Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Regul Toxicol Pharmacol ; 149: 105623, 2024 May.
Article in English | MEDLINE | ID: mdl-38631606

ABSTRACT

The Bone-Marrow derived Dendritic Cell (BMDC) test is a promising assay for identifying sensitizing chemicals based on the 3Rs (Replace, Reduce, Refine) principle. This study expanded the BMDC benchmarking to various in vitro, in chemico, and in silico assays targeting different key events (KE) in the skin sensitization pathway, using common substances datasets. Additionally, a Quantitative Structure-Activity Relationship (QSAR) model was developed to predict the BMDC test outcomes for sensitizing or non-sensitizing chemicals. The modeling workflow involved ISIDA (In Silico Design and Data Analysis) molecular fragment descriptors and the SVM (Support Vector Machine) machine-learning method. The BMDC model's performance was at least comparable to that of all ECVAM-validated models regardless of the KE considered. Compared with other tests targeting KE3, related to dendritic cell activation, BMDC assay was shown to have higher balanced accuracy and sensitivity concerning both the Local Lymph Node Assay (LLNA) and human labels, providing additional evidence for its reliability. The consensus QSAR model exhibits promising results, correlating well with observed sensitization potential. Integrated into a publicly available web service, the BMDC-based QSAR model may serve as a cost-effective and rapid alternative to lab experiments, providing preliminary screening for sensitization potential, compound prioritization, optimization and risk assessment.


Subject(s)
Benchmarking , Dendritic Cells , Quantitative Structure-Activity Relationship , Dendritic Cells/drug effects , Humans , Animals , Support Vector Machine , Computer Simulation , Dermatitis, Allergic Contact , Allergens/toxicity , Animal Testing Alternatives/methods , Bone Marrow Cells/drug effects , Local Lymph Node Assay , Mice
2.
Contact Dermatitis ; 90(2): 169-181, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37927141

ABSTRACT

BACKGROUND: Bisphenol (BP-)A is a chemical used in Europe to produce polycarbonate plastics and epoxy resin or as colour developer in thermal paper. Due to its toxicity, BPA presence was restricted by European regulations. Therefore, substitute chemicals are replacing BPA. OBJECTIVE: To assess the allergenic sensitizing potential of 27 substitutes to BPA used in the industry. METHODS: The expression of two costimulatory molecules and six cytokines were analysed by flow cytometry in mouse bone marrow-derived dendritic cells (BMDCs) exposed to the chemicals. RESULTS: All substances except one induced overexpression of at least one receptor and were thus identified as having allergenic sensitizing potential. Based on the BMDC model, they were classified as extreme (1 out of 27), strong (20 out of 27) and moderate (5 out of 27) sensitizers. BPA was classified as a moderate sensitizer and BPF was the only substitute classified as a non-sensitizer. The more potent substitutes induced more than 2-fold secretion of CCL3, CCL4 and/or CCL5 by dendritic cells. CONCLUSION: Most of the BPA substitutes tested in this study have an allergenic sensitizing potential; 24 of them being more potent than BPA itself. Only BPE, BPF and 2,4-BPS appeared to be weaker sensitizers than BPA.


Subject(s)
Allergens , Dermatitis, Allergic Contact , Animals , Mice , Allergens/adverse effects , Sulfones/analysis , Sulfones/pharmacology , Dermatitis, Allergic Contact/etiology , Phenols/toxicity , Benzhydryl Compounds/toxicity
3.
Contact Dermatitis ; 90(3): 211-234, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37852624

ABSTRACT

BACKGROUND: Chemical-induced allergies at workplace represent a significant occupational health issue. These substances must be properly identified as sensitizers. In previous studies, an original model using mouse bone marrow-derived dendritic cells (BMDC) was developed for this purpose. OBJECTIVES: The aim of this study was to evaluate the predictive capacity of the BMDC model with a large panel of sensitizers (including pre- and pro-haptens) and non-sensitizers. METHODS: The readout from the BMDC model is based on expression levels of six phenotypic markers measured by flow cytometry. RESULTS: The results indicate that 29 of the 37 non-sensitizers, and 81 of the 86 sensitizers were correctly classified compared to the Local Lymph Node Assay (LLNA). Statistical analysis revealed the BMDC model to have a sensitivity of 94%, a specificity of 78%, and an accuracy of 89%. The EC2 (Effective Concentration) values calculated with this model allow sensitizers to be categorized into four classes: extreme, strong, moderate and weak. CONCLUSIONS: These excellent predictive performances show that the BMDC model discriminates between sensitizers and non-sensitizers with outstanding precision equal to or better than existing validated alternative models. Moreover, this model allows to predict sensitization potency of chemicals. The BMDC test could therefore be proposed as an additional tool to assess the sensitizing potential and potency of chemicals.


Subject(s)
Dermatitis, Allergic Contact , Mice , Animals , Dermatitis, Allergic Contact/diagnosis , Dermatitis, Allergic Contact/etiology , Haptens , Local Lymph Node Assay , Flow Cytometry , Allergens/adverse effects
4.
Front Oncol ; 10: 564674, 2020.
Article in English | MEDLINE | ID: mdl-33489875

ABSTRACT

Colorectal cancer (CRC) is one of the most common cancer worldwide. Chronic inflammation contributes to CRC development and progression. Emodin, is a natural anthraquinone derivative with anti-oxidant, anti-inflammatory, and anti-tumor activities. We used the AOM/DSS model of colitis-associated intestinal tumorigenesis to characterize the effect of Emodin on inflammation and tumorigenesis at weeks 3, 5, and 14 after initiation with AOM. At all three time points, Emodin (50 mg/kg) reduced inflammatory cell (i.e. CD11b+ and F4/80+) recruitment, cytokine (i.e. TNFα, IL1α/ß, IL6, CCL2, CXCL5) and pro-inflammatory enzymes (i.e. COX-2, NOS2) expression in the tumor microenvironment, while promoting recruitment of CD3+ T lymphocytes at 14 weeks. Emodin decreased the incidence of premalignant lesions (adenoma) at week 3, the incidence of dysplastic lesions and carcinomas at week 5, and the incidence, size and the invasiveness of carcinomas at week 14. Emodin also reduced the acute clinical intestinal symptoms (i.e. bleeding and diarrhea) during DSS treatment. In vitro, Emodin inhibited the expression of pro-inflammatory mediators by LPS-stimulated RAW 264.7 macrophages, and reduced viability, adhesion, migration, and fibroblasts-induced invasion of SW620 and HCT116 colon cancer cells. In conclusion, this work demonstrates that Emodin suppresses carcinogenesis-associated intestinal inflammation and prevents AOM/DSS-induced intestinal tumorigenesis and progression. These results instigate further studies on Emodin as a natural agent for the prevention or treatment of colorectal cancer.

5.
Stem Cell Reports ; 13(1): 1-9, 2019 07 09.
Article in English | MEDLINE | ID: mdl-31257133

ABSTRACT

Cancer stem cells (CSCs) are defined by their ability to regenerate a tumor upon transplantation. However, it is not yet clear whether tumors contain a single CSC population or different subsets of cells with mixed capacities for initiating primary and secondary tumors. Using two different identification strategies, we studied the overlap between metastatic stem cells and tumor-initiating cells (TICs) in the MMTV-PyMT model. Our results show that in the MMTV-PyMT model, Lin-CD90-ALDHhigh cells retained a high tumor-initiating potential (TIP) in orthotopic transplants, in contrast to Lin-CD24+CD90+, which retained higher metastatic capacity. Interestingly, suppression of TGFß signaling increased TIC numbers. We here describe the existence of distinct populations of CSCs with differing capacities to initiate tumors in the primary or the secondary site. Inhibiting TGFß signaling shifts the balance toward the former, which may have unanticipated implications for the therapeutic use of TGFß/TGFBR1 inhibitors.


Subject(s)
Breast Neoplasms/etiology , Breast Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Receptor, Transforming Growth Factor-beta Type I/antagonists & inhibitors , Animals , Biomarkers , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Epithelial-Mesenchymal Transition , Female , Fluorescent Antibody Technique , Humans , Immunophenotyping , Mice , Neoplasm Metastasis , Neoplastic Stem Cells/pathology , Signal Transduction , Transforming Growth Factor beta/metabolism
6.
Cells ; 8(5)2019 04 27.
Article in English | MEDLINE | ID: mdl-31035633

ABSTRACT

Fluid shear stress stimulates endothelial nitric oxide synthase (eNOS) activation and nitric oxide (NO) production through multiple kinases, including protein kinase A (PKA), AMP-activated protein kinase (AMPK), AKT and Ca2+/calmodulin-dependent protein kinase II (CaMKII). Membrane-associated guanylate kinase (MAGUK) with inverted domain structure-1 (MAGI1) is an adaptor protein that stabilizes epithelial and endothelial cell-cell contacts. The aim of this study was to assess the unknown role of endothelial cell MAGI1 in response to fluid shear stress. We show constitutive expression and co-localization of MAGI1 with vascular endothelial cadherin (VE-cadherin) in endothelial cells at cellular junctions under static and laminar flow conditions. Fluid shear stress increases MAGI1 expression. MAGI1 silencing perturbed flow-dependent responses, specifically, Krüppel-like factor 4 (KLF4) expression, endothelial cell alignment, eNOS phosphorylation and NO production. MAGI1 overexpression had opposite effects and induced phosphorylation of PKA, AMPK, and CAMKII. Pharmacological inhibition of PKA and AMPK prevented MAGI1-mediated eNOS phosphorylation. Consistently, MAGI1 silencing and PKA inhibition suppressed the flow-induced NO production. Endothelial cell-specific transgenic expression of MAGI1 induced PKA and eNOS phosphorylation in vivo and increased NO production ex vivo in isolated endothelial cells. In conclusion, we have identified endothelial cell MAGI1 as a previously unrecognized mediator of fluid shear stress-induced and PKA/AMPK dependent eNOS activation and NO production.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Cell Adhesion Molecules/physiology , Endothelial Cells/metabolism , Guanylate Kinases/physiology , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/metabolism , Shear Strength , Stress, Mechanical , Animals , Antigens, CD/metabolism , Cadherins/metabolism , Endothelial Cells/cytology , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Kruppel-Like Factor 4 , Mice , Mice, Transgenic , Signal Transduction
7.
Breast Cancer Res ; 20(1): 104, 2018 09 04.
Article in English | MEDLINE | ID: mdl-30180888

ABSTRACT

BACKGROUND: Obesity is a strong predictor of poor prognosis in breast cancer, especially in postmenopausal women. In particular, tumors in obese patients tend to seed more distant metastases, although the biology behind this observation remains poorly understood. METHODS: To elucidate the effects of the obese microenvironment on metastatic spread, we ovariectomized C57BL/6 J female mice and fed them either a regular diet (RD) or a high-fat diet (HFD) to generate a postmenopausal diet-induced obesity model. We then studied tumor progression to metastasis of Py230 and EO771 grafts. We analyzed and phenotyped the RD and HFD tumors and the surrounding adipose tissue by flow cytometry, qPCR, immunohistochemistry (IHC) and western blot. The influence of the microenvironment on tumor cells was assessed by performing cross-transplantation of RD and HFD tumor cells into other RD and HFD mice. The results were analyzed using the unpaired Student t test when comparing two variables, otherwise we used one-way or two-way analysis of variance. The relationship between two variables was calculated using correlation coefficients. RESULTS: Our results show that tumors in obese mice grow faster, are also less vascularized, more hypoxic, of higher grade and enriched in CD11b+Ly6G+ neutrophils. Collectively, this favors induction of the epithelial-to-mesenchymal transition and progression to claudin-low breast cancer, a subtype of triple-negative breast cancer that is enriched in cancer stem cells. Interestingly, transplanting HFD-derived tumor cells in RD mice transfers enhanced tumor growth and lung metastasis formation. CONCLUSIONS: These data indicate that a pro-metastatic effect of obesity is acquired by the tumor cells in the primary tumor independently of the microenvironment of the secondary site. Effects of postmenopausal obesity on primary breast cancer tumoursᅟ.


Subject(s)
Cell Proliferation , Mammary Neoplasms, Experimental/metabolism , Neoplastic Stem Cells/metabolism , Obesity/metabolism , Adipose Tissue/metabolism , Animals , Cell Line, Tumor , Diet, High-Fat/adverse effects , Disease Progression , Epithelial-Mesenchymal Transition , Female , Humans , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Metastasis , Neoplastic Stem Cells/pathology , Obesity/etiology , Obesity/genetics , Ovariectomy
8.
Sci Signal ; 10(488)2017 Jul 18.
Article in English | MEDLINE | ID: mdl-28720716

ABSTRACT

The phosphoinositide 3-kinase γ (PI3Kγ) plays a major role in leukocyte recruitment during acute inflammation and has been proposed to inhibit classical macrophage activation by driving immunosuppressive gene expression. PI3Kγ plays an important role in diet-induced obesity and insulin resistance. In seeking to determine the underlying molecular mechanisms, we showed that PI3Kγ action in high-fat diet-induced inflammation and insulin resistance depended largely on its role in the control of adiposity, which was due to PI3Kγ activity in a nonhematopoietic cell type. However, PI3Kγ activity in leukocytes was required for efficient neutrophil recruitment to adipose tissue. Neutrophil recruitment was correlated with proinflammatory gene expression in macrophages in adipose tissue, which triggered insulin resistance early during the development of obesity. Our data challenge the concept that PI3Kγ is a general suppressor of classical macrophage activation and indicate that PI3Kγ controls macrophage gene expression by non-cell-autonomous mechanisms, the outcome of which is context-dependent.


Subject(s)
Adipose Tissue/pathology , Class Ib Phosphatidylinositol 3-Kinase/physiology , Diet, High-Fat/adverse effects , Inflammation/prevention & control , Insulin Resistance , Leukocytes/enzymology , Obesity/complications , Animals , Gene Expression Profiling , Inflammation/etiology , Leukocytes/pathology , Lipid Metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/physiopathology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism
9.
J Leukoc Biol ; 97(1): 9-18, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25387836

ABSTRACT

Increase of blood capillary density at the interface between normal and ischemic tissue after acute MI reduces infarct size and improves cardiac function. Cardiac injury triggers the production of the matricellular component TSP-1, but its role in angiogenesis is not clear, as both anti- and proangiogenic properties have been reported. It is unknown whether TSP-1 is modulated by other factors released during cardiac injury. Among these, Ado is a well-known promoter of angiogenesis. This study determined whether Ado modulates TSP-1 expression and the implication on angiogenesis. Ado dose dependently increased the production of TSP-1 by human macrophages. With the use of agonists and antagonists of AdoRs, coupled to RNA interference, we observed that this effect is mediated via A2AR and A2BR. The Ado effect was reproduced by cholera toxin (Gs protein activator) and forskolin (adenylate cyclase activator) and blocked by the PKA inhibitor H89. Conditioned medium from Ado-treated macrophages stimulated microvessel outgrowth from aortic ring explants by 400%, and induced vessel formation in matrigel plugs. Microvessel outgrowth and vessel formation were blocked completely by addition of anti-TSP-1 antibodies to conditioned medium. Chronic administration of Ado to rats after MI maintained long-term expression of TSP-1 in the infarct border zone, and this was associated with enhanced border-zone vascularization. Ado up-regulates TSP-1 production by macrophages, resulting in stimulation of angiogenesis. The mechanism involves A2AR and A2BR and is mediated through the cAMP/PKA pathway. This information may be important when designing Ado-based therapies of angiogenesis.


Subject(s)
Adenosine/metabolism , Macrophages/metabolism , Myocardial Infarction/metabolism , Neovascularization, Physiologic/physiology , Thrombospondin 1/biosynthesis , Animals , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorescent Antibody Technique , Humans , Male , Microscopy, Confocal , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Up-Regulation
10.
EJNMMI Res ; 3(1): 65, 2013 Sep 12.
Article in English | MEDLINE | ID: mdl-24028474

ABSTRACT

BACKGROUND: Adenosine may have beneficial effects on left ventricular function after myocardial infarction (MI), but the magnitude of this effect on remote and MI areas is controversial. We assessed the long-term effects of adenosine after MI using electrocardiogram-triggered 18 F-fluorodeoxyglucose positron emission tomography. METHODS: Wistar rats were subjected to coronary ligation and randomized into three groups treated daily for 2 months by NaCl (control; n = 7), 2-chloroadenosine (CADO; n = 8) or CADO with 8-sulfophenyltheophilline, an antagonist of adenosine receptors (8-SPT; n = 8). RESULTS: After 2 months, control rats exhibited left ventricular remodelling, with increased end-diastolic volume and decreased ejection fraction. Left ventricular remodelling was not significantly inhibited by CADO. Segmental contractility, as assessed by the change in myocardial thickening after 2 months, was improved in CADO rats compared to control rats (+1.6% ± 0.8% vs. -2.3% ± 0.8%, p < 0.001). This improvement was significant in border (+5.6% ± 0.8% vs. +1.5% ± 0.8%, p < 0.001) and remote (-4.0% ± 1.0% vs. -10.4% ± 1.3%, p < 0.001) segments, but absent in MI segments. Histological analyses revealed that CADO reduced fibrosis, cardiomyocyte hypertrophy and apoptosis. Protective effects of CADO were blunted by 8-SPT. CONCLUSION: Long-term administration of adenosine protects the left ventricle from contractile dysfunction following MI.

11.
PLoS One ; 8(1): e54135, 2013.
Article in English | MEDLINE | ID: mdl-23326587

ABSTRACT

BACKGROUND: Administration of endothelial progenitor cells (EPC) represents a promising option to regenerate the heart after myocardial infarction, but is limited because of low recruitment and engraftment in the myocardium. Mobilization and migration of EPC are mainly controlled by stromal cell-derived factor 1α (SDF-1α) and its receptor CXCR4. We hypothesized that adenosine, a cardioprotective molecule, may improve the recruitment of EPC to the heart. METHODS: EPC were obtained from peripheral blood mononuclear cells of healthy volunteers. Expression of chemokines and their receptors was evaluated using microarrays, quantitative PCR, and flow cytometry. A Boyden chamber assay was used to assess chemotaxis. Recruitment of EPC to the infarcted heart was evaluated in rats after permanent occlusion of the left anterior descending coronary artery. RESULTS: Microarray analysis revealed that adenosine modulates the expression of several members of the chemokine family in EPC. Among these, CXCR4 was up-regulated by adenosine, and this result was confirmed by quantitative PCR (3-fold increase, P<0.001). CXCR4 expression at the cell surface was also increased. This effect involved the A(2B) receptor. Pretreatment of EPC with adenosine amplified their migration towards recombinant SDF-1α or conditioned medium from cardiac fibroblasts. Both effects were abolished by CXCR4 blocking antibodies. Adenosine also increased CXCR4 under ischemic conditions, and decreased miR-150 expression. Binding of miR-150 to the 3' untranslated region of CXCR4 was verified by luciferase assay. Addition of pre-miR-150 blunted the effect of adenosine on CXCR4. Administration of adenosine to rats after induction of myocardial infarction stimulated EPC recruitment to the heart and enhanced angiogenesis. CONCLUSION: Adenosine increases the migration of EPC. The mechanism involves A(2B) receptor activation, decreased expression of miR-150 and increased expression of CXCR4. These results suggest that adenosine may be used to enhance the capacity of EPC to revascularize the ischemic heart.


Subject(s)
Adenosine/administration & dosage , Endothelial Cells , MicroRNAs , Receptors, CXCR4 , Adenosine/metabolism , Cell Movement/drug effects , Cell- and Tissue-Based Therapy , Endothelial Cells/cytology , Endothelial Cells/metabolism , HEK293 Cells , Humans , Leukocytes, Mononuclear , MicroRNAs/genetics , MicroRNAs/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/therapy , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Regeneration/genetics , Stem Cells/cytology , Stem Cells/metabolism , Up-Regulation/drug effects
12.
Int J Clin Exp Med ; 6(1): 84-5, 2013.
Article in English | MEDLINE | ID: mdl-23236563

ABSTRACT

There is evidence that survival remains possible for infarction greater than 50% of the left ventricle in human, as well as in the rat infarct model. To our knowledge, survival has not been documented for infarctions involving the anterior, inferior and lateral wall leaving intact only the septal wall. An adult rat underwent a ligation of the left anterior descending coronary artery. ECG-triggered (18)F-ffluorodeoxyglucose Positron Emission Tomography revealed that 72% of the left ventricle was necrotic and totally akinetic. Although the left ventricular ejection fraction was severely impaired (9%), this rat survived and was asymptomatic after 2 months. The exact reasons for this incredible survival are still unclear.

13.
Br J Pharmacol ; 167(4): 854-67, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22646737

ABSTRACT

BACKGROUND AND PURPOSE: The purpose of the study was to investigate renal endothelium-dependent vasodilatation in a model of severe hypertension associated with kidney injury. EXPERIMENTAL APPROACH: Changes in perfusion pressure were measured in isolated, perfused kidneys taken from 18-week-old Wistar-Kyoto rat (WKY), spontaneously hypertensive rats (SHR) and SHR treated for 2 weeks with N(ω) -nitro-L-arginine methyl ester in the drinking water (L-NAME-treated SHR, 6 mg·kg(-1) ·day(-1) ). KEY RESULTS: Acetylcholine caused similar dose-dependent renal dilatation in the three groups. In vitro administration of indomethacin did not alter the vasodilatation, while the addition of N(w) -nitro-L-arginine (L-NA) produced a differential inhibition of the vasodilatation, (inhibition in WKY > SHR > L-NAME-treated SHR). Further addition of ODQ, an inhibitor of soluble guanylyl cyclase, abolished the responses to sodium nitroprusside but did not affect the vasodilatation to acetylcholine. However, the addition of TRAM-34 (or charybdotoxin) inhibitors of Ca(2+) -activated K(+) channels of intermediate conductance (K(Ca) 3.1), blocked the vasodilatation to acetylcholine, while apamin, an inhibitor of Ca(2+) -activated K(+) channels of small conductance (K(Ca) 2.3), was ineffective. Dilatation induced by an opener of K(Ca) 3.1/K(Ca) 2.3 channels, NS-309, was also blocked by TRAM-34, but not by apamin. The magnitude and duration of NS-309-induced vasodilatation and the renal expression of mRNA for K(Ca) 3.1, but not K(Ca) 2.3, channels followed the same ranking order (WKY < SHR < L-NAME-treated SHR). CONCLUSIONS AND IMPLICATIONS: In SHR kidneys, an EDHF-mediated response, involving activation of K(Ca) 3.1 channels, contributed to the mechanism of endothelium-dependent vasodilatation. In kidneys from L-NAME-treated SHR, up-regulation of this pathway fully compensated for the decrease in NO availability.


Subject(s)
Endothelium, Vascular/physiology , Hypertension/physiopathology , Intermediate-Conductance Calcium-Activated Potassium Channels/physiology , Kidney/physiology , Vasodilation/physiology , Acetylcholine/pharmacology , Animals , Apamin/pharmacology , Biological Factors/physiology , Enzyme Inhibitors/pharmacology , In Vitro Techniques , Indoles/pharmacology , Indomethacin/pharmacology , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Kidney/drug effects , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitroarginine/pharmacology , Nitroprusside/pharmacology , Oximes/pharmacology , Potassium Channel Blockers/pharmacology , Pyrazoles/pharmacology , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Vasodilation/drug effects , Vasodilator Agents/pharmacology
14.
Cardiovasc Res ; 94(3): 519-25, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22492674

ABSTRACT

AIMS: Circulating angiogenic cells (CAC) participate in cardiac repair. CAC recruitment to the ischaemic heart is mainly induced by the chemokine (C-X-C motif) receptor 4 (CXCR4)/stromal-cell derived factor-1α axis. However, CAC mobilization is only partly prevented by CXCR4 blockade, indicating that other mechanisms are involved. Since the expression of monocyte chemotactic protein 3 (MCP3) is increased in ischaemic hearts, we hypothesized that it may participate in CAC mobilization. METHODS AND RESULTS: CAC were obtained from peripheral blood mononuclear cells of healthy volunteers. In vitro migration of CAC was concentration-dependently increased by recombinant MCP3 (one-fold increase, P= 0.001), and this effect was inhibited by antibodies neutralizing the chemokine (C-C motif) receptor 1 (CCR1). CCR1 expression at the surface of CAC was confirmed by flow cytometry. Conditioned medium from heparan sulfate-activated macrophages, which contained MCP3, induced the migration of CAC (one-fold increase, P= 0.01). This increase was partly inhibited by CCR1 antibodies. The migration of CAC was also stimulated by macrophage inflammatory protein 3ß. This effect was blocked by CCR7 antibodies and was of lower magnitude than that of MCP3. MCP3 induced the formation of blood vessels in Matrigel plugs implanted in mice (1.5-fold increase, P< 0.001). This effect was abrogated by anti-CCR1 antibodies. CONCLUSION: This study shows that MCP3 stimulates the migration of CAC and angiogenesis, suggesting that MCP3 may be useful to improve cardiac repair.


Subject(s)
Cell Movement , Chemokine CCL7/metabolism , Receptors, CXCR4/metabolism , Animals , Cells, Cultured , Chemotactic Factors/metabolism , Flow Cytometry , Humans , Macrophage Inflammatory Proteins/metabolism , Mice , Mice, Inbred C57BL , Recombinant Proteins/metabolism
15.
Int J Cardiovasc Imaging ; 28(6): 1407-15, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22116590

ABSTRACT

The rat myocardial infarction (MI) model is widely used to study left ventricular (LV) remodeling. In this study, acipimox-enhanced (18)F-Fluorodeoxyglucose (FDG) gated-positron emission tomography (PET) was assessed for characterizing and predicting early remodeling in the rat infarct model. Nineteen Wistar rats had surgical occlusion of the left anterior descending coronary artery and 7 were sham-operated. PET was scheduled 48 h and 2 weeks later for quantifying MI area and LV function. Segments with <50% of FDG uptake had histological evidence of MI (74 ± 9% decrease in parietal thickness, fibrosis development). At 48 h, MI area was large (>35% of LV) in 6 rats, moderate (15-35% of LV) in 8 rats, limited (<15% of LV) in 5 rats and absent in the 7 sham rats. LV remodeling, assessed through the 2 weeks increase in end-diastolic volume, increased between rats with limited, moderate and large MI (+72 ± 25, +109 ± 56, +190 ± 69 µl, respectively, P = 0.007). This 3-groups classification allowed predicting 44% of the 2 weeks increase in end-diastolic volume, and additional 34% were predicted by heart rate at 48 h. The acipimox-enhanced FDG gated-PET technique provides efficient characterization and prediction of early remodeling in the rat infarct model.


Subject(s)
Fluorodeoxyglucose F18 , Hypolipidemic Agents , Myocardial Infarction/diagnostic imaging , Myocardium/pathology , Positron-Emission Tomography , Pyrazines , Radiopharmaceuticals , Ventricular Remodeling , Animals , Diastole , Disease Models, Animal , Male , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Predictive Value of Tests , Rats , Rats, Wistar , Time Factors , Ventricular Function, Left
16.
BMC Med Genomics ; 4: 83, 2011 Dec 05.
Article in English | MEDLINE | ID: mdl-22136666

ABSTRACT

BACKGROUND: Prediction of left ventricular (LV) remodeling after acute myocardial infarction (MI) is clinically important and would benefit from the discovery of new biomarkers. METHODS: Blood samples were obtained upon admission in patients with acute ST-elevation MI who underwent primary percutaneous coronary intervention. Messenger RNA was extracted from whole blood cells. LV function was evaluated by echocardiography at 4-months. RESULTS: In a test cohort of 32 MI patients, integrated analysis of microarrays with a network of protein-protein interactions identified subgroups of genes which predicted LV dysfunction (ejection fraction ≤ 40%) with areas under the receiver operating characteristic curve (AUC) above 0.80. Candidate genes included transforming growth factor beta receptor 1 (TGFBR1). In a validation cohort of 115 MI patients, TGBFR1 was up-regulated in patients with LV dysfunction (P < 0.001) and was associated with LV function at 4-months (P = 0.003). TGFBR1 predicted LV function with an AUC of 0.72, while peak levels of troponin T (TnT) provided an AUC of 0.64. Adding TGFBR1 to the prediction of TnT resulted in a net reclassification index of 8.2%. When added to a mixed clinical model including age, gender and time to reperfusion, TGFBR1 reclassified 17.7% of misclassified patients. TGFB1, the ligand of TGFBR1, was also up-regulated in patients with LV dysfunction (P = 0.004), was associated with LV function (P = 0.006), and provided an AUC of 0.66. In the rat MI model induced by permanent coronary ligation, the TGFB1-TGFBR1 axis was activated in the heart and correlated with the extent of remodeling at 2 months. CONCLUSIONS: We identified TGFBR1 as a new candidate prognostic biomarker after acute MI.


Subject(s)
Myocardial Infarction/diagnosis , Myocardial Infarction/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Acute Disease , Adult , Aged , Aged, 80 and over , Angiogenic Proteins/genetics , Biomarkers/metabolism , Blood Cells/metabolism , Female , Gene Expression Profiling , Humans , Male , Middle Aged , Multigene Family/genetics , Myocardial Infarction/blood , Myocardial Infarction/physiopathology , Myocardium/metabolism , Prognosis , Protein Interaction Maps , Protein Serine-Threonine Kinases/genetics , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/genetics , Reproducibility of Results , Transforming Growth Factor beta1/metabolism , Ventricular Remodeling
SELECTION OF CITATIONS
SEARCH DETAIL