Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Eur J Clin Invest ; 51(7): e13539, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33729579

ABSTRACT

BACKGROUND: Gremlin-1 is a cystine knot protein and is expressed in organs developing fibrosis. Transient ischaemia leads to myocardial fibrosis, a major determinant of impaired myocardial function. MATERIALS AND METHODS: Expression of Gremlin-1 was investigated in infarcted myocardium by real-time PCR, Western blot analysis, histological and immunohistochemistry staining. We further elaborated the colocalization of Gremlin-1 and TGF-ß proteins by confocal microscopy and co-immunoprecipitation experiments. The interaction between Gremlin-1 and TGF-ß was analysed by photon correlation spectroscopy. Gremlin-1 modulation of the TGF-ß-dependent collagen I synthesis in fibroblasts was investigated using ELISA and immunohistochemistry experiments. The effect of prolonged administration of recombinant Gremlin-1 on myocardial function following ischaemia/reperfusion was accessed by echocardiography and immunohistochemistry. RESULTS: Gremlin-1 is expressed in myocardial tissue and infiltrating cells after transient myocardial ischaemia (P < .05). Gremlin-1 colocalizes with the pro-fibrotic cytokine transforming growth factor-ß (TGF-ß) expressed in fibroblasts and inflammatory cell infiltrates (P < .05). Gremlin-1 reduces TGF-ß-induced collagen production of myocardial fibroblasts by approximately 20% (P < .05). We found that Gremlin-1 binds with high affinity to TGF-ß (KD  = 54 nmol/L) as evidenced by photon correlation spectroscopy and co-immunoprecipitation. intravenous administration of m Gremlin-1-Fc, but not of equivalent amount of Fc control, significantly reduced infarct size by approximately 20%. In the m Gremlin-1-Fc group, infarct area was reduced by up to 30% in comparison with mice treated with Fc control (I/LV: 4.8 ± 1.2% vs 6.0 ± 1.2% P < .05; I/AaR: 15.2 ± 1.5% vs 21.1 ± 5%, P < .05). CONCLUSIONS: The present data disclose Gremlin-1 as an antagonist of TGF-ß and presume a role for Gremlin-1/TGF-ß interaction in myocardial remodelling following myocardial ischaemia.


Subject(s)
Fibroblasts/metabolism , Heart/physiopathology , Intercellular Signaling Peptides and Proteins/genetics , Myocardial Infarction/genetics , Myocardial Reperfusion Injury/genetics , Myocardium/pathology , Transforming Growth Factor beta/metabolism , Animals , Collagen Type I/metabolism , Echocardiography , Endothelial Cells/metabolism , Fibroblasts/drug effects , Fibrosis , Heart/diagnostic imaging , Heart/drug effects , Humans , Immunoprecipitation , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Mice , Microscopy, Confocal , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Recombinant Proteins , Transforming Growth Factor beta/drug effects , Ventricular Remodeling/genetics
2.
Clin Cardiol ; 39(4): 201-6, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27101443

ABSTRACT

BACKGROUND: Chronic inflammation promotes atherosclerosis and is a prognostic factor in coronary artery disease (CAD). Patients with type 2 diabetes mellitus (DM2) are at risk for progressive atherosclerosis. Macrophage migration inhibitory factor (MIF) is a key player in atherosclerosis, mediating pro-inflammatory responses. Its endogenous antagonist Gremlin-1 inhibits foam-cell formation and atheroprogression by binding MIF, neutralizing its proatherosclerotic functions. HYPOTHESIS: Plasma levels of MIF and Gremlin-1 correlate with the stability of CAD in patients with DM2. METHOD: We assessed plasma levels of Gremlin-1 and MIF in 198 nondiabetic and 88 diabetic patients with symptomatic CAD using enzyme-linked immunosorbent assays. RESULTS: Plasma levels of Gremlin-1 were higher DM2 patients (278.8 ± 16.6 vs 224.7 ± 6.7 ng/mL; P = 0.001). MIF levels were elevated but not significantly increased in DM2 (P = 0.098). Interestingly, we found that Gremlin-1 plasma levels were significantly higher in diabetic patients with stable angina pectoris (SAP; n = 53) or acute coronary syndrome (ACS; n = 35) compared with nondiabetic patients with SAP (P = 0.008 and P = 0.011, respectively). MIF levels were significantly higher in diabetic patients with ACS compared with SAP (P < 0.001). Although the single plasma parameters showed an association with DM2 and CAD status, we could not confirm that the Gremlin-1/MIF ratio is significantly different in patients stratified by DM2 and CAD (P = 0.072). Hence, Gremlin-1/MIF ratio was significantly lower in patients with ACS compared with SAP (1.1 ± 0.1 vs 4.4 ± 1.1; P = 0.003). CONCLUSIONS: Diabetic patients with ACS show increased levels of Gremlin-1 and MIF, leading to unfavorable Gremlin-1/MIF ratios. However, DM2 alone is not associated with low Gremlin-1/MIF ratios.


Subject(s)
Acute Coronary Syndrome/etiology , Diabetes Mellitus, Type 2/blood , Intercellular Signaling Peptides and Proteins/blood , Intramolecular Oxidoreductases/blood , Macrophage Migration-Inhibitory Factors/blood , Acute Coronary Syndrome/diagnosis , Aged , Aged, 80 and over , Biomarkers/blood , Case-Control Studies , Cross-Sectional Studies , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/diagnosis , Female , Germany , Humans , Male , Middle Aged , Pilot Projects , Predictive Value of Tests , Prognosis , Registries , Risk Assessment , Risk Factors , Up-Regulation
4.
Arterioscler Thromb Vasc Biol ; 35(3): 547-57, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25614279

ABSTRACT

OBJECTIVE: Atherosclerosis, an inflammatory disease of arterial vessel walls, requires migration and matrix metalloproteinase (MMP)-9-dependent invasion of monocytes/macrophages into the vascular wall. MMP-9 expression is stimulated by transcription factor nuclear factor-κB, which is regulated by inhibitor κB (IκB) and thus IκB kinase. Regulators of nuclear factor-κB include serum- and glucocorticoid-inducible kinase 1 (SGK1). The present study explored involvement of SGK1 in vascular inflammation and atherogenesis. APPROACH AND RESULTS: Gene-targeted apolipoprotein E (ApoE)-deficient mice without (apoe(-/-)sgk1(+/+)) or with (apoe(-/-)sgk1(-/-)) additional SGK1 knockout received 16-week cholesterol-rich diet. According to immunohistochemistry atherosclerotic lesions in aorta and carotid artery, vascular CD45(+) leukocyte infiltration, Mac-3(+) macrophage infiltration, vascular smooth muscle cell content, MMP-2, and MMP-9 positive areas in atherosclerotic tissue were significantly less in apoe(-/-)sgk1(-/-)mice than in apoe(-/-)sgk1(+/+)mice. As determined by Boyden chamber, thioglycollate-induced peritonitis and air pouch model, migration of SGK1-deficient CD11b(+)F4/80(+) macrophages was significantly diminished in vitro and in vivo. Zymographic MMP-2 and MMP-9 production, MMP-9 activity and invasion through matrigel in vitro were significantly less in sgk1(-/-) than in sgk1(+/+)macrophages and in control plasmid-transfected or inactive (K127N)SGK1-transfected than in constitutively active (S422D)SGK1-transfected THP-1 cells. Confocal microscopy revealed reduced macrophage number and macrophage MMP-9 content in plaques of apoe(-/-)sgk1(-/-) mice. In THP-1 cells, MMP-inhibitor GM6001 (25 µmol/L) abrogated (S422D)SGK1-induced MMP-9 production and invasion. According to reverse transcription polymerase chain reaction, MMP-9 transcript levels were significantly reduced in sgk1(-/-)macrophages and strongly upregulated in (S422D)SGK1-transfected THP-1 cells compared with control plasmid-transfected or (K127N)SGK1-transfected THP-1 cells. According to immunoblotting and confocal microscopy, phosphorylation of IκB kinase and inhibitor κB and nuclear translocation of p50 were significantly lower in sgk1(-/-)macrophages than in sgk1(+/+)macrophages and significantly higher in (S422D)SGK1-transfected THP-1 cells than in control plasmid-transfected or (K127N)SGK1-transfected THP-1 cells. Treatment of (S422D)SGK1-transfected THP-1 cells with IκB kinase-inhibitor BMS-345541 (10 µmol/L) abolished (S422D)SGK1-induced increase of MMP-9 transcription and gelatinase activity. CONCLUSIONS: SGK1 plays a pivotal role in vascular inflammation during atherogenesis. SGK1 participates in the regulation of monocyte/macrophage migration and MMP-9 transcription via regulation of nuclear factor-κB.


Subject(s)
Aortic Diseases/enzymology , Atherosclerosis/enzymology , Carotid Artery Diseases/enzymology , Chemotaxis , Immediate-Early Proteins/metabolism , Inflammation/enzymology , Protein Serine-Threonine Kinases/metabolism , Active Transport, Cell Nucleus , Animals , Aorta/enzymology , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/pathology , Carotid Arteries/enzymology , Carotid Arteries/pathology , Carotid Artery Diseases/genetics , Carotid Artery Diseases/pathology , Cell Line , Disease Models, Animal , Gene Expression Regulation, Enzymologic , Humans , I-kappa B Kinase/metabolism , I-kappa B Proteins/metabolism , Immediate-Early Proteins/deficiency , Immediate-Early Proteins/genetics , Inflammation/genetics , Inflammation/pathology , Macrophages/enzymology , Macrophages/pathology , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mutation , NF-kappa B p50 Subunit/metabolism , Peritonitis/chemically induced , Peritonitis/enzymology , Peritonitis/genetics , Plaque, Atherosclerotic , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Thioglycolates , Transcription, Genetic , Transfection , Vascular Remodeling
5.
Atherosclerosis ; 237(2): 426-32, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25463068

ABSTRACT

OBJECTIVE: Monocyte infiltration is a critical step in the pathophysiology of plaque instability in coronary artery disease (CAD). Macrophage migration inhibitory factor (MIF) is involved in atherosclerotic plaque progression and instability leading to intracoronary thrombosis. Gremlin-1 (Grem1) has been recently identified as endogenous inhibitor of MIF. To date there are no data on the clinical impact of this interaction in cardiovascular patients. METHODS AND RESULTS: Plasma levels of MIF and Grem1 were determined by enzyme-linked immunoassay in patients with acute coronary syndromes (ACS, n = 120; stable CAD, n = 166 and healthy control subjects, n = 25). MIF levels were significantly increased in ACS compared to stable CAD and healthy control (ACS: median 2.85; IQR 3.52 ng/ml; versus SAP: median 1.22; IQR 2.99 ng/ml; versus healthy control: median 0.10; IQR 0.09 ng/ml, p < 0.001). Grem1 levels were significantly higher in ACS and stable CAD patients compared to healthy control (ACS: median 211.00; IQR 130.47 ng/ml; SAP: median 220.20; IQR 120.93 ng/ml, versus healthy control: median 90.57; IQR 97.68 ng/ml, p < 0.001). Grem1/MIF ratio was independently associated with ACS, whereas the single parameters were not associated with the presence of ACS. Furthermore, Grem1/MIF ratio was associated with angiographic signs of intracoronary thrombi and severity of thrombus burden. CONCLUSION: These novel findings suggest a potential role of Grem1/MIF ratio to indicate acuity of CAD and the grade of plaque stability. Prospective angiographic cohort studies involving plaque imaging techniques are warranted to further characterize the prognostic role of this novel risk marker in CAD patients.


Subject(s)
Acute Coronary Syndrome/blood , Coronary Artery Disease/genetics , Intercellular Signaling Peptides and Proteins/blood , Intramolecular Oxidoreductases/blood , Macrophage Migration-Inhibitory Factors/blood , Adult , Aged , Aged, 80 and over , Angiography , Atherosclerosis , Case-Control Studies , Coronary Artery Disease/metabolism , Coronary Thrombosis/physiopathology , Enzyme-Linked Immunosorbent Assay , Female , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intramolecular Oxidoreductases/genetics , Macrophage Migration-Inhibitory Factors/genetics , Male , Middle Aged , Multivariate Analysis , Plaque, Atherosclerotic/metabolism , Recombinant Proteins/chemistry , Retrospective Studies , Risk Factors , Thrombosis/pathology , Young Adult
6.
Int J Cardiol ; 176(3): 923-9, 2014 Oct 20.
Article in English | MEDLINE | ID: mdl-25194861

ABSTRACT

BACKGROUND: Monocyte migration and their differentiation into macrophages critically regulate vascular inflammation and atherogenesis and are governed by macrophage migration inhibitory factor (MIF). Gremlin-1 binds to MIF. Current experimental evidences present Gremlin-1 as a potential physiological agent that might counter-regulate the inflammatory attributes of MIF. METHODS AND RESULTS: We found that Gremlin-1 inhibited MIF-dependent monocyte migration and adhesion to activated endothelial cells in flow chamber perfusion assay in vitro and to the injured carotid artery of WT and ApoE-/- mice in vivo as deciphered by intravital microscopy. Intravenous administration of Gremlin-1, but not of control protein, significantly reduced leukocyte recruitment towards the inflamed carotid artery of ApoE-/- mice. Besides, leukocytes from MIF-/- when administered into ApoE-/- mice showed lesser adhesion as compared to wild type. In the presence of Gremlin-1 however, adhesion of wild type, but not of MIF-/- leukocytes, to the carotid artery was significantly inhibited as compared to control. Gremlin-1 also inhibited the MIF-induced differentiation of monocytes into macrophages. Gremlin-1 substantially inhibited the anti-apoptotic impact of MIF on monocytes against BH3 mimetic ABT-737-induced apoptosis as verified by Annexin V-binding, caspase 3 activity, and mitochondrial depolarization. CONCLUSIONS: Therefore Gremlin-1 can modulate MIF dependent monocyte adhesion, migration, differentiation and survival.


Subject(s)
Cell Movement/physiology , Intercellular Signaling Peptides and Proteins/pharmacology , Macrophage Migration-Inhibitory Factors/antagonists & inhibitors , Macrophage Migration-Inhibitory Factors/physiology , Monocytes/physiology , Animals , Cell Line , Cell Movement/drug effects , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Monocytes/drug effects
7.
Dis Model Mech ; 6(2): 332-41, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23223679

ABSTRACT

Atrial fibrillation (AF) is the most common cardiac arrhythmia and carries a significant risk of stroke and heart failure. The molecular etiologies of AF are poorly understood, leaving patients with limited therapeutic options. AF has been recognized as an inherited disease in almost 30% of patient cases. However, few genetic loci have been identified and the mechanisms linking genetic variants to AF susceptibility remain unclear. By sequencing 193 probands with lone AF, we identified a Q76E variant within the coding sequence of the bone morphogenetic protein (BMP) antagonist gremlin-2 (GREM2) that increases its inhibitory activity. Functional modeling in zebrafish revealed that, through regulation of BMP signaling, GREM2 is required for cardiac laterality and atrial differentiation during embryonic development. GREM2 overactivity results in slower cardiac contraction rates in zebrafish, and induction of previously identified AF candidate genes encoding connexin-40, sarcolipin and atrial natriuretic peptide in differentiated mouse embryonic stem cells. By live heart imaging in zebrafish overexpressing wild-type or variant GREM2, we found abnormal contraction velocity specifically in atrial cardiomyocytes. These results implicate, for the first time, regulators of BMP signaling in human AF, providing mechanistic insights into the pathogenesis of the disease and identifying potential new therapeutic targets.


Subject(s)
Atrial Fibrillation/genetics , Cell Differentiation/genetics , Disease Models, Animal , Heart Atria/physiopathology , Intercellular Signaling Peptides and Proteins/genetics , Myocytes, Cardiac/pathology , Zebrafish Proteins/genetics , Amino Acid Sequence , Amino Acid Substitution , Animals , Arrhythmias, Cardiac/complications , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/pathology , Arrhythmias, Cardiac/physiopathology , Atrial Fibrillation/complications , Atrial Fibrillation/physiopathology , Bone Morphogenetic Proteins/metabolism , Cytokines , Female , Gene Expression Regulation, Developmental , Heart Atria/embryology , Heart Atria/pathology , Heart Rate/physiology , Humans , Intercellular Signaling Peptides and Proteins/chemistry , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mice , Middle Aged , Molecular Sequence Data , Myocytes, Cardiac/metabolism , Organogenesis/genetics , Pedigree , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/chemistry , Zebrafish Proteins/metabolism
8.
PLoS One ; 7(6): e38376, 2012.
Article in English | MEDLINE | ID: mdl-22693633

ABSTRACT

BACKGROUND: Chronic inflammation promotes atherosclerosis in cardiovascular disease and is a major prognostic factor for patients undergoing percutaneous coronary intervention (PCI). Macrophage migration inhibitory factor (MIF) is involved in the progress of atherosclerosis and plaque destabilization and plays a pivotal role in the development of acute coronary syndromes (ACS). Little is known to date about the clinical impact of MIF in patients with symptomatic coronary artery disease (CAD). METHODS AND RESULTS: In a pilot study, 286 patients with symptomatic CAD (n = 119 ACS, n = 167 stable CAD) undergoing PCI were consecutively evaluated. 25 healthy volunteers served as control. Expression of MIF was consecutively measured in patients at the time of PCI. Baseline levels of interleukin 6 (IL-6), "regulated upon activation, normal T-cell expressed, and secreted" (RANTES) and monocyte chemoattractant protein-1 (MCP-1) were measured by Bio-Plex Cytokine assay. C-reactive protein (CRP) was determined by Immunoassay. Patients with ACS showed higher plasma levels of MIF compared to patients with stable CAD and control subjects (median 2.85 ng/mL, interquartile range (IQR) 3.52 versus median 1.22 ng/mL, IQR 2.99, versus median 0.1, IQR 0.09, p<0.001). Increased MIF levels were associated with CRP and IL-6 levels and correlated with troponin I (TnI) release (spearman rank coefficient: 0.31, p<0.001). Patients with ACS due to plaque rupture showed significantly higher plasma levels of MIF than patients with flow limiting stenotic lesions (p = 0.002). CONCLUSION: To our knowledge this is the first study, demonstrating enhanced expression of MIF in ACS. It is associated with established inflammatory markers, correlates with the extent of cardiac necrosis marker release after PCI and is significantly increased in ACS patients with "culprit" lesions. Further attempts should be undertaken to characterize the role of MIF for risk assessment in the setting of ACS.


Subject(s)
Acute Coronary Syndrome/blood , Acute Coronary Syndrome/immunology , Inflammation/blood , Inflammation/immunology , Macrophage Migration-Inhibitory Factors/blood , Aged , Aged, 80 and over , C-Reactive Protein/metabolism , Chemokine CCL2/blood , Coronary Artery Disease/blood , Female , Humans , Immunoassay , Interleukin-6/blood , Male , Middle Aged
10.
Dev Dyn ; 235(10): 2881-8, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16921498

ABSTRACT

The protein related to Dan and Cerberus, or PRDC, is a secreted glycoprotein, which belongs to the DAN subfamily of bone morphogenetic protein (BMP) antagonists. In zebrafish, prdc is expressed initially around 17 hours postfertilization in the developing eyes and the first two pharyngeal arches. Expression in the eye starts in the outer layers of the optic cup. Later, prdc expression domains are juxtaposed at the edges of the optic cup surrounding the choroid fissure, then gradually becoming restricted to a small site in the ventral marginal zone. Prdc expression in the arch mesenchyme expands stepwise to the remaining posterior arches. Prdc is also detectable in the ventral part of the somites and the mesenchyme of the swim bladder. The relatively late appearance during development is a unique feature of Prdc among BMP antagonists. Moreover, the complexity of the prdc expression pattern suggests possible roles in eye development, pharyngeal arch remodeling, somitogenesis, and swim bladder organogenesis.


Subject(s)
Air Sacs/metabolism , Branchial Region/metabolism , Eye/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Somites/metabolism , Zebrafish Proteins/genetics , Zebrafish/genetics , Air Sacs/embryology , Amino Acid Sequence , Animals , Branchial Region/embryology , Eye/embryology , Gene Expression Regulation, Developmental/genetics , In Situ Hybridization , Molecular Sequence Data , Phylogeny , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Somites/enzymology , Zebrafish/embryology
SELECTION OF CITATIONS
SEARCH DETAIL
...