Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 7(3): e32440, 2012.
Article in English | MEDLINE | ID: mdl-22403661

ABSTRACT

OBJECTIVE: A genomic region near the CDKN2A locus, encoding p16(INK4a), has been associated to type 2 diabetes and atherosclerotic vascular disease, conditions in which inflammation plays an important role. Recently, we found that deficiency of p16(INK4a) results in decreased inflammatory signaling in murine macrophages and that p16(INK4a) influences the phenotype of human adipose tissue macrophages. Therefore, we investigated the influence of immune cell p16(INK4a) on glucose tolerance and atherosclerosis in mice. METHODS AND RESULTS: Bone marrow p16(INK4a)-deficiency in C57Bl6 mice did not influence high fat diet-induced obesity nor plasma glucose and lipid levels. Glucose tolerance tests showed no alterations in high fat diet-induced glucose intolerance. While bone marrow p16(INK4a)-deficiency did not affect the gene expression profile of adipose tissue, hepatic expression of the alternative markers Chi3l3, Mgl2 and IL10 was increased and the induction of pro-inflammatory Nos2 was restrained on the high fat diet. Bone marrow p16(INK4a)-deficiency in low density lipoprotein receptor-deficient mice did not affect western diet-induced atherosclerotic plaque size or morphology. In line, plasma lipid levels remained unaffected and p16(INK4a)-deficient macrophages displayed equal cholesterol uptake and efflux compared to wild type macrophages. CONCLUSION: Bone marrow p16(INK4a)-deficiency does not affect plasma lipids, obesity, glucose tolerance or atherosclerosis in mice.


Subject(s)
Atherosclerosis/metabolism , Atherosclerosis/pathology , Bone Marrow/metabolism , Cyclin-Dependent Kinase Inhibitor p16/deficiency , Glucose/metabolism , Homeostasis , Obesity/metabolism , Animals , Diet, High-Fat/adverse effects , Glucose Intolerance/chemically induced , Glucose Intolerance/metabolism , Humans , Hyperlipidemias/metabolism , Hyperlipidemias/pathology , Male , Mice , Mice, Inbred C57BL , Obesity/chemically induced , Receptors, LDL/deficiency
2.
Cell Metab ; 6(2): 137-43, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17681149

ABSTRACT

Th1 cytokines promote monocyte differentiation into proatherogenic M1 macrophages, while Th2 cytokines lead to an "alternative" anti-inflammatory M2 macrophage phenotype. Here we show that in human atherosclerotic lesions, the expression of M2 markers and PPARgamma, a nuclear receptor controlling macrophage inflammation, correlate positively. Moreover, PPARgamma activation primes primary human monocytes into M2 differentiation, resulting in a more pronounced anti-inflammatory activity in M1 macrophages. However, PPARgamma activation does not influence M2 marker expression in resting or M1 macrophages, nor does PPARgamma agonist treatment influence the expression of M2 markers in atherosclerotic lesions, indicating that only native monocytes can be primed by PPARgamma activation to an enhanced M2 phenotype. Furthermore, PPARgamma activation significantly increases expression of the M2 marker MR in circulating peripheral blood mononuclear cells. These data demonstrate that PPARgamma activation skews human monocytes toward an anti-inflammatory M2 phenotype.


Subject(s)
Inflammation/metabolism , Inflammation/pathology , Macrophages/cytology , Macrophages/metabolism , Monocytes/cytology , PPAR gamma/metabolism , Benzophenones/pharmacology , Biomarkers , Blood Cells/drug effects , Carotid Artery Diseases/pathology , Cell Differentiation/drug effects , Cells, Cultured , Foam Cells/drug effects , Foam Cells/pathology , Humans , Macrophages/drug effects , Monocytes/drug effects , Monocytes/metabolism , PPAR gamma/agonists , Paracrine Communication/drug effects , Phenotype , Stem Cells/drug effects , Tyrosine/analogs & derivatives , Tyrosine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...