Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
PLoS Biol ; 19(9): e3001397, 2021 09.
Article in English | MEDLINE | ID: mdl-34499640

ABSTRACT

In 2018, the first registry dedicated to preregistration of animal study protocols was launched. Despite international support, the overall number of (pre)registered protocols is still low, illustrating the need for pushing the preregistration agenda among researchers and policymakers.


Subject(s)
Registries , Research Design , Animal Experimentation/standards , Animals
2.
Heart Fail Rev ; 26(6): 1515-1524, 2021 11.
Article in English | MEDLINE | ID: mdl-32335789

ABSTRACT

The transverse aortic constriction (TAC) model is frequently used to study adverse cardiac remodeling upon pressure overload. We set out to define the most important characteristics that define the degree of cardiac remodeling in this model. A systematic review and meta-analyses were performed on studies using the TAC mouse/rat model and reporting echocardiographic outcome parameters. We included all animal studies in which a constriction around the transverse aorta and at least one of the predefined echocardiography or MRI outcome parameters were assessed. A total of 502 articles and > 3000 wild-type, untreated animals undergoing TAC were included in this study and referenced to a control group. The duration of aortic constriction correlated to the degree of adverse remodeling. However, the mouse data is strongly biased by the preferential use of male C57Bl/6 mice (66% of studies). Furthermore, mostly ketamine/xylazine anesthetics, 27G needle constriction, and silk sutures are used. Nonetheless, despite the homogeneity in experimental design, the model contained a substantial degree of heterogeneity in the functional outcome measures. When looking at study quality, only 12% reported randomization, 23% mentioned any sort of blinding, 25% adequately addressed the outcomes, and an amazingly low percentage (2%) showed sample size calculation. Meta-analyses did not detect specific study characteristics that explained the heterogeneity in the reported outcome measures, however this might be related to the strong bias towards the use of specific mouse lines, sex as well as age or to poor reporting of characteristics of study quality.


Subject(s)
Aorta , Heart Failure , Animals , Constriction , Echocardiography , Female , Humans , Male , Mice , Mice, Inbred C57BL , Rats
3.
Eur Heart J ; 38(11): 828-836, 2017 Mar 14.
Article in English | MEDLINE | ID: mdl-27432019

ABSTRACT

AIMS: Myocardial infarction (MI) triggers an intense inflammatory response that is associated with infarct expansion and is detrimental for cardiac function. Interleukin (IL)-1ß and IL-18 are key players in this response and are controlled by the NLRP3-inflammasome. In the current study, we therefore hypothesized that selective inhibition of the NLRP3-inflammasome reduces infarct size and preserves cardiac function in a porcine MI model. METHODS AND RESULTS: Thirty female landrace pigs were subjected to 75 min transluminal balloon occlusion and treated with the NLRP3-inflammasome inhibitor MCC950 (6 or 3 mg/kg) or placebo for 7 days in a randomized, blinded fashion. After 7 days, 3D-echocardiography was performed to assess cardiac function and Evans blue/TTC double staining was executed to assess the area at risk (AAR) and infarct size (IS). The IS/AAR was lower in the 6 mg/kg group (64.6 ± 8.8%, P = 0.004) and 3 mg/kg group (69.7 ± 7.2%, P = 0.038) compared with the control group (77.5 ± 6.3%). MCC950 treatment markedly preserved left ventricular ejection fraction in treated animals (6 mg/kg 47 ± 8%, P = 0.001; 3 mg/kg 45 ± 7%, P = 0.031; control 37 ± 6%). Myocardial neutrophil influx was attenuated in treated compared with non-treated animals (6 mg/kg 132 ± 72 neutrophils/mm2, P = 0.035; 3 mg/kg 207 ± 210 neutrophils/mm2, P = 0.5; control 266 ± 158 neutrophils/mm2). Myocardial IL-1ß levels were dose-dependently reduced in treated animals. CONCLUSIONS: NLRP3-inflammasome inhibition reduces infarct size and preserves cardiac function in a randomized, blinded translational large animal MI model. Hence, NLRP3-inflammasome inhibition may have therapeutic potential in acute MI patients.


Subject(s)
Heterocyclic Compounds, 4 or More Rings/pharmacology , Myocardial Infarction/prevention & control , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Sulfones/pharmacology , Animals , Balloon Occlusion , Biomarkers/metabolism , Echocardiography/methods , Female , Furans , Hemodynamics/physiology , Indenes , Inflammasomes/antagonists & inhibitors , Leukocytes, Mononuclear/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Random Allocation , Sulfonamides , Swine
4.
Cell Rep ; 38(1): 110189, 2022 01 04.
Article in English | MEDLINE | ID: mdl-34986347

ABSTRACT

Fibrosis is a major cause of mortality worldwide, characterized by myofibroblast activation and excessive extracellular matrix deposition. Systemic sclerosis is a prototypic fibrotic disease in which CXCL4 is increased and strongly correlates with skin and lung fibrosis. Here we aim to elucidate the role of CXCL4 in fibrosis development. CXCL4 levels are increased in multiple inflammatory and fibrotic mouse models, and, using CXCL4-deficient mice, we demonstrate the essential role of CXCL4 in promoting fibrotic events in the skin, lungs, and heart. Overexpressing human CXCL4 in mice aggravates, whereas blocking CXCL4 reduces, bleomycin-induced fibrosis. Single-cell ligand-receptor analysis predicts CXCL4 to affect endothelial cells and fibroblasts. In vitro, we confirm that CXCL4 directly induces myofibroblast differentiation and collagen synthesis in different precursor cells, including endothelial cells, by stimulating endothelial-to-mesenchymal transition. Our findings identify a pivotal role of CXCL4 in fibrosis, further substantiating the potential role of neutralizing CXCL4 as a therapeutic strategy.


Subject(s)
Extracellular Matrix/pathology , Myofibroblasts/metabolism , Platelet Factor 4/metabolism , Pulmonary Fibrosis/pathology , Scleroderma, Systemic/pathology , Animals , Bleomycin/toxicity , Cell Line , Collagen/biosynthesis , Disease Models, Animal , Endothelial Cells/cytology , Endothelial Cells/metabolism , Epithelial-Mesenchymal Transition/physiology , Human Umbilical Vein Endothelial Cells , Humans , Lung/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Myofibroblasts/cytology , Pericytes/metabolism , Platelet Factor 4/genetics , Stromal Cells/cytology , Stromal Cells/metabolism
5.
J Am Heart Assoc ; 6(9)2017 Aug 30.
Article in English | MEDLINE | ID: mdl-28855167

ABSTRACT

BACKGROUND: Peripheral artery disease (PAD) is one of the most common clinical presentations of atherosclerosis, and its prevalence is still increasing. Despite improvement of health care, morbidity and mortality risks remain high, including the risk of amputation. GDF15 (growth differentiation factor 15) is a member of the transforming growth factor family that is involved in apoptosis and inflammation; therefore, GDF15 is a potential biomarker to identify patients at high risk of adverse clinical outcomes. METHODS AND RESULTS: Circulating GDF15 levels were measured using a multiplex immunoassay in patients with critical limb ischemia and PAD from 2 different patient cohorts that included patients with clinically manifest PAD: the JUVENTAS (Rejuvenating Endothelial Progenitor Cells via Transcutaneous Intra-Arterial Supplementation) trial (n=160, 67 major events; critical limb ischemia) and the Athero-Express Biobank (n=386, 64 major events; PAD). Kaplan-Meier curves demonstrated that high levels of GDF15 were associated with increased risk of major events, defined as major amputation (at or above the ankle joint) and all-cause mortality, in both cohorts (highest versus lowest, JUVENTAS: hazard ratio: 4.01 [95% confidence interval, 2.05-7.84; P<0.0001]; Athero-Express: hazard ratio: 3.27 [95% confidence interval, 1.64-6.54; P=0.0008]). In the JUVENTAS trial, this was more pronounced in women. Cox proportional multivariable regression models with median follow-up of 3 years, corrected for common confounders, showed hazard ratios of 1.70 (95% confidence interval, 1.18-2.69; P=0.0053) and 1.57 (95% confidence interval, 1.02-2.41; P=0.041) per 2.78-fold increase of GDF15 in JUVENTAS and Athero-Express, respectively. CONCLUSIONS: High GDF15 levels are associated with increased risk of major amputation and/or death in PAD patients. GDF15 levels could be of additive value to identify patients who are at high risk of amputation or death and could help guide treatment choices.


Subject(s)
Amputation, Surgical , Growth Differentiation Factor 15/blood , Ischemia/blood , Peripheral Arterial Disease/blood , Aged , Amputation, Surgical/adverse effects , Amputation, Surgical/mortality , Biological Specimen Banks , Biomarkers/blood , Chi-Square Distribution , Clinical Decision-Making , Critical Illness , Endothelial Progenitor Cells/transplantation , Female , Humans , Ischemia/diagnosis , Ischemia/mortality , Ischemia/surgery , Kaplan-Meier Estimate , Logistic Models , Male , Middle Aged , Multivariate Analysis , Patient Selection , Peripheral Arterial Disease/diagnosis , Peripheral Arterial Disease/mortality , Peripheral Arterial Disease/surgery , Proportional Hazards Models , Randomized Controlled Trials as Topic , Risk Assessment , Risk Factors , Time Factors , Treatment Outcome , Up-Regulation
6.
Sci Rep ; 7(1): 17045, 2017 12 06.
Article in English | MEDLINE | ID: mdl-29213128

ABSTRACT

Hypertension is one of the most common risk factors for the development heart failure in the general population. Inflammation plays a central role in this adverse remodeling and eventually to the development of heart failure. Circulating levels of Complement factor 5a (C5a) are increased in hypertensive patients and the C5a receptor is associated with the presence of cardiac fibrosis and inflammation in an experimental hypertension model. To test if C5aR is involved in adverse cardiac remodeling following pressure-overload, we induced transverse aortic constriction (TAC) in wildtype and C5a receptor deficient mice (C5aR-/-). Six weeks after TAC, C5aR-/- animals showed a similar degree of cardiac hypertrophy and decrease in cardiac function as wild type mice (End Systolic Volume; 50.30±5.32 µl vs. 55.81±8.16 µl). In addition, other features of adverse cardiac remodeling like cardiomyocyte cell size (WGA staining), fibrosis (picrosirius red staining) or collagen degradation (matrix metalloproteinase activity assay) did not differ either. In conclusion, full body C5aR deficiency does not affect adverse cardiac remodeling after pressure-overload. However, our finding are in contrast with C5a inhibition studies. Our observations do present the role of C5a-C5aR in adverse cardiac remodeling and heart failure as controversial at the least.


Subject(s)
Cardiomegaly/pathology , Receptor, Anaphylatoxin C5a/genetics , Ventricular Remodeling , Actins/metabolism , Animals , Collagen/genetics , Collagen/metabolism , Constriction, Pathologic , Fibrosis , Leukocytes/cytology , Leukocytes/metabolism , Matrix Metalloproteinases/metabolism , Mice , Mice, Inbred BALB C , Myocardium/pathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Receptor, Anaphylatoxin C5a/deficiency
7.
Cardiovasc Res ; 113(8): 892-905, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28371893

ABSTRACT

AIMS: Tissue transglutaminase (tTG) is induced in injured and remodelling tissues, and modulates cellular phenotype, while contributing to matrix cross-linking. Our study tested the hypothesis that tTG may be expressed in the pressure-overloaded myocardium, and may regulate cardiac function, myocardial fibrosis and chamber remodelling. METHODS AND RESULTS: In order to test the hypothesis, wild-type and tTG null mice were subjected to pressure overload induced through transverse aortic constriction. Moreover, we used isolated cardiac fibroblasts and macrophages to dissect the mechanisms of tTG-mediated actions. tTG expression was upregulated in the pressure-overloaded mouse heart and was localized in cardiomyocytes, interstitial cells, and in the extracellular matrix. In contrast, expression of transglutaminases 1, 3, 4, 5, 6, 7 and FXIII was not induced in the remodelling myocardium. In vitro, transforming growth factor (TGF)-ß1 stimulated tTG synthesis in cardiac fibroblasts and in macrophages through distinct signalling pathways. tTG null mice had increased mortality and enhanced ventricular dilation following pressure overload, but were protected from diastolic dysfunction. tTG loss was associated with a hypercellular cardiac interstitium, reduced collagen cross-linking, and with accentuated matrix metalloproteinase (MMP)2 activity in the pressure-overloaded myocardium. In vitro, tTG did not modulate TGF-ß-mediated responses in cardiac fibroblasts; however, tTG loss was associated with accentuated proliferative activity. Moreover, when bound to the matrix, recombinant tTG induced synthesis of tissue inhibitor of metalloproteinases (TIMP)-1 through transamidase-independent actions. CONCLUSIONS: Following pressure overload, endogenous tTG mediates matrix cross-linking, while protecting the remodelling myocardium from dilation by exerting matrix-preserving actions.


Subject(s)
Extracellular Matrix/metabolism , GTP-Binding Proteins/metabolism , Myocardium/metabolism , Transglutaminases/metabolism , Ventricular Remodeling/physiology , Animals , Female , Fibroblasts/metabolism , Fibrosis/metabolism , Hypertrophy, Left Ventricular/physiopathology , Male , Mice, Knockout , Myocytes, Cardiac/metabolism , Pressure , Protein Glutamine gamma Glutamyltransferase 2 , Transforming Growth Factor beta/metabolism
8.
Sci Rep ; 7(1): 9193, 2017 08 23.
Article in English | MEDLINE | ID: mdl-28835616

ABSTRACT

An involement of Toll-like receptor 2 (TLR2) has been established in cardiac dysfunction after acute myocardial infarction; however, its role in chronic pressure overload is unclear. We sought to evaluate the role of TLR2 in cardiac hypertrophy, fibrosis and dysfunction in sustained pressure overload. We induced pressure overload via transverse aortic constriction (TAC) in TLR2-/- and wild type (WT) mice, and followed temporal changes over 8 weeks. Despite similar increases in heart weight, left ventricular (LV) ejection fraction (EF) and diastolic function (mitral E/A ratio) were preserved in TLR2-/- mice but impaired in WT mice following TAC. TAC produced less LV fibrosis in TLR2-/- mice associated with lower mRNA levels of collagen genes (Col1a1 and Col3a1) and lower protein level of TGFbeta1, compared to WT mice. Following TAC, the influx of macrophages and CD3 T cells into LV was similar between TLR2-/- and WT mice, whereas levels of cyto/chemokines were lower in the heart and plasma in TLR2-/- mice. TLR2-/- bone marrow-derived cells protected against LVEF decline and fibrosis following TAC. Our findings show that leukocytic TLR2 deficiency protects against LV dysfunction and fibrosis probably via a reduction in inflammatory signaling in sustained pressure overload.


Subject(s)
Blood Pressure , Heart Diseases/etiology , Heart Diseases/physiopathology , Leukocytes/metabolism , Toll-Like Receptor 2/deficiency , Animals , Biopsy , Cytokines/metabolism , Disease Models, Animal , Fibrosis , Heart Diseases/metabolism , Heart Diseases/pathology , Heart Function Tests , Immunohistochemistry , Inflammation Mediators/metabolism , Male , Mice , Mice, Knockout , Ventricular Function, Left , Ventricular Remodeling/genetics
9.
Sci Rep ; 7(1): 18039, 2017 12 21.
Article in English | MEDLINE | ID: mdl-29269840

ABSTRACT

Heart failure after myocardial infarction (MI) depends on infarct size and adverse left ventricular (LV) remodelling, both influenced by the inflammatory response. Leukocyte-associated immunoglobulin-like receptor 1 (LAIR-1) is an inhibitory receptor of ITAM-dependent cell activation, present on almost all immune cells. We investigated regulation of LAIR-1 leukocyte expression after MI in patients and hypothesized that its absence in a mouse model of MI would increase infarct size and adverse remodelling. In patients, LAIR-1 expression was increased 3 days compared to 6 weeks after MI on circulating monocytes (24.8 ± 5.3 vs. 21.2 ± 5.1 MFI, p = 0.008) and neutrophils (12.9 ± 4.7 vs. 10.6 ± 3.1 MFI, p = 0.046). In WT and LAIR-1-/- mice, infarct size after ischemia-reperfusion injury was comparable (37.0 ± 14.5 in WT vs. 39.4 ± 12.2% of the area at risk in LAIR-1-/-, p = 0.63). Remodelling after permanent left coronary artery ligation did not differ between WT and LAIR-1-/- mice (end-diastolic volume 133.3 ± 19.3 vs. 132.1 ± 27.9 µL, p = 0.91 and end-systolic volume 112.1 ± 22.2 vs. 106.9 ± 33.5 µL, p = 0.68). Similarly, no differences were observed in inflammatory cell influx or fibrosis. In conclusion, LAIR-1 expression on monocytes and neutrophils is increased in the acute phase after MI in patients, but the absence of LAIR-1 in mice does not influence infarct size, inflammation, fibrosis or adverse cardiac remodelling.


Subject(s)
Myocardial Infarction/metabolism , Myocardium/metabolism , Receptors, Immunologic/metabolism , Reperfusion Injury/metabolism , Aged , Animals , Disease Models, Animal , Female , Fibrosis/genetics , Fibrosis/metabolism , Fibrosis/pathology , Gene Expression Regulation , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Knockout , Middle Aged , Monocytes/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardium/pathology , Neutrophils/metabolism , Receptors, Immunologic/genetics , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Ventricular Remodeling/physiology
12.
J Cardiovasc Transl Res ; 5(6): 837-47, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22956156

ABSTRACT

Cardiac fibroblasts are key cellular effectors of cardiac repair; their phenotype and function are modulated by interactions with extracellular matrix proteins. This review manuscript discusses the effects of the extracellular matrix on the inflammatory and reparative properties of fibroblasts in the infarcted myocardium. Early generation of matrix fragments in the infarct induces a pro-inflammatory and matrix-degrading fibroblast phenotype. Formation of a fibrin/fibronectin-rich provisional matrix serves as a conduit for migration of fibroblasts into the infarcted area. Induction of ED-A fibronectin and nonfibrillar collagens may contribute to myofibroblast transdifferentiation. Upregulation of matricellular proteins promotes transduction of growth factor and cytokine-mediated signals. As the scar matures, matrix cross-linking, clearance of matricellular proteins, and reduced growth factor signaling cause deactivation and apoptosis of reparative infarct fibroblasts. Understanding the effects of matrix components on infarct fibroblasts may guide the design of peptides that reproduce, or inhibit, specific matricellular functions, attenuating adverse remodeling.


Subject(s)
Extracellular Matrix/metabolism , Fibroblasts/metabolism , Myocardial Infarction/metabolism , Myocardium/metabolism , Animals , Cytokines/metabolism , Extracellular Matrix/pathology , Fibroblasts/pathology , Fibrosis , Humans , Inflammation Mediators/metabolism , Myocardial Infarction/pathology , Myocardium/pathology , Myofibroblasts/metabolism , Myofibroblasts/pathology , Phenotype , Signal Transduction , Ventricular Remodeling
13.
Atherosclerosis ; 222(2): 355-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22498256

ABSTRACT

BACKGROUND: Platelets foster an inflammatory environment that influences atherosclerotic lesion progression and facilitates plaque rupture, in addition to their role in acute thrombus formation. The route of entry of platelets into the atherosclerotic plaque and their exact location inside the plaque are however not completely understood. METHODS AND RESULTS: 188 carotid plaques were examined for the presence of platelets using immunohistochemistry (CD42b), and 76/188 (40.4%) were platelet positive. Platelets were observed in intraplaque hemorrhages, around plaque microvessels, mostly without leakage of erythrocytes; and in mural thrombi. Platelet positive staining was associated with a higher plaque microvessel density, and elevated plaque-levels of interleukin-8. CONCLUSION: Due to their short life span, platelets reflect recent bleeding. It can be hypothesized that platelets might serve as a marker for leaky microvessels inside atherosclerotic plaques that are at risk for development, or progression of plaque hemorrhage.


Subject(s)
Blood Platelets/pathology , Capillary Permeability , Carotid Arteries/pathology , Carotid Artery Diseases/pathology , Hemorrhage/pathology , Microvessels/pathology , Aged , Biomarkers/analysis , Blood Platelets/immunology , Carotid Arteries/immunology , Carotid Artery Diseases/blood , Carotid Artery Diseases/complications , Carotid Artery Diseases/immunology , Disease Progression , Female , Hemorrhage/blood , Hemorrhage/etiology , Hemorrhage/immunology , Humans , Immunohistochemistry , Interleukin-8/analysis , Male , Microvessels/immunology , Microvessels/metabolism , Middle Aged , Plaque, Atherosclerotic , Platelet Glycoprotein GPIb-IX Complex/analysis , Rupture, Spontaneous
SELECTION OF CITATIONS
SEARCH DETAIL