Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Blood ; 118(26): e192-208, 2011 Dec 22.
Article in English | MEDLINE | ID: mdl-22012065

ABSTRACT

Macrophages are either classically (M1) or alternatively-activated (M2). Whereas this nomenclature was generated from monocyte-derived macrophages treated in vitro with defined cytokine stimuli, the phenotype of in vivo-derived macrophages is less understood. We completed Affymetrix-based transcriptomic analysis of macrophages from the resolution phase of a zymosan-induced peritonitis. Compared with macrophages from hyperinflamed mice possessing a pro-inflammatory nature as well as naive macrophages from the uninflamed peritoneum, resolution-phase macrophages (rM) are similar to monocyte-derived dendritic cells (DCs), being CD209a positive but lacking CD11c. They are enriched for antigen processing/presentation (MHC class II [H2-Eb1, H2-Ab1, H2-Ob, H2-Aa], CD74, CD86), secrete T- and B-lymphocyte chemokines (Xcl1, Ccl5, Cxcl13) as well as factors that enhance macrophage/DC development, and promote DC/T cell synapse formation (Clec2i, Tnfsf4, Clcf1). rM are also enriched for cell cycle/proliferation genes as well as Alox15, Timd4, and Tgfb2, key systems in the termination of leukocyte trafficking and clearance of inflammatory cells. Finally, comparison with in vitro-derived M1/M2 shows that rM are neither classically nor alternatively activated but possess aspects of both definitions consistent with an immune regulatory phenotype. We propose that macrophages in situ cannot be rigidly categorized as they can express many shades of the inflammatory spectrum determined by tissue, stimulus, and phase of inflammation.


Subject(s)
Macrophages/immunology , Macrophages/metabolism , Transcriptome , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cells, Cultured , Female , Flow Cytometry , Gene Expression Profiling , Male , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Peritonitis/chemically induced , Peritonitis/genetics , Peritonitis/immunology , Reverse Transcriptase Polymerase Chain Reaction , Zymosan
2.
Blood ; 118(22): 5918-27, 2011 Nov 24.
Article in English | MEDLINE | ID: mdl-21911834

ABSTRACT

Females are protected against mortality arising from severe sepsis; however, the precise mechanisms that confer this survival advantage in females over males are unclear. Resident leukocytes in resting tissues have a significant influence on circulating cytokine levels and recruitment of blood leukocytes during acute inflammatory responses. Whether the phenotype of resident leukocytes is distinct in females is unknown. In the present study, we show that the numbers of leukocytes occupying the naive peritoneal and pleural cavities is higher in female than in male mice and rats, comprising more T and B lymphocytes and macrophages. The altered immune cell composition of the female peritoneum is controlled by elevated tissue chemokine expression. Female resident macrophages also exhibit greater TLR expression and enhanced phagocytosis and NADPH oxidase-mediated bacterial killing. However, macrophage-derived cytokine production is diminished by proportionally more resident immunomodulatory CD4+ T lymphocytes. Ovarian hormones regulate macrophage phenotype, function, and numbers, but have no significant impact on T-lymphocyte populations in females. We have identified a fundamental sex difference in phenotype of resident leukocytes. We propose that the distinct resident leukocyte population in females allows aggressive recognition and elimination of diverse infectious stimuli without recruitment of circulating neutrophils or excessive cytokine production.


Subject(s)
Inflammation/immunology , Leukocytes/immunology , Sex Characteristics , Acute Disease , Animals , Efficiency , Female , Inflammation/metabolism , Inflammation/pathology , Leukocyte Count , Leukocytes/cytology , Leukocytes/metabolism , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/pathology , Male , Mice , Mice, Inbred C57BL , Peritonitis/immunology , Peritonitis/mortality , Peritonitis/pathology , Phenotype , Pleurisy/immunology , Pleurisy/mortality , Pleurisy/pathology , Rats , Rats, Wistar
3.
Prog Lipid Res ; 50(1): 35-51, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20655950

ABSTRACT

Originally regarded as just membrane constituents and energy storing molecules, lipids are now recognised as potent signalling molecules that regulate a multitude of cellular responses via receptor-mediated pathways, including cell growth and death, and inflammation/infection. Derived from polyunsaturated fatty acids (PUFAs), such as arachidonic acid (AA), eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA), each lipid displays unique properties, thus making their role in inflammation distinct from that of other lipids derived from the same PUFA. The diversity of their actions arises because such metabolites are synthesised via discrete enzymatic pathways and because they elicit their response via different receptors. This review will collate the bioactive lipid research to date and summarise the findings in terms of the major pathways involved in their biosynthesis and their role in inflammation and its resolution. It will include lipids derived from AA (prostanoids, leukotrienes, 5-oxo-6,8,11,14-eicosatetraenoic acid, lipoxins and epoxyeicosatrienoic acids), EPA (E-series resolvins), and DHA (D-series resolvins, protectins and maresins).


Subject(s)
Inflammation/metabolism , Lipid Metabolism , Animals , Humans , Inflammation/enzymology , Lipids/biosynthesis , Lipids/chemistry
4.
Blood ; 116(16): 2950-9, 2010 Oct 21.
Article in English | MEDLINE | ID: mdl-20606163

ABSTRACT

Inhibition of cyclooxygenase (COX)-derived prostaglandins (PGs) by nonsteroidal anti-inflammatory drugs (NSAIDs) mediates leukocyte killing of bacteria. However, the relative contribution of COX1 versus COX2 to this process, as well as the mechanisms controlling it in mouse and humans, are unknown. Indeed, the potential of NSAIDs to facilitate leukocyte killing of drug-resistant bacteria warrants investigation. Therefore, we carried out a series of experiments in mice and humans, finding that COX1 is the predominant isoform active in PG synthesis during infection and that its prophylactic or therapeutic inhibition primes leukocytes to kill bacteria by increasing phagocytic uptake and reactive oxygen intermediate-mediated killing in a cyclic adenosine monophosphate (cAMP)-dependent manner. Moreover, NSAIDs enhance bacterial killing in humans, exerting an additive effect when used in combination with antibiotics. Finally, NSAIDs, through the inhibition of COX prime the innate immune system to mediate bacterial clearance of penicillin-resistant Streptococcus pneumoniae serotype 19A, a well-recognized vaccine escape serotype of particular concern given its increasing prevalence and multi-antibiotic resistance. Therefore, these data underline the importance of lipid mediators in host responses to infection and the potential of inhibitors of PG signaling pathways as adjunctive therapies, particularly in the con-text of antibiotic resistance.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cyclooxygenase 1/immunology , Cyclooxygenase Inhibitors/pharmacology , Drug Resistance, Bacterial/drug effects , Immunity, Innate/drug effects , Pneumococcal Infections/drug therapy , Streptococcus pneumoniae/drug effects , Adult , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Cyclic AMP/immunology , Cyclic AMP/metabolism , Cyclooxygenase 2/immunology , Cyclooxygenase Inhibitors/therapeutic use , Cytokines/immunology , Humans , Male , Mice , Mice, Inbred C57BL , Phagocytosis/drug effects , Prostaglandins/immunology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...