Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Nat Commun ; 8: 13944, 2017 01 11.
Article in English | MEDLINE | ID: mdl-28074841

ABSTRACT

Bacterial pneumonia is a significant healthcare burden worldwide. Failure to resolve inflammation after infection precipitates lung injury and an increase in morbidity and mortality. Gram-negative bacteria are common in pneumonia and increased levels of the mito-damage-associated molecular pattern (DAMP) cardiolipin can be detected in the lungs. Here we show that mice infected with Klebsiella pneumoniae develop lung injury with accumulation of cardiolipin. Cardiolipin inhibits resolution of inflammation by suppressing production of anti-inflammatory IL-10 by lung CD11b+Ly6GintLy6CloF4/80+ cells. Cardiolipin induces PPARγ SUMOylation, which causes recruitment of a repressive NCOR/HDAC3 complex to the IL-10 promoter, but not the TNF promoter, thereby tipping the balance towards inflammation rather than resolution. Inhibition of HDAC activity by sodium butyrate enhances recruitment of acetylated histone 3 to the IL-10 promoter and increases the concentration of IL-10 in the lungs. These findings identify a mechanism of persistent inflammation during pneumonia and indicate the potential of HDAC inhibition as a therapy.


Subject(s)
Cardiolipins/physiology , Inflammation/metabolism , Interleukin-10/biosynthesis , Klebsiella Infections/physiopathology , Klebsiella pneumoniae/isolation & purification , Pneumonia, Bacterial/metabolism , Animals , Cardiolipins/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Interleukin-10/genetics , Interleukin-10/metabolism , Klebsiella Infections/microbiology , Lipopolysaccharides/administration & dosage , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid-Derived Suppressor Cells/immunology , Oxidation-Reduction , PPAR gamma/agonists , PPAR gamma/metabolism , Pneumonia, Bacterial/drug therapy , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/pathology , Promoter Regions, Genetic , RAW 264.7 Cells , Sumoylation , Tumor Necrosis Factor-alpha/genetics
2.
Cell Rep ; 15(8): 1700-14, 2016 05 24.
Article in English | MEDLINE | ID: mdl-27184852

ABSTRACT

Inhalation of environmental antigens such as allergens does not always induce inflammation in the respiratory tract. While antigen-presenting cells (APCs), including dendritic cells and macrophages, take up inhaled antigens, the cell-intrinsic molecular mechanisms that prevent an inflammatory response during this process, such as activation of the transcription factor NF-κB, are not well understood. Here, we show that the nuclear receptor PPARγ plays a critical role in blocking NF-κB activation in response to inhaled antigens to preserve immune tolerance. Tolerance induction promoted mitochondrial respiration, generation of H2O2, and suppression of NF-κB activation in WT, but not PPARγ-deficient, APCs. Forced restoration of H2O2 in PPARγ-deficient cells suppressed IκBα degradation and NF-κB activation. Conversely, scavenging reactive oxygen species from mitochondria promoted IκBα degradation with loss of regulatory and promotion of inflammatory T cell responses in vivo. Thus, communication between PPARγ and the mitochondria maintains immune quiescence in the airways.


Subject(s)
Antigen-Presenting Cells/immunology , Hydrogen Peroxide/metabolism , Lung/cytology , Mitochondria/metabolism , NF-kappa B/metabolism , Animals , CD11c Antigen/metabolism , Cell Proliferation , Cytokines/genetics , Dendritic Cells/metabolism , Fatty Acids/metabolism , Gene Expression Regulation , Immune Tolerance , Inflammation Mediators/metabolism , Mice, Inbred C57BL , PPAR gamma/deficiency , PPAR gamma/metabolism , Promoter Regions, Genetic/genetics , Reactive Oxygen Species/metabolism , T-Lymphocytes/cytology
3.
J Clin Invest ; 125(8): 3037-50, 2015 Aug 03.
Article in English | MEDLINE | ID: mdl-26121748

ABSTRACT

Severe asthma (SA) is a challenge to control, as patients are not responsive to high doses of systemic corticosteroids (CS). In contrast, mild-moderate asthma (MMA) is responsive to low doses of inhaled CS, indicating that Th2 cells, which are dominant in MMA, do not solely orchestrate SA development. Here, we analyzed broncholalveolar lavage cells isolated from MMA and SA patients and determined that IFN-γ (Th1) immune responses are exacerbated in the airways of individuals with SA, with reduced Th2 and IL-17 responses. We developed a protocol that recapitulates the complex immune response of human SA, including the poor response to CS, in a murine model. Compared with WT animals, Ifng-/- mice subjected to this SA model failed to mount airway hyperresponsiveness (AHR) without appreciable effect on airway inflammation. Conversely, AHR was not reduced in Il17ra-/- mice, although airway inflammation was lower. Computer-assisted pathway analysis tools linked IFN-γ to secretory leukocyte protease inhibitor (SLPI), which is expressed by airway epithelial cells, and IFN-γ inversely correlated with SLPI expression in SA patients and the mouse model. In mice subjected to our SA model, forced SLPI expression decreased AHR in the absence of CS, and it was further reduced when SLPI was combined with CS. Our study identifies a distinct immune response in SA characterized by a dysregulated IFN-γ/SLPI axis that affects lung function.


Subject(s)
Asthma/immunology , Gene Expression Regulation/immunology , Interferon-gamma/immunology , Secretory Leukocyte Peptidase Inhibitor/immunology , Th2 Cells/immunology , Adolescent , Adult , Animals , Asthma/genetics , Asthma/pathology , Bronchoalveolar Lavage , Disease Models, Animal , Gene Expression Regulation/genetics , Humans , Interferon-gamma/genetics , Interleukin-17/genetics , Interleukin-17/immunology , Lung/immunology , Lung/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Middle Aged , Receptors, Interleukin-17/genetics , Receptors, Interleukin-17/immunology , Secretory Leukocyte Peptidase Inhibitor/genetics , Severity of Illness Index , Th1 Cells/immunology , Th1 Cells/pathology , Th2 Cells/pathology
4.
J Immunol ; 195(2): 431-5, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26062999

ABSTRACT

The respiratory tract maintains immune homeostasis despite constant provocation by environmental Ags. Failure to induce tolerogenic responses to allergens incites allergic inflammation. Despite the understanding that APCs have a crucial role in maintaining immune tolerance, the underlying mechanisms are poorly understood. Using mice with a conditional deletion of peroxisome proliferator-activated receptor γ (PPARγ) in CD11c(+) cells, we show that PPARγ performs two critical functions in CD11c(+) cells to induce tolerance, thereby preserving immune homeostasis. First, PPARγ was crucial for the induction of retinaldehyde dehydrogenase (aldh1a2) selectively in CD103(+) dendritic cells, which we recently showed promotes Foxp3 expression in naive CD4(+) T cells. Second, in all CD11c(+) cells, PPARγ was required to suppress expression of the Th17-skewing cytokines IL-6 and IL-23p19. Also, lack of PPARγ in CD11c(+) cells induced p38 MAPK activity, which was recently linked to Th17 development. Thus, PPARγ favors immune tolerance by promoting regulatory T cell generation and blocking Th17 differentiation.


Subject(s)
Hypersensitivity/immunology , Immune Tolerance , PPAR gamma/immunology , Respiratory System/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase/immunology , Aldehyde Dehydrogenase 1 Family , Animals , Antigens, CD/genetics , Antigens, CD/immunology , CD11c Antigen/genetics , CD11c Antigen/immunology , Cell Differentiation , Dendritic Cells/immunology , Dendritic Cells/pathology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Gene Expression Regulation , Homeostasis , Hypersensitivity/genetics , Hypersensitivity/pathology , Immunity, Innate , Integrin alpha Chains/genetics , Integrin alpha Chains/immunology , Interleukin-23 Subunit p19/genetics , Interleukin-23 Subunit p19/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Mice , Mice, Knockout , PPAR gamma/genetics , Respiratory System/pathology , Retinal Dehydrogenase , Signal Transduction , T-Lymphocytes, Regulatory/pathology , Th17 Cells/pathology , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/immunology
5.
J Allergy Clin Immunol ; 136(3): 747-756.e4, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25930197

ABSTRACT

BACKGROUND: Single nucleotide polymorphisms in the human gene for the receptor for advanced glycation end-products (RAGE) are associated with an increased incidence of asthma. RAGE is highly expressed in the lung and has been reported to play a vital role in the pathogenesis of murine models of asthma/allergic airway inflammation (AAI) by promoting expression of the type 2 cytokines IL-5 and IL-13. IL-5 and IL-13 are prominently secreted by group 2 innate lymphoid cells (ILC2s), which are stimulated by the proallergic cytokine IL-33. OBJECTIVE: We sought to test the hypothesis that pulmonary RAGE is necessary for allergen-induced ILC2 accumulation in the lung. METHODS: AAI was induced in wild-type and RAGE knockout mice by using IL-33, house dust mite extract, or Alternaria alternata extract. RAGE's lung-specific role in type 2 responses was explored with bone marrow chimeras and induction of gastrointestinal type 2 immune responses. RESULTS: RAGE was found to drive AAI by promoting IL-33 expression in response to allergen and by coordinating the inflammatory response downstream of IL-33. Absence of RAGE impedes pulmonary accumulation of ILC2s in models of AAI. Bone marrow chimera studies suggest that pulmonary parenchymal, but not hematopoietic, RAGE has a central role in promoting AAI. In contrast to the lung, the absence of RAGE does not affect IL-33-induced ILC2 influx in the spleen, type 2 cytokine production in the peritoneum, or mucus hypersecretion in the gastrointestinal tract. CONCLUSIONS: For the first time, this study demonstrates that a parenchymal factor, RAGE, mediates lung-specific accumulation of ILC2s.


Subject(s)
Asthma/immunology , Immunity, Innate , Interleukin-33/immunology , Lung/immunology , Lymphocytes/immunology , Receptor for Advanced Glycation End Products/immunology , Allergens/administration & dosage , Allergens/immunology , Alternaria/chemistry , Animals , Antigens, Dermatophagoides/administration & dosage , Antigens, Dermatophagoides/immunology , Asthma/chemically induced , Asthma/genetics , Asthma/pathology , Bone Marrow/immunology , Bone Marrow/pathology , Cell Proliferation , Gastrointestinal Tract/immunology , Gastrointestinal Tract/pathology , Gene Expression Regulation , Interleukin-13/genetics , Interleukin-13/immunology , Interleukin-33/genetics , Interleukin-5/genetics , Interleukin-5/immunology , Lung/pathology , Lymphocytes/pathology , Mice , Organ Specificity , Peritoneum/immunology , Peritoneum/pathology , Pyroglyphidae/chemistry , Receptor for Advanced Glycation End Products/genetics , Signal Transduction , Spleen/immunology , Spleen/pathology , Transplantation Chimera
6.
J Immunol ; 192(12): 5471-5475, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24829419

ABSTRACT

We reported previously that c-kit ligation by membrane-bound stem cell factor (mSCF) boosts IL-6 production in dendritic cells (DCs) and a Th17-immune response. However, Th17 establishment also requires heterodimeric IL-23, but the mechanisms that regulate IL-23 gene expression in DCs are not fully understood. We show that IL-23p19 gene expression in lung DCs is dependent on mSCF, which is regulated by the metalloproteinase MMP-9. Th1-inducing conditions enhanced MMP-9 activity, causing cleavage of mSCF, whereas the opposite was true for Th17-promoting conditions. In MMP-9(-/-) mice, a Th1-inducing condition could maintain mSCF and enhance IL-23p19 in DCs, promoting IL-17-producing CD4(+) T cells in the lung. Conversely, mSCF cleavage from bone marrow DCs in vitro by rMMP-9 led to reduced IL-23p19 expression under Th17-inducing conditions, with dampening of intracellular AKT phosphorylation. Collectively, these results show that the c-kit/mSCF/MMP-9 axis regulates IL-23 gene expression in DCs to control IL-17 production in the lung.


Subject(s)
Dendritic Cells/immunology , Gene Expression Regulation/immunology , Interleukin-17/immunology , Interleukin-23 Subunit p19/immunology , Lung/immunology , Matrix Metalloproteinase 9/immunology , Stem Cell Factor/immunology , Animals , Cell Membrane/genetics , Cell Membrane/immunology , Cell Membrane/metabolism , Dendritic Cells/cytology , Dendritic Cells/metabolism , Gene Expression Regulation/genetics , Interleukin-17/genetics , Interleukin-17/metabolism , Interleukin-23 Subunit p19/biosynthesis , Interleukin-23 Subunit p19/genetics , Lung/cytology , Lung/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Knockout , Phosphorylation/genetics , Phosphorylation/immunology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Proto-Oncogene Proteins c-akt/metabolism , Stem Cell Factor/genetics , Stem Cell Factor/metabolism , Th1 Cells/cytology , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/cytology , Th17 Cells/immunology , Th17 Cells/metabolism
7.
J Immunol ; 191(1): 25-9, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23733880

ABSTRACT

Dendritic cell (DC)-T cell interactions that underlie inducible/adaptive regulatory T cell generation and airway tolerance are not well understood. In this study, we show that mice lacking CD11c(hi) lung DCs, but containing plasmacytoid DCs (pDCs), fail tolerization with inhaled Ag and cannot support Foxp3 induction in vivo in naive CD4(+) T cells. CD103(+) DCs from tolerized mice efficiently induced Foxp3 in cocultured naive CD4(+) T cells but pDCs and lung macrophages failed to do so. CD103(+) DCs, but not pDCs or lung macrophages, upregulated the expression of retinaldehyde dehydrogenase 2 (aldh1a2), which is key for the production of retinoic acid, a cofactor for TGF-ß for Foxp3 induction. Batf3(-/-) mice, selectively lacking CD103(+) DCs, failed tolerization by inhaled Ag. Collectively, our data show that pulmonary tolerance is dependent on CD103(+) DCs, correlating with their ability to upregulate aldh1a2, which can promote Foxp3 expression in T cells.


Subject(s)
Aldehyde Dehydrogenase/biosynthesis , Antigens, CD/biosynthesis , CD11c Antigen/physiology , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Forkhead Transcription Factors/biosynthesis , Integrin alpha Chains/biosynthesis , Lung/immunology , Up-Regulation/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Coculture Techniques , Dendritic Cells/enzymology , Dendritic Cells/metabolism , Immune Tolerance/immunology , Lung/enzymology , Lung/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
8.
Nat Med ; 18(10): 1525-30, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22961107

ABSTRACT

Immune tolerance is instituted early in life, during which time regulatory T (T(reg)) cells have an important role. Recurrent infections with respiratory syncytial virus (RSV) in early life increase the risk for asthma in adult life. Repeated infection of infant mice tolerized to ovalbumin (OVA) through their mother's milk with RSV induced allergic airway disease in response to OVA sensitization and challenge, including airway inflammation, hyper-reactivity and higher OVA-specific IgE, as compared to uninfected tolerized control mice. Virus infection induced GATA-3 expression and T helper type 2 (T(H)2) cytokine production in forkhead box P3 (FOXP3)(+) T(reg) cells and compromised the suppressive function of pulmonary T(reg) cells in a manner that was dependent on interleukin-4 receptor α (IL-4Rα) expression in the host. Thus, by promoting a T(H)2-type inflammatory response in the lung, RSV induced a T(H)2-like effector phenotype in T(reg) cells and attenuated tolerance to an unrelated antigen (allergen). Our findings highlight a mechanism by which viral infection targets a host-protective mechanism in early life and increases susceptibility to allergic disease.


Subject(s)
Asthma/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Asthma/virology , Forkhead Transcription Factors/biosynthesis , GATA3 Transcription Factor/biosynthesis , Immune Tolerance , Immunoglobulin E/blood , Interleukin-4 Receptor alpha Subunit/immunology , Interleukin-4 Receptor alpha Subunit/metabolism , Lung/immunology , Lung/metabolism , Lung/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Ovalbumin/immunology , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/metabolism
9.
Clin Transl Sci ; 4(4): 268-73, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21884514

ABSTRACT

Prostaglandin E2 (PGE2), interleukin (IL)-23, and IL-1beta (ß) propagate inflammatory bowel disease (IBD) by enhancing the development and function of IL-17 producing CD4(+) T helper (Th17) cells. CD4(+) T cells that express the C-type lectin-like receptor CD161 have been proposed to be the physiologic pool of circulating Th17 cells implicated in IBD. We sought to understand how PGE2, alone and in combination with IL-23 and IL-1ß, modulate human peripheral CD161(+) CD4(+) memory T cells. We found that CD161(+) cells comprise a significant proportion of human peripheral CD4(+) memory T cells. PGE2 and IL-23 plus IL-1ß synergistically induced early IL-17A secretion from CD161(+) CD4(+) memory T cells and the selective enrichment of IL-17A(+) CD161(+) CD4(+) memory T cells in culture. Conversely, IL-23 plus IL-1ß partially opposed the PGE2-mediated repression of early interferon gamma (IFN-γ) secretion from CD161(+) cells, as well as the PGE2-mediated depletion of IFN-γ(+) CD161(+) cells. Our results suggest that PGE2 and IL-23 plus IL-1ß induce the Th17 immune response preferentially in CD161(+) CD4(+) memory T cells, while divergently regulating their ability to express IFN-γ. We hypothesize that Th17-mediated chronic inflammation in IBD depends on the net response of CD161(+) CD4(+) memory T cells to both PGE2 and IL-23 plus IL-1ß.


Subject(s)
Dinoprostone/pharmacology , Immunologic Memory/drug effects , Interleukin-1beta/pharmacology , Interleukin-23/pharmacology , NK Cell Lectin-Like Receptor Subfamily B/metabolism , Th1 Cells/immunology , Th17 Cells/immunology , CD4 Antigens/metabolism , Cells, Cultured , Humans , Integrin alpha4/metabolism , Interferon-gamma/metabolism , Interleukin-17/metabolism , Th1 Cells/drug effects , Th1 Cells/metabolism , Th17 Cells/drug effects , Th17 Cells/metabolism
10.
Proc Natl Acad Sci U S A ; 108(13): 5360-5, 2011 Mar 29.
Article in English | MEDLINE | ID: mdl-21402950

ABSTRACT

Aspergillus fumigatus is commonly associated with allergic bronchopulmonary aspergillosis in patients with severe asthma in which chronic airway neutrophilia predicts a poor outcome. We were able to recapitulate fungus-induced neutrophilic airway inflammation in a mouse model in our efforts to understand the underlying mechanisms. However, neutrophilia occurred in a mouse strain-selective fashion, providing us with an opportunity to perform a comparative study to elucidate the mechanisms involved. Here we show that TNF-α, largely produced by Ly6c(+)CD11b(+) dendritic cells (DCs), plays a central role in promoting IL-17A from CD4(+) T cells and collaborating with it to induce airway neutrophilia. Compared with C57BL/6 mice, BALB/c mice displayed significantly more TNF-α-producing DCs and macrophages in the lung. Lung TNF-α levels were drastically reduced in CD11c-DTR BALB/c mice depleted of CD11c+ cells, and TNF-α-producing Ly6c(+)CD11b(+) cells were abolished in Dectin-1(-/-) and MyD88(-/-) BALB/c mice. TNF-α deficiency itself blunted accumulation of inflammatory Ly6c(+)CD11b(+) DCs. Also, lack of TNF-α decreased IL-17A but promoted IL-5 levels, switching inflammation from a neutrophil to eosinophil bias resembling that in C57BL/6 mice. The TNF-α(low) DCs in C57BL/6 mice contained more NF-κB p50 homodimers, which are strong repressors of TNF-α transcription. Functionally, collaboration between TNF-α and IL-17A triggered significantly higher levels of the neutrophil chemoattractants keratinocyte cytokine and macrophage inflammatory protein 2 in BALB/c mice. Our study identifies TNF-α as a molecular switch that orchestrates a sequence of events in DCs and CD4 T cells that promote neutrophilic airway inflammation.


Subject(s)
Dendritic Cells/immunology , Eosinophilia/immunology , Interleukin-17/immunology , Interleukin-5/immunology , Lung/immunology , Neutrophils/immunology , Pulmonary Aspergillosis/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Antigens, CD/immunology , CD4-Positive T-Lymphocytes/immunology , Humans , Lung/cytology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , NF-kappa B/metabolism , Toll-Like Receptor 2/immunology
11.
Apoptosis ; 16(4): 334-46, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21193961

ABSTRACT

Macrophages and polymorphonuclear cells (PMNs) rapidly respond to microbial and immune inflammatory stimuli and die during these responses. We have shown earlier that many macrophage and PMN functions are compromised in x-linked immunodeficient (Xid) mice with functional deficiency in Bruton's tyrosine kinase (Btk). We now report that Btk-deficient macrophages show enhanced susceptibility to apoptotic death on exposure to the microbial and immune inflammatory signals bacterial lipopolysaccharide (LPS) and interferon-gamma (IFNγ) in vitro. In vivo in mixed bone marrow (BM) chimeras Btk deficiency leads primarily to loss of peripheral macrophage numbers without affecting BM development, suggesting a role of inflammation-induced apoptosis in regulating macrophage life span. Surprisingly, Btk deficiency does not affect macrophage apoptosis induced by DNA damage or CD95 engagement. Reactive nitrogen and oxygen species also do not contribute to inflammation-induced apoptosis, but apoptotic process involves loss of mitochondrial potential, shows increased activation of caspase 9 and enhanced loss of Bcl-xL. The lack of pro-survival signaling through the Btk-phosphotidylinositol 3-kinase-Akt pathway, and persistent MEK signaling, lead to enhanced death in Btk-deficient macrophages only downstream of inflammatory triggers. These data underline the complex role of Btk in the regulation of macrophage survival and function.


Subject(s)
Apoptosis , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/enzymology , Protein-Tyrosine Kinases/metabolism , Agammaglobulinaemia Tyrosine Kinase , Animals , Apoptosis/drug effects , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Cell Count , Cell Survival/drug effects , DNA Damage , Fas Ligand Protein/metabolism , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/drug effects , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Monocytes/cytology , Monocytes/drug effects , Protein-Tyrosine Kinases/deficiency , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , bcl-X Protein/metabolism , fas Receptor/metabolism
12.
J Immunol ; 179(2): 797-803, 2007 Jul 15.
Article in English | MEDLINE | ID: mdl-17617569

ABSTRACT

Apoptosis-inducing factor (Aif) is a mitochondrial flavoprotein with multiple roles in apoptosis as well as in cellular respiration and redox regulation. The harlequin (Hq) mouse strain carries an aif locus modification causing reduced Aif expression. We demonstrate that activated CD4(+) and CD8(+) peripheral T cells from Hq mice show resistance to neglect-induced death (NID) triggered by growth factor withdrawal, but not to death induced by multiple agents that trigger DNA damage. Aif translocates to the nucleus in cells undergoing NID, and, in Hq T cell blasts, resistance to NID is associated with reduced cytosolic release of mitochondrial cytochrome c, implicating Aif in this event. In contrast, Hq T cell blasts express higher levels of CD95L, demonstrating increased susceptibility to activation-induced cell death (AICD) and apoptosis triggered by hydrogen peroxide. Superoxide scavenging protects from AICD in wild-type, but not Hq, T cell blasts, suggesting that Aif plays a crucial superoxide-scavenging role to regulate T cell AICD. Finally, the altered pattern of death susceptibility is reproduced by siRNA-mediated reduction of Aif expression in normal T cells. Thus, Aif serves nonredundant roles, both proapoptotic and antiapoptotic, in activated peripheral T cells.


Subject(s)
Apoptosis Inducing Factor/metabolism , Apoptosis/physiology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Animals , Apoptosis Inducing Factor/immunology , Blotting, Western , Cell Differentiation/immunology , Flow Cytometry , Mice , Mice, Mutant Strains , Microscopy, Confocal , Mitochondria/metabolism , Mitochondria/pathology , RNA, Small Interfering , Reactive Oxygen Species/metabolism , T-Lymphocytes/cytology , p38 Mitogen-Activated Protein Kinases/immunology , p38 Mitogen-Activated Protein Kinases/metabolism
13.
Blood ; 104(4): 1191-7, 2004 Aug 15.
Article in English | MEDLINE | ID: mdl-15117762

ABSTRACT

Bruton tyrosine kinase (Btk), a non-receptor-associated tyrosine kinase of the Tec family, appears to participate in many myeloid cell functions. We show that macrophages from X-linked immunodeficient (XID) mice lacking functional Btk cannot generate efficient bursts of reactive oxygen intermediates (ROIs). The induction of apoptotic cell death by inflammatory stimuli is also enhanced in XID macrophages. Phagocytosis of bacterial particles is only marginally affected in them. In vivo, XID mice show reduced severity of inflammatory diseases in models of experimental autoimmune encephalomyelitis (EAE), dextran sulfate sodium (DSS)-induced colitis, and carrageenan-induced acute edema. Also, polymorphonuclear neutrophil granulocytes (PMNs) in XID mice show poor ROI and nitric oxide (NO) induction, along with a reduction in PMN recruitment to peritoneal inflammation. XID mice show reduction in PMN numbers in peripheral blood, and their bone marrow shows a reduction in the numbers of both monocytic and granulocytic lineages, extending to the earliest progenitor populations. Thus, Btk is likely to play a significant role at multiple points during the development and functioning of the myeloid lineages, affecting the outcome of many infectious as well as noninfectious inflammatory events in vivo.


Subject(s)
Inflammation/enzymology , Myeloid Cells/enzymology , Protein-Tyrosine Kinases/deficiency , Protein-Tyrosine Kinases/physiology , Agammaglobulinaemia Tyrosine Kinase , Animals , Apoptosis , Bone Marrow/pathology , Cell Count , Cell Lineage , Inflammation/pathology , Macrophages/enzymology , Macrophages/pathology , Mice , Mice, Mutant Strains , Myeloid Cells/pathology , Neutrophils/enzymology , Neutrophils/pathology , Phagocytosis , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...