Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Shock ; 36(2): 162-9, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21430603

ABSTRACT

Burn injury initiates an enhanced inflammatory condition referred to as the systemic inflammatory response syndrome or the two-hit response phenotype. Prior reports indicated that macrophages respond to injury and demonstrate a heightened reactivity to Toll-like receptor stimulation. Since we and others observed a significant increase in splenic GR-1 F4/80 CD11b macrophages in burn-injured mice, we wished to test if these macrophages might be the primary macrophage subset that shows heightened LPS reactivity. We report here that burn injury promoted higher level TNF-α expression in GR-1, but not GR-1 macrophages, after LPS activation both in vivo and ex vivo. We next tested whether CD4 T cells, which are known to suppress injury-induced inflammatory responses, might control the activation and expansion of GR-1 macrophages. Interestingly, we found that GR-1 macrophage expansion and LPS-induced TNF-α expression were not significantly different between wild-type and CD4 T cell-deficient CD4(-/-) mice. However, further investigations showed that LPS-induced TNF-α production was significantly influenced by CD4 T cells. Taken together, these data indicate that GR-1 F4/80 CD11b macrophages represent the primary macrophage subset that expands in response to burn injury and that CD4 T cells do not influence the GR-1 macrophage expansion process, but do suppress LPS-induced TNF-α production. These data suggest that modulating GR-1 macrophage activation as well as CD4 T cell responses after severe injury may help control the development of systemic inflammatory response syndrome and the two-hit response phenotype.


Subject(s)
Burns/immunology , Burns/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Macrophages/immunology , Macrophages/metabolism , Receptors, Cell Surface/metabolism , Animals , CD11b Antigen/metabolism , Flow Cytometry , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL , Tumor Necrosis Factor-alpha/metabolism
2.
J Leukoc Biol ; 89(1): 137-47, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20884652

ABSTRACT

Injury initiates local and systemic host responses and is known to increase CD4 Treg activity in mice and humans. This study uses a TCR transgenic T cell adoptive transfer approach and in vivo Treg depletion to determine specifically the in vivo influence of Tregs on antigen-driven CD4 T cell reactivity following burn injury in mice. We report here that injury in the absence of recipient and donor Tregs promotes high antigen-driven CD4 T cell expansion and increases the level of CD4 T cell reactivity. In contrast, CD4 T cell expansion and reactivity were suppressed significantly in injured Treg-replete mice. In additional experiments, we found that APCs prepared from burn- or sham-injured, Treg-depleted mice displayed significantly higher antigen-presenting activity than APCs prepared from normal mice, suggesting that Tregs may suppress injury responses by controlling the intensity of APC activity. Taken together, these findings demonstrate that Tregs can actively control the in vivo expansion and reactivity of antigen-stimulated, naïve CD4 T cells following severe injury.


Subject(s)
Burns/immunology , CD4 Antigens/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Antigen-Presenting Cells/immunology , B7-1 Antigen/metabolism , B7-2 Antigen/metabolism , Burns/pathology , Cell Proliferation , Dendritic Cells/metabolism , Epitopes/immunology , Lymph Nodes/immunology , Lymphocyte Activation/immunology , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Transgenic , Models, Immunological , T-Lymphocytes, Regulatory/cytology , Th1 Cells/immunology
3.
J Leukoc Biol ; 85(5): 862-70, 2009 May.
Article in English | MEDLINE | ID: mdl-19228816

ABSTRACT

Severe injury disrupts normal immune regulation causing a transient hyperinflammatory reaction and suppressed adaptive immune function. This report addresses the potential contribution of dendritic cells (DC) to changes in adaptive immune function after injury by specifically measuring injury-induced changes in splenic DC numbers and subsets, cell-surface markers, TLR responses, and APC function. Using a mouse burn injury model, we found that injury did not markedly alter the relative percentage of lymphoid, myeloid, or plasmacytoid DC in the spleens of burn-injured mice. Moreover, we did not observe a significant reduction in cell-surface expression of several major costimulatory molecules, CD40, CD80, CD86, programmed death 1 ligand, ICOS ligand, and B7-H3, on DC. Instead, we observed increased cell-surface expression of CD86 at 1 day after injury with no significant changes in costimulatory molecule expression at 7 days after injury, suggesting that burn injury causes an early activation of DC. In addition, injury did not suppress DC reactivity to TLR2, TLR4, or TLR9 agonists. Most important, DC prepared from injured mice were able to present peptide antigen to naive OTII TCR transgenic CD4+ T cells as efficiently and effectively as DC from sham-injured mice. We also found that CD4 T cells stimulated with antigen presented by DC from sham or burn mice showed similar levels of IL-2, IFN-gamma, IL-10, and IL-13 production. Taken together, these findings support the conclusion that DC do not acquire a suppressive phenotype following severe injury in mice.


Subject(s)
Antigen Presentation/immunology , Burns/immunology , Dendritic Cells/immunology , Spleen/cytology , Animals , B7-2 Antigen/immunology , B7-2 Antigen/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cytokines/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Transgenic , Spleen/immunology , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Toll-Like Receptors/immunology
4.
J Immunol ; 180(4): 2450-8, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18250454

ABSTRACT

Major injury is widely thought to predispose the injured host to opportunistic infections. This idea is supported by animal studies showing that major injury causes reduced resistance to polymicrobial sepsis induced by cecal ligation and puncture. Although cecal ligation and puncture represents a clinically relevant sepsis model, we wanted to test whether injury might also lead to greater susceptibility to peritoneal infection caused by a single common pathogen, Escherichia coli. Contrary to our expectation, we show herein that the LD(50) for sham-injured mice was 10(3) CFU of E. coli, whereas the LD(50) for burn-injured mice was 50 x 10(3) CFU at 7 days postinjury. This injury-associated enhanced resistance was apparent as early as 1 day after injury, and maximal resistance was observed at days 7 and 14. We found that burn-injured mice had higher numbers of circulating neutrophils and monocytes than did sham mice before infection and that injured mice were able to recruit greater numbers of neutrophils to the site of infection. Moreover, the peritoneal neutrophils in burn-injured mice were more highly activated than neutrophils from sham mice as determined by Mac-1 expression, superoxide generation, and bactericidal activity. Our findings suggest that the enhanced innate immune response that develops following injury, although it is commonly accepted as the mediator of the detrimental systemic inflammatory response syndrome, may also, in some cases, benefit the injured host by boosting innate immune antimicrobial defenses.


Subject(s)
Burns/immunology , Burns/microbiology , Escherichia coli Infections/immunology , Escherichia coli Infections/prevention & control , Immunity, Innate , Animals , Ascitic Fluid/immunology , Ascitic Fluid/microbiology , Burns/mortality , Escherichia coli Infections/mortality , Injections, Intraperitoneal , Lethal Dose 50 , Macrophage Activation/immunology , Male , Mice , Mice, Inbred C57BL , Neutrophil Activation/immunology , Opportunistic Infections/immunology , Opportunistic Infections/mortality , Opportunistic Infections/prevention & control
5.
Physiol Genomics ; 32(3): 299-310, 2008 Feb 19.
Article in English | MEDLINE | ID: mdl-17986522

ABSTRACT

A primary objective of the large collaborative project entitled "Inflammation and the Host Response to Injury" was to identify leukocyte genes that are differentially expressed after two different types of injury in mouse models and to test the hypothesis that both forms of injury would induce similar changes in gene expression. We report here the genes that are expressed in white blood cells (WBCs) and in splenocytes at 2 h, 1 day, 3 days, and 7 days after burn and sham injury or trauma-hemorrhage (T-H) and sham T-H. Affymetrix Mouse Genome 430 2.0 GeneChips were used to profile gene expression, and the results were analyzed by dCHIP, BRB Array Tools, and Ingenuity Pathway Analysis (IPA) software. We found that the highest number of genes differentially expressed following burn injury were at day 1 for both WBCs (4,989) and for splenocytes (4,715) and at day 1 for WBCs (1,167) and at day 3 for splenocytes (1,117) following T-H. The maximum overlap of genes that were expressed after both forms of injury were at day 1 in WBCs (136 genes) and at day 7 in splenocytes (433 genes). IPA revealed that the cell-to-cell signaling, cell death, immune response, antiapoptosis, and cell cycle control pathways were affected most significantly. In summary, this report provides a database of genes that are modulated in WBCs and splenocytes at sequential time points after burn or T-H in mice and reveals that relatively few leukocyte genes are expressed in common after these two forms of injury.


Subject(s)
Burns/genetics , Gene Expression Regulation , Gene Regulatory Networks/genetics , Hemorrhage/genetics , Leukocytes/metabolism , Animals , Blood Loss, Surgical , Gene Expression Profiling , Inflammation/genetics , Male , Mice , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Spleen/pathology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...