Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Cardiovasc Ultrasound ; 13: 1, 2015 Jan 08.
Article in English | MEDLINE | ID: mdl-25567641

ABSTRACT

BACKGROUND: Macrophages may concentrate ultrasound contrast agents and exhibit selective adhesion to activated endothelium. The present study investigates in mice the potential of perfluorohexane (PFH) loaded macrophages to act as ultrasound contrast agent with high reflectivity and specifically targeted at (atherosclerotic) vascular lesions. METHODS: Lung passage was evaluated with a mouse echo scanner after injection, at a slow pace or as a bolus, of varying doses of PFH-loaded and unloaded bone marrow macrophages (BMM) into the jugular vein. The interaction of PFH-loaded and unloaded BMM with TNF-α stimulated carotid artery endothelium after tail vein injection was assessed by means of intravital microscopy. RESULTS: High doses of jugular vein injected PFH-loaded BMM were visible with ultrasound in the pulmonary artery and detectable in the carotid artery. At intravital microscopy, tail vein injected BMM exhibited rolling and adhesion behavior at the TNF-α stimulated carotid endothelium, similar to that of native blood leukocytes. Rolling behavior was not different between PFH-loaded and unloaded BMM (p = 0.38). CONCLUSION: In vivo, perfluorohexane loaded macrophages pass the pulmonary circulation and appear on the arterial side. Moreover, they roll and adhere selectively to activated endothelium under physiological flow conditions. These findings indicate that perfluorohexane loaded BMM could be used to study processes in vivo where endothelial activation plays a role, such as atherosclerosis.


Subject(s)
Endothelium, Vascular/diagnostic imaging , Fluorocarbons/administration & dosage , Leukocytes/physiology , Macrophages/physiology , Pulmonary Artery/diagnostic imaging , Animals , Cell Adhesion/physiology , Cell Communication/physiology , Contrast Media , Drug Carriers , Female , Injections, Intravenous , Male , Mice , Mice, Inbred C57BL , Ultrasonography
2.
PLoS One ; 6(7): e22327, 2011.
Article in English | MEDLINE | ID: mdl-21814576

ABSTRACT

Activation of the transcription factor NF-κB appears to be involved in different stages of atherogenesis. In this paper we investigate the role of NF-κB inhibitor IκBα in atherosclerosis. Myeloid-specific deletion of IκBα results in larger and more advanced lesions in LDL-R-deficient mice without affecting the compositional phenotype of the plaques or systemic inflammatory markers in the plasma. We show that IκBα-deleted macrophages display enhanced adhesion to an in vitro endothelial cell layer, coinciding with an increased expression of the chemokine CCL5. Also, in vivo we found that IκBα(del) mice had more leukocytes adhering to the luminal side of the endothelial cell layers that cover the atherosclerotic plaques. Moreover, we introduce ER-MP58 in this paper as a new immunohistochemical tool for quantifying newly recruited myeloid cells in the atherosclerotic lesion. This staining confirms that in IκBα(del) mice more leukocytes are attracted to the plaques. In conclusion, we show that IκBα deletion in myeloid cells promotes atherogenesis, probably through an induced leukocyte recruitment to plaques.


Subject(s)
Atherosclerosis/etiology , Atherosclerosis/pathology , I-kappa B Proteins/physiology , Leukocytes/pathology , Myeloid Cells/pathology , Receptors, LDL/physiology , Animals , Apolipoproteins E/metabolism , Atherosclerosis/metabolism , Blotting, Western , Bone Marrow Transplantation , Cell Adhesion , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Female , Immunoenzyme Techniques , Inflammation/metabolism , Inflammation/pathology , Leukocytes/metabolism , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/metabolism , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism
3.
Mol Imaging Biol ; 10(5): 264-70, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18536974

ABSTRACT

PURPOSE: We investigated in vitro the potential of macrophages to act as targeted vehicle for ultrasound molecular imaging. PROCEDURES: Murine bone marrow-derived macrophages (BMM), incubated for 3 h with different concentrations of perfluorohexane (PFH) emulsions, were monitored by microscopy, flow cytometry, and ultrasound. Effects of PFH loading on BMM adhesion molecule (PSGL-1, VLA-4, Mac-1, LFA-1) expression were analyzed by flow cytometry. Static adhesion of PFH loaded BMM to unstimulated and TNF-alpha stimulated b.End5 endothelial cells was assessed by microscopy. RESULTS: Incubation of BMM with PFH emulsions resulted in dose-dependent uptake and increased echogenicity (max. 17 dB). Flow cytometry analyses revealed no down-regulation related to PFH loading of BMM adhesion molecule expression. Endothelial adhesion remained functional, even after 24 h, although PFH loading dose-dependently attenuated static adhesion. CONCLUSION: PFH loaded BMM may potentially serve as ultrasound contrast agent for noninvasive detection of atherogenic hotspots in arteries.


Subject(s)
Contrast Media/metabolism , Fluorocarbons/metabolism , Macrophages/diagnostic imaging , Macrophages/metabolism , Animals , Bone Marrow/metabolism , Cell Adhesion , Cell Adhesion Molecules/metabolism , Cells, Cultured , Feasibility Studies , Macrophages/cytology , Mice , Mice, Inbred C57BL , Ultrasonography
4.
J Biol Chem ; 283(31): 21640-8, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18511424

ABSTRACT

Secreted phospholipase A2 group X (sPLA(2)-X) is one of the most potent enzymes of the phospholipase A(2) lipolytic enzyme superfamily. Its high catalytic activity toward phosphatidylcholine (PC), the major phospholipid of cell membranes and low-density lipoproteins (LDL), has implicated sPLA(2)-X in chronic inflammatory conditions such as atherogenesis. We studied the role of sPLA(2)-X enzyme activity in vitro and in vivo, by generating sPLA(2)-X-overexpressing macrophages and transgenic macrophage-specific sPLA(2)-X mice. Our results show that sPLA(2)-X expression inhibits macrophage activation and inflammatory responses upon stimulation, characterized by reduced cell adhesion and nitric oxide production, a decrease in tumor necrosis factor (TNF), and an increase in interleukin (IL)-10. These effects were mediated by an increase in IL-6, and enhanced production of prostaglandin E(2) (PGE(2)) and 15-deoxy-Delta12,14-prostaglandin J(2) (PGJ(2)). Moreover, we found that overexpression of active sPLA(2)-X in macrophages strongly increases foam cell formation upon incubation with native LDL but also oxidized LDL (oxLDL), which is mediated by enhanced expression of scavenger receptor CD36. Transgenic sPLA(2)-X mice died neonatally because of severe lung pathology characterized by interstitial pneumonia with massive granulocyte and surfactant-laden macrophage infiltration. We conclude that overexpression of the active sPLA(2)-X enzyme results in enhanced foam cell formation but reduced activation and inflammatory responses in macrophages in vitro. Interestingly, enhanced sPLA(2)-X activity in macrophages in vivo leads to fatal pulmonary defects, suggesting a crucial role for sPLA(2)-X in inflammatory lung disease.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Group X Phospholipases A2/physiology , Lipids/chemistry , Lung/pathology , Macrophages/metabolism , Animals , Cell Line , Cells, Cultured , Group X Phospholipases A2/metabolism , Humans , Interleukin-10/metabolism , Lipopolysaccharides/metabolism , Lung/abnormalities , Mice , Mice, Transgenic , Models, Biological , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/metabolism
5.
Mutat Res ; 621(1-2): 87-94, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17418875

ABSTRACT

Increased oxidative stress and subsequent lipid peroxidation (LPO) are thought to be critical events in the formation of atherosclerotic lesions in apolipoprotein E deficient mice (ApoE-KO). LPO derived reactive aldehydes react with DNA to form exocyclic etheno-DNA adducts. These pro-mutagenic DNA lesions are known to be involved in the initiation of carcinogenesis, but their role in the development of atherosclerosis is unknown. In the present study we show that levels of the LPO derived 1,N(6)-ethenodeoxyadenosine (varepsilondA) and 3,N(4)-ethenodeoxycytidine (varepsilondC) were both significantly lower in aorta of 12 weeks old ApoE-KO mice as compared to their wild type controls (1.6+/-0.3 versus 3.2+/-0.8 varepsilondA per 10(8) parent nucleotides, P=0.04 and 4.8+/-0.8 versus 9.2+/-2.1 for varepsilondC, P=0.02). Moreover, levels of both DNA adduct types were inversely related with total plasma cholesterol levels. Consequently, lowest etheno-DNA adduct levels were observed in ApoE-KO mice on a high fat diet. Hypercholesterolemia has previously been associated with increased expression of base excision repair (BER) enzymes, which could explain the lower levels of etheno-DNA adducts in ApoE-KO mice as compared to wild type controls. Indeed, increased staining for the BER-specific DNA repair enzyme apurinic/apyrimidinic endonuclease (Ape1/Ref1) was observed by immunohistochemistry in the endothelium and the first layers of arterial smooth muscle cells of ApoE-KO mice as compared to their wild type counterparts. A high fat diet further increased overall Ape1/Ref1 protein expression in ApoE-KO mice. Although these data suggest no role for increased LPO derived DNA damage in the onset of atherogenesis in ApoE-KO mice, the potentially modulating role of Ape1/Ref1 in the arterial wall deserves further attention.


Subject(s)
Apolipoproteins E/deficiency , Atherosclerosis/metabolism , DNA Damage , Lipid Peroxidation/genetics , Animals , Apolipoproteins E/genetics , Atherosclerosis/genetics , DNA Adducts/metabolism , DNA Repair Enzymes/metabolism , Female , Immunohistochemistry , Lipids/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress/genetics
6.
Curr Opin Lipidol ; 16(5): 536-42, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16148538

ABSTRACT

PURPOSE OF REVIEW: Atherosclerosis is a chronic inflammatory disease of the medium and large-sized arteries. Nuclear factor kappaB transcription factors are major regulators of inflammatory responses, and aberrant nuclear factor kappaB regulation is linked to a large number of diseases. Focusing on macrophages, this review will discuss recent literature on the role of nuclear factor kappaB and the signaling pathways regulating its activity in atherosclerosis. RECENT FINDINGS: After the initial identification of activated nuclear factor kappaB in human atherosclerotic lesions, the involvement of this family of transcription factors in atherogenesis has gained growing attention. It is now clear that signaling pathways activating nuclear factor kappaB, and nuclear factor kappaB action, constitute major players at all stages of the atherosclerotic process. Long considered a pro-atherogenic factor, recent studies indicate that the actual role of nuclear factor kappaB might prove to be far more complex. Apart from activating many pro-inflammatory genes linked to atherogenesis, nuclear factor kappaB regulates cellular processes such as cell survival and proliferation. In addition, its important role in inflammatory resolution and anti-inflammatory gene transcription suggests that its activation at different cell types or different stages of the atherosclerotic process might have distinct and opposing results. SUMMARY: The numerous diseases in which aberrant nuclear factor kappaB action is found to play a crucial role makes it an intensively studied target for drug interventions. However, given its pleiotropic functions in inflammation and immunity, a more targeted modulation of its activity, at a cell type-specific or disease stage-specific level, could provide safer therapeutic solutions.


Subject(s)
Atherosclerosis/metabolism , Inflammation Mediators/physiology , Macrophages/metabolism , Macrophages/pathology , NF-kappa B/physiology , Signal Transduction , Animals , Atherosclerosis/immunology , Atherosclerosis/pathology , Disease Models, Animal , Humans , Macrophages/immunology , Signal Transduction/immunology
7.
FASEB J ; 19(10): 1290-2, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15939734

ABSTRACT

Although it has been demonstrated that carcinogenic environmental polycyclic aromatic hydrocarbons (PAHs) cause progression of atherosclerosis, the underlying mechanism remains unclear. In the present study, we aimed to investigate whether DNA binding events are critically involved in the progression of PAH-mediated atherogenesis. Apolipoprotein E knockout mice were orally (24 wk, once/wk) exposed to 5 mg/kg benzo[a]pyrene (B[a]P), or its nonmutagenic, noncarcinogenic structural isoform benzo[e]pyrene (B[e]P). 32P-postlabeling of lung tissue confirmed the presence of promutagenic PAH-DNA adducts in B[a]P-exposed animals, whereas in B[e]P-exposed and vehicle control animals, these adducts were undetectable. Morphometrical analysis showed that both B[a]P and B[e]P caused an increase in plaque size, whereas location or number of plaques was unaffected. Immunohistochemistry revealed no differences in oxidative DNA damage (8-OHdG) or apoptosis in the plaques. Also plasma lipoprotein levels remained unchanged after PAH-exposure. However, T lymphocytes were increased > or =2-fold in the plaques of B[a]P- and B[e]P-exposed animals. Additionally, B[a]P and to a lesser extent B[e]P exposure resulted in increased TGFbeta protein levels in the plaques, that was mainly localized in the plaque macrophages. In vitro studies using the murine macrophage like RAW264.7 cells showed that inhibition of TGFbeta resulted in decreased tumor necrosis factor (TNF) alpha release, suggesting that enhanced TGFbeta expression in the plaque macrophages contributes to the proinflammatory effects in the vessel wall. In general, this inflammatory reaction in the plaques appeared to be a local response since peripheral blood cell composition (T cells, B cells, granulocytes, and macrophages) was not changed upon PAH exposure. In conclusion, we showed that both B[a]P and B[e]P cause progression of atherosclerosis, irrespective of their DNA binding properties. Moreover, our data revealed a possible novel mechanism of PAH-mediated atherogenesis, which likely involves a TGFbeta-mediated local inflammatory reaction in the vessel wall.


Subject(s)
Atherosclerosis/chemically induced , Benzo(a)pyrene/toxicity , Benzopyrenes/toxicity , DNA Adducts/metabolism , DNA/metabolism , Animals , Apolipoproteins E/genetics , Apolipoproteins E/physiology , Benzo(a)pyrene/metabolism , Benzopyrenes/metabolism , Cells, Cultured , Flow Cytometry , Male , Mice , Mice, Knockout , Phenotype , Transforming Growth Factor beta/analysis , Transforming Growth Factor beta/physiology , Transforming Growth Factor beta1
8.
Am J Pathol ; 164(1): 101-8, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14695324

ABSTRACT

Benzo[a]pyrene (B[a]P) is a polycyclic aromatic hydrocarbon with atherogenic and carcinogenic properties. The role of B[a]P in carcinogenesis is well established, and thought to exert via enzymatic activation into reactive metabolites that are capable of binding to the DNA leading to uncontrolled proliferation. However, the mechanism underlying the atherogenic properties of B[a]P is still unclear. Therefore, the effects of chronic B[a]P exposure on atherosclerotic plaque development in apolipoprotein E knockout (apoE-KO) mice were studied. ApoE-KO mice were orally treated with 5 mg/kg/bw B[a]P once per week for 12 or 24 consecutive weeks. Levels of reactive B[a]P metabolites in the arterial tree (from the aortic arch until the iliac artery bifurcations) were high as shown by the level of B[a]P DNA-binding products measured in DNA isolated from the entire aorta (38.9 +/- 4.8 adducts/10(8) nucleotides). Analysis of atherosclerotic lesions in the aortic arch showed no influence of B[a]P on location or number of lesions. Moreover, no increased levels of p53 nuclear protein accumulation or cell proliferation, as detected by immunohistochemistry, were seen in the plaques of the B[a]P-exposed animals. However, the effects of B[a]P on advanced lesions were obvious: advanced plaques were larger and more prone to lipid core development and plaque layering at both 12 and 24 weeks (P < 0.05). In the B[a]P-exposed animals advanced plaques contained more T-lymphocytes and macrophages than in the control animals at both end points (P < 0.05). These data suggest that B[a]P does not initiate atherosclerosis in apoE-KO mice, but accelerates the progression of atherosclerotic plaques via a local inflammatory response.


Subject(s)
Apolipoproteins E/genetics , Arteriosclerosis/chemically induced , Arteriosclerosis/pathology , Benzo(a)pyrene/toxicity , Animals , Apoptosis/drug effects , Benzo(a)pyrene/analysis , Benzo(a)pyrene/metabolism , DNA Adducts/analysis , DNA Adducts/drug effects , Immunohistochemistry , Male , Mice , Mice, Knockout , Phenotype , Time Factors , Tumor Suppressor Protein p53/drug effects
9.
Environ Mol Mutagen ; 42(4): 243-9, 2003.
Article in English | MEDLINE | ID: mdl-14673869

ABSTRACT

The role of plasma lipids in the uptake, transportation, and distribution of lipophilic carcinogens like benzo[a]pyrene (B[a]P) remains unclear. Therefore, we studied the effects of dietary-modulated plasma lipids on B[a]P-induced DNA damage in several organs of two hyperlipidemic mouse models. Male apolipoprotein E (ApoE)*3-Leiden (n = 22) and ApoE knockout (ApoE-KO) mice (n = 20) were fed a high-fat cholesterol (HFC) diet or low-fat cholesterol (LFC; standard mouse chow) diet for 3 weeks, after which the animals were exposed to a single oral dose of 5 mg/kg bw B[a]P or vehicle and killed 4 days later. Plasma lipids were determined and DNA adducts were measured in aorta, heart, lung, liver, brain, and stomach. Total cholesterol and low-density lipoprotein (LDL) cholesterol were increased in all animals on a HFC diet, whereas a decrease of triglycerides was seen only in the ApoE-KO mice. In ApoE-KO mice on a normal diet, DNA-adduct levels were highest in aorta (10.8 +/- 1.4 adducts/10(8) nucleotides), followed by brain (7.8 +/- 1.3), lung (3.3 +/- 0.7), heart (3.1 +/- 0.6), liver (1.5 +/- 0.2) and stomach (1.2 +/- 0.2). In the ApoE*3-Leiden mice, adduct levels were equally high in aorta, heart, and lung (4.6 +/- 0.7, 5.0 +/- 0.5 and 4.6 +/- 0.4, respectively), followed by stomach (2.7 +/- 0.4), brain (2.3 +/- 0.2), and liver (1.7 +/- 0.2). In the ApoE-KO mice, the HFC diet intervention resulted in lower adduct levels in lung (2.1 +/- 0.2), heart (1.9 +/- 0.2), and brain (2.9 +/- 0.5), as compared with the LFC group. In contrast, a nonsignificant increase of adducts was found in aorta (13.1 +/- 1.5). A similar but nonsignificant trend was observed in the ApoE*3-Leiden mice. Multiple regression analysis showed that in aorta, DNA adducts were inversely related to plasma triglycerides (P = 0.004) and were also modulated by the ApoE genotype (P < 0.001). The results of the present study support further investigation into the role of dietary modulation of plasma lipids, ApoE, and polycyclic aromatic hydrocarbon exposure on the formation of DNA adducts in chronic degenerative diseases.


Subject(s)
Benzo(a)pyrene/pharmacokinetics , Carcinogens/pharmacokinetics , DNA Damage , Hyperlipidemias/chemically induced , Lipids/blood , Animals , Apolipoprotein E3 , Apolipoproteins E/genetics , DNA/metabolism , DNA Adducts/metabolism , Disease Models, Animal , Male , Mice , Mice, Knockout , Pyrenes/metabolism , Regression Analysis , Time Factors , Tissue Distribution , Triglycerides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...