant proportion of adults residing in Greece have low nutrient intake and poor food selections. These results provide guidance to public health policy makers for developing strategies to improve the dietary quality in Greece.


Subject(s)
Diet/standards , Food/classification , Micronutrients/administration & dosage , Nutritional Requirements , Nutritional Status , Adult , Aged , Diet/statistics & numerical data , Female , Food/statistics & numerical data , Greece/epidemiology , Humans , Male , Middle Aged , Nutrition Surveys , Recommended Dietary Allowances
5.
Eur Heart J ; 39(10): 876-887, 2018 03 07.
Article in English | MEDLINE | ID: mdl-29136142

ABSTRACT

Aims: Foxo3 is a transcription factor involved in cell metabolism, survival, and inflammatory disease. However, mechanistic insight in Foxo3 effects is still limited. Here, we investigated the role of Foxo3 on natural killer (NK) cell responses and its effects in viral myocarditis. Methods and results: Effects of Foxo3 on viral load and immune responses were investigated in a model of coxsackie virus B3 myocarditis in wild-type (WT) and Foxo3 deficient mice. Reduced immune cell infiltration, viral titres, and pro-inflammatory cytokines in cardiac tissue were observed in Foxo3-/- mice 7 days post-infection (p.i.). Viral titres were also attenuated in hearts of Foxo3-/- mice at Day 3 while interferon-γ (IFNγ) and NKp46 expression were up-regulated suggesting early viral control by enhanced NK cell activity. CD69 expression of NK cells, frequencies of CD11b+CD27+ effector NK cells and cytotoxicity of Foxo3-/- mice was enhanced compared to WT littermates. Moreover, microRNA-155 expression, essential in NK cell activation, was elevated in Foxo3-/- NK cells while its inhibition led to diminished IFNγ production. Healthy humans carrying the longevity-associated FOXO3 single nucleotide polymorphism (SNP) rs12212067 exhibited reduced IFNγ and cytotoxic degranulation of NK cells. Viral inflammatory cardiomyopathy (viral CMI) patients with this SNP showed a poorer outcome due to less efficient virus control. Conclusion: Our results implicate Foxo3 in regulating NK cell function and suggest Foxo3 playing an important role in the antiviral innate immunity. Thus, enhanced FOXO3 activity such as in the polymorphism rs12212067 may be protective in chronic inflammation such as cancer and cardiovascular disease but disadvantageous to control acute viral infection.


Subject(s)
Forkhead Box Protein O3 , Killer Cells, Natural/immunology , Myocarditis , Adult , Animals , Coxsackievirus Infections/immunology , Coxsackievirus Infections/virology , Cytokines/metabolism , Disease Models, Animal , Female , Forkhead Box Protein O3/genetics , Forkhead Box Protein O3/immunology , Forkhead Box Protein O3/metabolism , Heart/virology , Humans , Male , Mice , Mice, Knockout , Middle Aged , Myocarditis/immunology , Myocarditis/pathology , Myocarditis/virology , Myocardium/immunology , Myocardium/pathology , Polymorphism, Single Nucleotide
6.
Circulation ; 136(8): 747-761, 2017 Aug 22.
Article in English | MEDLINE | ID: mdl-28611091

ABSTRACT

BACKGROUND: Cardiovascular diseases remain the predominant cause of death worldwide, with the prevalence of heart failure continuing to increase. Despite increased knowledge of the metabolic alterations that occur in heart failure, novel therapies to treat the observed metabolic disturbances are still lacking. METHODS: Mice were subjected to pressure overload by means of angiotensin-II infusion or transversal aortic constriction. MicroRNA-146a was either genetically or pharmacologically knocked out or genetically overexpressed in cardiomyocytes. Furthermore, overexpression of dihydrolipoyl succinyltransferase (DLST) in the murine heart was performed by means of an adeno-associated virus. RESULTS: MicroRNA-146a was upregulated in whole heart tissue in multiple murine pressure overload models. Also, microRNA-146a levels were moderately increased in left ventricular biopsies of patients with aortic stenosis. Overexpression of microRNA-146a in cardiomyocytes provoked cardiac hypertrophy and left ventricular dysfunction in vivo, whereas genetic knockdown or pharmacological blockade of microRNA-146a blunted the hypertrophic response and attenuated cardiac dysfunction in vivo. Mechanistically, microRNA-146a reduced its target DLST-the E2 subcomponent of the α-ketoglutarate dehydrogenase complex, a rate-controlling tricarboxylic acid cycle enzyme. DLST protein levels significantly decreased on pressure overload in wild-type mice, paralleling a decreased oxidative metabolism, whereas DLST protein levels and hence oxidative metabolism were partially maintained in microRNA-146a knockout mice. Moreover, overexpression of DLST in wild-type mice protected against cardiac hypertrophy and dysfunction in vivo. CONCLUSIONS: Altogether we show that the microRNA-146a and its target DLST are important metabolic players in left ventricular dysfunction.


Subject(s)
Acyltransferases/biosynthesis , Cardiomegaly/metabolism , Gene Expression Regulation, Enzymologic , MicroRNAs/antagonists & inhibitors , MicroRNAs/biosynthesis , Ventricular Dysfunction, Left/metabolism , Acyltransferases/genetics , Animals , Animals, Newborn , Cardiomegaly/genetics , Cardiomegaly/prevention & control , Cells, Cultured , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Myocytes, Cardiac/metabolism , Rats , Rats, Inbred Lew , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/prevention & control
7.
Cell Mol Life Sci ; 74(8): 1511-1525, 2017 04.
Article in English | MEDLINE | ID: mdl-27878326

ABSTRACT

BACKGROUND: Viral myocarditis can severely damage the myocardium through excessive infiltration of immune cells. Osteoglycin (OGN) is part of the small leucine-rich repeat proteoglycan (SLRP) family. SLRP's may affect inflammatory and fibrotic processes, but the implication of OGN in cardiac inflammation and the resulting injury upon viral myocarditis is unknown. METHODS AND RESULTS: This study uncovered a previously unidentified 72-kDa variant of OGN that is predominant in cardiac human and mouse samples of viral myocarditis. Its absence in mice significantly decreased cardiac inflammation and injury in Coxsackievirus-B3-induced myocarditis. It also delayed mortality in lipopolysaccharide-induced endotoxemia going along with a reduced systemic production of pro-inflammatory cytokines. This 72-kDa OGN is expressed in the cell membrane of circulating and resident cardiac macrophages and neutrophils. Co-immunoprecipitation and OGN siRNA experiments revealed that this 72-kDa variant activates the toll-like receptor-4 (TLR4) with a concomitant increase in IL-6, TNF-α, IL-1ß, and IL-12 expression. This immune cell activation by OGN occurred via MyD88 and increased phosphorylation of c-jun. Finally, the 72-kDa chondroitin sulfate is the result of O-linked glycosylation of the 32-kDa protein core of OGN. In contrast, the 34-kDa dermatan sulfate-OGN, involved in collagen cross linking, was also the result of O-linked glycosylation. CONCLUSION: The current study discovered a novel 72-kDa chondroitin sulfate-OGN that is specific for innate immune cells. This variant is able to bind and activate TLR4. The absence of OGN decreases cytokine production by both circulating and cardiac leukocytes upon (systemic) LPS exposure, and reduces cardiac inflammation and injury in viral myocarditis.


Subject(s)
Intercellular Signaling Peptides and Proteins/immunology , Leukocytes/pathology , Myocarditis/immunology , Myocarditis/pathology , Myocardium/pathology , Toll-Like Receptor 4/immunology , Animals , Cytokines/immunology , Disease Models, Animal , Female , Glycosylation , HEK293 Cells , Heart/virology , Humans , Immunity, Cellular , Immunity, Innate , Intercellular Signaling Peptides and Proteins/analysis , Leukocytes/immunology , Leukocytes/virology , Male , Mice , Mice, Inbred C57BL , Myocarditis/virology , Myocardium/immunology
8.
Angew Chem Int Ed Engl ; 57(2): 486-490, 2018 01 08.
Article in English | MEDLINE | ID: mdl-28980372

ABSTRACT

Tudor domains bind to dimethylarginine (DMA) residues, which are post-translational modifications that play a central role in gene regulation in eukaryotic cells. NMR spectroscopy and quantum calculations are combined to demonstrate that DMA recognition by Tudor domains involves conformational selection. The binding mechanism is confirmed by a mutation in the aromatic cage that perturbs the native recognition mode of the ligand. General mechanistic principles are delineated from the combined results, indicating that Tudor domains utilize cation-π interactions to achieve ligand recognition.


Subject(s)
Arginine/analogs & derivatives , Motor Neurons/metabolism , Tudor Domain , Arginine/chemistry , Arginine/metabolism , Protein Conformation , Protein Processing, Post-Translational , Quantum Theory , Thermodynamics
9.
Basic Res Cardiol ; 112(4): 42, 2017 07.
Article in English | MEDLINE | ID: mdl-28540528

ABSTRACT

Optimal healing after myocardial infarction requires not only the induction of inflammation, but also its timely resolution. In patients, 30 days post myocardial infarction, circulating monocytes have increased expression of Semaphorin3A (Sema3A) as compared to directly after admission. This increased expression coincides with increased expression of Cx3CR1-a marker of non-classical monocytes that are important for immune resolution hence proper wound healing. In mice, the expression of Sema3A also increases in response to myocardial ischemia being expressed by infiltrating leukocytes. Comparing Sema3A heterozygote (HZ) and wild type (WT) mice post myocardial infarction, revealed increased presence of leukocytes in the cardiac tissues of HZ mice as compared to WT, with no differences in capillary density, collagen deposition, cardiomyocyte surface area, chemokine-or adhesion molecules expression. Whilst infarct sizes were similar 14 days after myocardial infarction in both genotypes, Sema3A HZ mice had thinner infarcts and reduced cardiac function as compared to their WT littermates. In vitro experiments were conducted to study the role of Sema3A in inflammation and resolution of inflammation as a potential explanation for the differences in leukocyte recruitment and cardiac function observed in our in vivo experiments. Here, recombinant Sema3A protein was able to affect the pro-inflammatory state of cultured bone marrow derived macrophages. First, the pro-inflammatory state was altered by the induced apoptosis of classical macrophages in the presence of Sema3A. Second, Sema3A promoted the polarization of classical macrophages to resolution-phase macrophages and enhanced their efferocytotic ability, findings that were reflected in the infarcted cardiac tissue of the Sema3A HZ mice. Finally, we demonstrated that besides promoting resolution of inflammation, Sema3A was also able to retard the migration of monocytes to the myocardium. Collectively our data demonstrate that Sema3A reduces cardiac inflammation and improves cardiac function after myocardial infarction by promoting the resolution of inflammation.


Subject(s)
Myocardial Infarction/metabolism , Myocarditis/metabolism , Myocardium/metabolism , Semaphorin-3A/metabolism , Wound Healing , Animals , Apoptosis , Cells, Cultured , Chemotaxis, Leukocyte , Disease Models, Animal , Female , Heterozygote , Macrophage Activation , Macrophages/metabolism , Macrophages/pathology , Male , Mice, Knockout , Monocytes/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocarditis/genetics , Myocarditis/pathology , Myocarditis/physiopathology , Myocardium/pathology , Phenotype , Recovery of Function , Semaphorin-3A/deficiency , Semaphorin-3A/genetics , Signal Transduction , Time Factors
10.
FASEB J ; 30(8): 2651-61, 2016 08.
Article in English | MEDLINE | ID: mdl-27080639

ABSTRACT

Small leucine-rich proteoglycans are emerging as important regulatory proteins within the extracellular matrix, where they exert both structural and nonstructural functions and hence are modulators of numerous biological processes, such as inflammation, fibrosis, and cell proliferation. One proteoglycan in particular, osteoglycin (OGN), also known as mimecan, shows great structural and functional diversity in normal physiology and in disease states, therefore making it a very interesting candidate for the development of novel therapeutic strategies. Unfortunately, the literature on OGN is confusing, as it has different names, and different transcript and protein variants have been identified. This review will give a clear overview of the different structures and functions of OGN that have been identified to date, portray its central role in pathophysiology, and highlight the importance of posttranslational processing, such as glycosylation, for the diversity of its functions.-Deckx, S., Heymans, S., Papageorgiou, A.-P. The diverse functions of osteoglycin: a deceitful dwarf, or a master regulator of disease?


Subject(s)
Extracellular Matrix/metabolism , Gene Expression Regulation/physiology , Intercellular Signaling Peptides and Proteins/metabolism , Humans , Intercellular Signaling Peptides and Proteins/genetics , Protein Conformation
11.
Circ Res ; 116(3): 425-36, 2015 Jan 30.
Article in English | MEDLINE | ID: mdl-25520363

ABSTRACT

RATIONALE: To maintain cardiac mechanical and structural integrity after an ischemic insult, profound alterations occur within the extracellular matrix. Osteoglycin is a small leucine-rich proteoglycan previously described as a marker of cardiac hypertrophy. OBJECTIVE: To establish whether osteoglycin may play a role in cardiac integrity and function after myocardial infarction (MI). METHODS AND RESULTS: Osteoglycin expression is associated with collagen deposition and scar formation in mouse and human MI. Absence of osteoglycin in mice resulted in significantly increased rupture-related mortality with tissue disruption, intramyocardial bleeding, and increased cardiac dysfunction, despite equal infarct sizes. Surviving osteoglycin null mice had greater infarct expansion in comparison with wild-type mice because of impaired collagen fibrillogenesis and maturation in the infarcts as revealed by electron microscopy and collagen polarization. Absence of osteoglycin did not affect cardiomyocyte hypertrophy in the remodeling remote myocardium. In cultured fibroblasts, osteoglycin knockdown or supplementation did not alter transforming growth factor-ß signaling. Adenoviral overexpression of osteoglycin in wild-type mice significantly improved collagen quality, thereby blunting cardiac dilatation and dysfunction after MI. In osteoglycin null mice, adenoviral overexpression of osteoglycin was unable to prevent rupture-related mortality because of insufficiently restoring osteoglycin protein levels in the heart. Finally, circulating osteoglycin levels in patients with heart failure were significantly increased in the patients with a previous history of MI compared with those with nonischemic heart failure and correlated with survival, left ventricular volumes, and other markers of fibrosis. CONCLUSIONS: Increased osteoglycin expression in the infarct scar promotes proper collagen maturation and protects against cardiac disruption and adverse remodeling after MI. In human heart failure, osteoglycin is a promising biomarker for ischemic heart failure.


Subject(s)
Cardiomegaly/metabolism , Collagen/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Myocardial Infarction/metabolism , Animals , Cardiomegaly/pathology , Cicatrix/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/physiology , Humans , Intercellular Signaling Peptides and Proteins/blood , Intercellular Signaling Peptides and Proteins/genetics , Lymphotoxin-alpha/metabolism , Mice , Mice, Inbred C57BL , Myocardial Infarction/pathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Rats , Rats, Inbred Lew , Ventricular Remodeling
12.
J Mol Cell Cardiol ; 91: 172-8, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26791544

ABSTRACT

More than 20years ago, Paul Bornstein coined the term matricellular protein to describe a group of secreted extracellular matrix proteins with de-adhesive properties. Though this is still true today, this family of proteins is vastly expanding with new emerging functions pushing the boundaries of this classic definition. In the heart, matricellular proteins have been extensively investigated in models of myocardial infarction, pressure overload, viral myocarditis and age-related cardiomyopathy with clear implications during cardiac fibrosis yet their involvement in regulating cardiac inflammation is less established. In this review, we describe our current understanding of the immune activation by damage- or pathogen-associated molecular pattern molecules during cardiac injury making a distinction between sterile versus non-sterile cardiac inflammation, and explain how matricellular proteins influence this crucial pathophysiological response in the heart.


Subject(s)
CCN Intercellular Signaling Proteins/genetics , Gene Expression Regulation , Myocardial Infarction/genetics , Myocarditis/genetics , Myocardium/metabolism , Animals , CCN Intercellular Signaling Proteins/metabolism , Fibrosis , Galectins/genetics , Galectins/metabolism , Inflammation , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocarditis/metabolism , Myocarditis/pathology , Myocardium/pathology , Osteonectin/genetics , Osteonectin/metabolism , Osteopontin/genetics , Osteopontin/metabolism , Signal Transduction , Tenascin/genetics , Tenascin/metabolism , Thrombospondins/genetics , Thrombospondins/metabolism
13.
Circ Res ; 114(5): 872-88, 2014 Feb 28.
Article in English | MEDLINE | ID: mdl-24577967

ABSTRACT

The cardiac extracellular matrix (ECM) is a complex architectural network consisting of structural and nonstructural proteins, creating strength and plasticity. The nonstructural compartment of the ECM houses a variety of proteins, which are vital for ECM plasticity, and can be divided into 3 major groups: glycoproteins, proteoglycans, and glycosaminoglycans. The common denominator for these groups is glycosylation, which refers to the decoration of proteins or lipids with sugars. This review will discuss the fundamental role of the matrix in cardiac development, homeostasis, and remodeling, from a glycobiology point of view. Glycoproteins (eg, thrombospondins, secreted protein acidic and rich in cysteine, tenascins), proteoglycans (eg, versican, syndecans, biglycan), and glycosaminoglycans (eg, hyaluronan, heparan sulfate) are upregulated on cardiac injury and regulate key processes in the remodeling myocardium such as inflammation, fibrosis, and angiogenesis. Albeit some parallels can be made regarding the processes these proteins are involved in, their specific functions are extremely diverse. In fact, under varying conditions, individual proteins can even have opposing functions, making spatiotemporal contribution of these proteins in the rearrangement of multifaceted ECM very hard to grasp. Alterations of protein characteristics by the addition of sugars may explain the immense, yet tightly regulated, variability of the remodeling cardiac matrix. Understanding the role of glycosylation in altering the ultimate function of glycoproteins, proteoglycans, and glycosaminoglycans in the myocardium may lead to the development of new biochemical structures or compounds with great therapeutic potential for patients with heart disease.


Subject(s)
Extracellular Matrix Proteins/metabolism , Extracellular Matrix/metabolism , Glycoproteins/metabolism , Homeostasis/physiology , Myocardium/metabolism , Animals , Glycosylation , Humans
14.
Extremophiles ; 20(5): 695-709, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27342116

ABSTRACT

The histone-like DNA-binding proteins (HU) serve as model molecules for protein thermostability studies, as they function in different bacteria that grow in a wide range of temperatures and show sequence diversity under a common fold. In this work, we report the cloning of the hutth gene from Thermus thermophilus, the purification and crystallization of the recombinant HUTth protein, as well as its X-ray structure determination at 1.7 Å. Detailed structural and thermodynamic analyses were performed towards the understanding of the thermostability mechanism. The interaction of HUTth protein with plasmid DNA in solution has been determined for the first time with MST. Sequence conservation of an exclusively thermophilic order like Thermales, when compared to a predominantly mesophilic order (Deinococcales), should be subject, to some extent, to thermostability-related evolutionary pressure. This hypothesis was used to guide our bioinformatics and evolutionary studies. We discuss the impact of thermostability adaptation on the structure of HU proteins, based on the detailed evolutionary analysis of the Deinococcus-Thermus phylum, where HUTth belongs. Furthermore, we propose a novel method of engineering thermostable proteins, by combining consensus-based design with ancestral sequence reconstruction. Finally, through the structure of HUTth, we are able to examine the validity of these predictions. Our approach represents a significant advancement, as it explores for the first time the potential of ancestral sequence reconstruction in the divergence between a thermophilic and a mainly mesophilic taxon, combined with consensus-based engineering.


Subject(s)
Bacterial Proteins/metabolism , DNA-Binding Proteins/metabolism , Evolution, Molecular , Hot Temperature , Thermus thermophilus/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Conserved Sequence , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Protein Binding , Protein Stability , Thermus thermophilus/metabolism
15.
Eur Heart J ; 36(42): 2909-19, 2015 11 07.
Article in English | MEDLINE | ID: mdl-26206211

ABSTRACT

AIMS: Viral myocarditis (VM) is an important cause of heart failure and sudden cardiac death in young healthy adults; it is also an aetiological precursor of dilated cardiomyopathy. We explored the role of the miR-221/-222 family that is up-regulated in VM. METHODS AND RESULTS: Here, we show that microRNA-221 (miR-221) and miR-222 levels are significantly elevated during acute VM caused by Coxsackievirus B3 (CVB3). Both miRs are expressed by different cardiac cells and by infiltrating inflammatory cells, but their up-regulation upon myocarditis is mostly exclusive for the cardiomyocyte. Systemic inhibition of miR-221/-222 in mice increased cardiac viral load, prolonged the viraemic state, and strongly aggravated cardiac injury and inflammation. Similarly, in vitro, overexpression of miR-221 and miR-222 inhibited enteroviral replication, whereas knockdown of this miR-cluster augmented viral replication. We identified and confirmed a number of miR-221/-222 targets that co-orchestrate the increased viral replication and inflammation, including ETS1/2, IRF2, BCL2L11, TOX, BMF, and CXCL12. In vitro inhibition of IRF2, TOX, or CXCL12 in cardiomyocytes significantly dampened their inflammatory response to CVB3 infection, confirming the functionality of these targets in VM and highlighting the importance of miR-221/-222 as regulators of the cardiac response to VM. CONCLUSIONS: The miR-221/-222 cluster orchestrates the antiviral and inflammatory immune response to viral infection of the heart. Its inhibition increases viral load, inflammation, and overall cardiac injury upon VM.


Subject(s)
Coxsackievirus Infections/virology , MicroRNAs/physiology , Myocarditis/virology , Animals , Coxsackievirus Infections/immunology , Humans , Immunity, Cellular/immunology , Macrophages/immunology , Male , Mice, Inbred C3H , Mice, Inbred C57BL , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Myocarditis/immunology , Myocytes, Cardiac/immunology , T-Lymphocytes/immunology , Up-Regulation , Viral Load/immunology , Virus Replication/immunology
16.
Circulation ; 128(13): 1420-32, 2013 Sep 24.
Article in English | MEDLINE | ID: mdl-23956210

ABSTRACT

BACKGROUND: Cardiac hypertrophy and subsequent heart failure triggered by chronic hypertension represent major challenges for cardiovascular research. Beyond neurohormonal and myocyte signaling pathways, growing evidence suggests inflammatory signaling pathways as therapeutically targetable contributors to this process. We recently reported that microRNA-155 is a key mediator of cardiac inflammation and injury in infectious myocarditis. Here, we investigated the impact of microRNA-155 manipulation in hypertensive heart disease. METHODS AND RESULTS: Genetic loss or pharmacological inhibition of the leukocyte-expressed microRNA-155 in mice markedly reduced cardiac inflammation, hypertrophy, and dysfunction on pressure overload. These alterations were macrophage dependent because in vivo cardiomyocyte-specific microRNA-155 manipulation did not affect cardiac hypertrophy or dysfunction, whereas bone marrow transplantation from wild-type mice into microRNA-155 knockout animals rescued the hypertrophic response of the cardiomyocytes and vice versa. In vitro, media from microRNA-155 knockout macrophages blocked the hypertrophic growth of stimulated cardiomyocytes, confirming that macrophages influence myocyte growth in a microRNA-155-dependent paracrine manner. These effects were at least partly mediated by the direct microRNA-155 target suppressor of cytokine signaling 1 (Socs1) because Socs1 knockdown in microRNA-155 knockout macrophages largely restored their hypertrophy-stimulating potency. CONCLUSIONS: Our findings reveal that microRNA-155 expression in macrophages promotes cardiac inflammation, hypertrophy, and failure in response to pressure overload. These data support the causative significance of inflammatory signaling in hypertrophic heart disease and demonstrate the feasibility of therapeutic microRNA targeting of inflammation in heart failure.


Subject(s)
Cardiomegaly/pathology , Heart Failure/pathology , Macrophages/pathology , MicroRNAs/genetics , Myocytes, Cardiac/pathology , Animals , Cardiomegaly/genetics , Cells, Cultured , Heart Failure/genetics , Humans , Inflammation/genetics , Inflammation/pathology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/metabolism , Rats
17.
Circ Res ; 111(4): 415-25, 2012 Aug 03.
Article in English | MEDLINE | ID: mdl-22715471

ABSTRACT

RATIONALE: Viral myocarditis results from an adverse immune response to cardiotropic viruses, which causes irreversible myocyte destruction and heart failure in previously healthy people. The involvement of microRNAs and their usefulness as therapeutic targets in this process are unknown. OBJECTIVE: To identify microRNAs involved in viral myocarditis pathogenesis and susceptibility. METHODS AND RESULTS: Cardiac microRNAs were profiled in both human myocarditis and in Coxsackievirus B3-injected mice, comparing myocarditis-susceptible with nonsusceptible mouse strains longitudinally. MicroRNA responses diverged depending on the susceptibility to myocarditis after viral infection in mice. MicroRNA-155, -146b, and -21 were consistently and strongly upregulated during acute myocarditis in both humans and susceptible mice. We found that microRNA-155 expression during myocarditis was localized primarily in infiltrating macrophages and T lymphocytes. Inhibition of microRNA-155 by a systemically delivered LNA-anti-miR attenuated cardiac infiltration by monocyte-macrophages, decreased T lymphocyte activation, and reduced myocardial damage during acute myocarditis in mice. These changes were accompanied by the derepression of the direct microRNA-155 target PU.1 in cardiac inflammatory cells. Beyond the acute phase, microRNA-155 inhibition reduced mortality and improved cardiac function during 7 weeks of follow-up. CONCLUSIONS: Our data show that cardiac microRNA dysregulation is a characteristic of both human and mouse viral myocarditis. The inflammatory microRNA-155 is upregulated during acute myocarditis, contributes to the adverse inflammatory response to viral infection of the heart, and is a potential therapeutic target for viral myocarditis.


Subject(s)
Coxsackievirus Infections/genetics , Gene Expression Profiling , MicroRNAs/metabolism , Myocarditis/genetics , Myocardium/metabolism , Animals , Coxsackievirus Infections/immunology , Coxsackievirus Infections/pathology , Coxsackievirus Infections/physiopathology , Coxsackievirus Infections/therapy , Coxsackievirus Infections/virology , Disease Models, Animal , Enterovirus B, Human/pathogenicity , Female , Gene Expression Profiling/methods , Humans , Lymphocyte Activation , Macrophages/immunology , Macrophages/metabolism , Macrophages/virology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Myocarditis/immunology , Myocarditis/pathology , Myocarditis/physiopathology , Myocarditis/therapy , Myocarditis/virology , Myocardium/immunology , Myocardium/pathology , Oligonucleotides/administration & dosage , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/virology , Time Factors
18.
Eur Heart J ; 34(25): 1930-41, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23139380

ABSTRACT

AIMS: The cardiac extracellular matrix is highly involved in regulating inflammation, remodelling, and function of the heart. Whether matrix alterations relate to the degree of inflammation, fibrosis, and overall rejection in the human transplanted heart remained, until now, unknown. METHODS AND RESULTS: Expression of matricellular proteins, proteoglycans, and metalloproteinases (MMPs) and their inhibitors (TIMPs) were investigated in serial endomyocardial biopsies (n = 102), in a cohort of 39 patients within the first year after cardiac transplantation. Out of 15 matrix-related proteins, intragraft transcript and protein levels of syndecan-1 and MMP-9 showed a strong association with the degree of cardiac allograft rejection (CAR), the expression of pro-inflammatory cytokines tumour necrosis factor (TNF)-α, interleukin (IL)-6 and transforming growth factor (TGF)-ß, and with infiltrating CD3⁺ T-cells and CD68⁺ monocytes. In addition, SPARC, CTGF, TSP-2, MMP-14, TIMP-1, Testican-1, TSP-1, Syndecan-1, MMP-2, -9, and -14, as well as IL-6 and TGF-ß transcript levels and inflammatory infiltrates all strongly relate to collagen expression in the transplanted heart. More importantly, receiver operating characteristic curve analysis demonstrated that syndecan-1 and MMP-9 transcript levels had the highest area under the curve (0.969 and 0.981, respectively), thereby identifying both as a potential decision-making tool to discriminate rejecting from non-rejecting hearts. CONCLUSION: Out of 15 matrix-related proteins, we identified synd-1 and MMP-9 intragraft transcript levels of as strong predictors of human CAR. In addition, a multitude of non-structural matrix-related proteins closely associate with collagen expression in the transplanted heart. Therefore, we are convinced that these findings deserve further investigation and are likely to be of clinical value to prevent human CAR.


Subject(s)
Extracellular Matrix/metabolism , Graft Rejection/pathology , Heart Transplantation , Matrix Metalloproteinases/metabolism , Myocardium/pathology , Allografts , Biomarkers/metabolism , Cytokines/metabolism , Female , Fibrosis/metabolism , Fibrosis/pathology , Graft Rejection/metabolism , Humans , Male , Middle Aged , Monocytes/pathology , Myocarditis/metabolism , Myocarditis/pathology , Proteoglycans/metabolism , T-Lymphocytes/pathology , Tissue Inhibitor of Metalloproteinases/metabolism
19.
Nat Commun ; 14(1): 6751, 2023 10 24.
Article in English | MEDLINE | ID: mdl-37875529

ABSTRACT

Biomolecular polyelectrolyte complexes can be formed between oppositely charged intrinsically disordered regions (IDRs) of proteins or between IDRs and nucleic acids. Highly charged IDRs are abundant in the nucleus, yet few have been functionally characterized. Here, we show that a positively charged IDR within the human ATP-dependent DNA helicase Q4 (RECQ4) forms coacervates with G-quadruplexes (G4s). We describe a three-step model of charge-driven coacervation by integrating equilibrium and kinetic binding data in a global numerical model. The oppositely charged IDR and G4 molecules form a complex in the solution that follows a rapid nucleation-growth mechanism leading to a dynamic equilibrium between dilute and condensed phases. We also discover a physical interaction with Replication Protein A (RPA) and demonstrate that the IDR can switch between the two extremes of the structural continuum of complexes. The structural, kinetic, and thermodynamic profile of its interactions revealed a dynamic disordered complex with nucleic acids and a static ordered complex with RPA protein. The two mutually exclusive binding modes suggest a regulatory role for the IDR in RECQ4 function by enabling molecular handoffs. Our study extends the functional repertoire of IDRs and demonstrates a role of polyelectrolyte complexes involved in G4 binding.


Subject(s)
G-Quadruplexes , Intrinsically Disordered Proteins , RecQ Helicases , Humans , Intrinsically Disordered Proteins/metabolism , Nucleic Acids , Polyelectrolytes , RecQ Helicases/metabolism
20.
Children (Basel) ; 9(1)2022 Jan 08.
Article in English | MEDLINE | ID: mdl-35053711

ABSTRACT

Two lifestyle intervention programs of a health initiative named "Evrostia" were conducted at (a) an outpatient obesity clinic of a children's hospital and (b) summer camp (SC), respectively. Thirty overweight/obese children were randomly selected to participate in each intervention arm to assess the efficacy of the SC intervention and its possible superiority over usual hospital consultation (HC) practice. There was a statistically significant decrease in body weight (BW), and body mass index (BMI) in both programs. A higher duration of reduced BW was observed in the SC compared to HC intervention. Regarding the nutritional behavior, there was a significant increase in the consumption of breakfast, fruit and vegetables, and a reduction in the consumption of beverages and sweets in the SC group. A significant increase in the hours of weekly physical activity was also observed in children of the SC program. The comparison between the two lifestyle intervention programs showed that the SC program improved nutritional behaviors and physical activity and promoted longer preservation of BW loss than that of the HC program. Thus, the holistic and experiential approach of the SC program was more successful in the treatment of overweight and obesity in children than a conventional HC program.

SELECTION OF CITATIONS
SEARCH DETAIL