Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Sci Immunol ; 6(61)2021 07 02.
Article in English | MEDLINE | ID: mdl-34215679

ABSTRACT

Excessive cytokine activity underlies many autoimmune conditions, particularly through the interleukin-17 (IL-17) and tumor necrosis factor-α (TNFα) signaling axis. Both cytokines activate nuclear factor κB, but appropriate induction of downstream effector genes requires coordinated activation of other transcription factors, notably, CCAAT/enhancer binding proteins (C/EBPs). Here, we demonstrate the unexpected involvement of a posttranscriptional "epitranscriptomic" mRNA modification [N6-methyladenosine (m6A)] in regulating C/EBPß and C/EBPδ in response to IL-17A, as well as IL-17F and TNFα. Prompted by the observation that C/EBPß/δ-encoding transcripts contain m6A consensus sites, we show that Cebpd and Cebpb mRNAs are subject to m6A modification. Induction of C/EBPs is enhanced by an m6A methylase "writer" and suppressed by a demethylase "eraser." The only m6A "reader" found to be involved in this pathway was IGF2BP2 (IMP2), and IMP2 occupancy of Cebpd and Cebpb mRNA was enhanced by m6A modification. IMP2 facilitated IL-17-mediated Cebpd mRNA stabilization and promoted translation of C/EBPß/δ in response to IL-17A, IL-17F, and TNFα. RNA sequencing revealed transcriptome-wide IL-17-induced transcripts that are IMP2 influenced, and RNA immunoprecipitation sequencing identified the subset of mRNAs that are directly occupied by IMP2, which included Cebpb and Cebpd Lipocalin-2 (Lcn2), a hallmark of autoimmune kidney injury, was strongly dependent on IL-17, IMP2, and C/EBPß/δ. Imp2-/- mice were resistant to autoantibody-induced glomerulonephritis (AGN), showing impaired renal expression of C/EBPs and Lcn2 Moreover, IMP2 deletion initiated only after AGN onset ameliorated disease. Thus, posttranscriptional regulation of C/EBPs through m6A/IMP2 represents a previously unidentified paradigm of cytokine-driven autoimmune inflammation.


Subject(s)
Adenosine/analogs & derivatives , CCAAT-Enhancer-Binding Proteins/immunology , Interleukin-17/immunology , RNA-Binding Proteins/immunology , Tumor Necrosis Factor-alpha/immunology , Adenosine/immunology , Animals , Autoimmunity/immunology , CCAAT-Enhancer-Binding Proteins/genetics , Cell Line , Female , Humans , Inflammation/immunology , Interleukin-17/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , RNA-Binding Proteins/genetics
2.
Stem Cell Reports ; 16(8): 1999-2013, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34242616

ABSTRACT

Hematopoietic stem cell transplantation (HSCT) is a frequent therapeutic approach to restore hematopoiesis in patients with hematologic diseases. Patients receive a hematopoietic stem cell (HSC)-enriched donor cell infusion also containing immune cells, which may have a beneficial effect by eliminating residual neoplastic cells. However, the effect that donor innate immune cells may have on the donor HSCs has not been deeply explored. Here, we evaluate the influence of donor natural killer (NK) cells on HSC fate, concluded that NK cells negatively affect HSC frequency and function, and identified interferon-gamma (IFNγ) as a potential mediator. Interestingly, improved HSC fitness was achieved by NK cell depletion from murine and human donor infusions or by blocking IFNγ activity. Thus, our data suggest that suppression of inflammatory signals generated by donor innate immune cells can enhance engraftment and hematopoietic reconstitution during HSCT, which is particularly critical when limited HSC numbers are available and the risk of engraftment failure is high.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/immunology , Interferon-gamma/immunology , Killer Cells, Natural/immunology , Tissue Donors , Animals , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/immunology , CCAAT-Enhancer-Binding Proteins/metabolism , Cells, Cultured , Coculture Techniques , Gene Expression Profiling/methods , Graft Survival/genetics , Graft Survival/immunology , Hematopoietic Stem Cells/metabolism , Humans , Interferon-gamma/genetics , Interferon-gamma/metabolism , Killer Cells, Natural/metabolism , Lymphocyte Depletion/methods , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mice, Transgenic
3.
J Clin Invest ; 130(12): 6571-6587, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32897881

ABSTRACT

Tregs require Foxp3 expression and induction of a specific DNA hypomethylation signature during development, after which Tregs persist as a self-renewing population that regulates immune system activation. Whether maintenance DNA methylation is required for Treg lineage development and stability and how methylation patterns are maintained during lineage self-renewal remain unclear. Here, we demonstrate that the epigenetic regulator ubiquitin-like with plant homeodomain and RING finger domains 1 (Uhrf1) is essential for maintenance of methyl-DNA marks that stabilize Treg cellular identity by repressing effector T cell transcriptional programs. Constitutive and induced deficiency of Uhrf1 within Foxp3+ cells resulted in global yet nonuniform loss of DNA methylation, derepression of inflammatory transcriptional programs, destabilization of the Treg lineage, and spontaneous inflammation. These findings support a paradigm in which maintenance DNA methylation is required in distinct regions of the Treg genome for both lineage establishment and stability of identity and suppressive function.


Subject(s)
CCAAT-Enhancer-Binding Proteins/immunology , DNA Methylation/immunology , Forkhead Transcription Factors/immunology , T-Lymphocytes, Regulatory/immunology , Ubiquitin-Protein Ligases/immunology , Animals , CCAAT-Enhancer-Binding Proteins/genetics , Forkhead Transcription Factors/genetics , Mice , Mice, Transgenic , Ubiquitin-Protein Ligases/genetics
4.
J Immunol ; 203(11): 3045-3053, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31611260

ABSTRACT

Macrophages drive the pathological process of inflammatory bowel diseases (IBD) mostly by secreting proinflammatory cytokines, such as Tnf-α. Recent studies have indicated the association between epigenetic modifications and macrophage functions. However, epigenetic mechanisms regulating macrophages' functional involvement in IBD remain unknown. In this study, we investigated whether the epigenetic regulator Uhrf1 plays a role in innate immunity by functionally regulating macrophages in intestines. We employed two transgenic strains of mice (one with Uhrf1 deficiency in macrophages [Uhrf1fl/flLyz2-Cre mice] and the other with the two mutations at Uhrf1's DNA methylation regulatory site [Uhrf1YP187/188AA mice]) to assess their susceptibility to dextran sodium sulfate-induced colitis. We examined the cytokines derived from Uhrf1fl/flLyz2-Cre and Uhrf1YP187/188AA macrophages in response to LPS stimulation. We also analyzed the effects of proinflammatory cytokines on Uhrf1 expression in macrophages. The data demonstrated that Uhrf1 deficiency and Uhrf1YP187/188AA mutation resulted in severe colitis in the dextran sodium sulfate-treated mice. In vitro analysis revealed the hypomethylation of Tnf-α promoter and the increased Tnf-α expression in Uhrf1fl/flLyz2-Cre and Uhrf1YP187/188AA macrophages in response to LPS stimulation, and anti-Tnf-α therapy implied the key role of Tnf-α to the aggravated colitis in Uhrf1-deficient mice. Exogenous Tnf-α destabilized Uhrf1 protein through ubiquitination-mediated protein degradation, triggering macrophage activation. In conclusion, we identified that Uhrf1-mediated DNA methylation controls Tnf-α expression of macrophages in the experimental colitis resembling IBD. The epigenetic mechanisms that activate macrophages may provide new therapeutic targets for IBD treatment.


Subject(s)
CCAAT-Enhancer-Binding Proteins/immunology , Colitis/immunology , Inflammation/immunology , Inflammatory Bowel Diseases/immunology , Macrophages/immunology , Tumor Necrosis Factor-alpha/immunology , Ubiquitin-Protein Ligases/immunology , Animals , CCAAT-Enhancer-Binding Proteins/deficiency , CCAAT-Enhancer-Binding Proteins/genetics , Disease Models, Animal , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Methylation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Tumor Necrosis Factor-alpha/genetics , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics
5.
Stem Cell Res Ther ; 9(1): 278, 2018 10 25.
Article in English | MEDLINE | ID: mdl-30359317

ABSTRACT

BACKGROUND: Mesenchymal stem cells (MSCs) isolated from adult tissues (Ad-MSCs) have shown great promise for use in regenerative medicine. However, their poor in vitro expansion capacity and tissue scarcity have been major limitations. In this study, we demonstrate that mouse embryonic stem cells (mESCs) can differentiate into cells with MSC properties. METHODS: Using previously established methods that characterize Ad-MSCs, we analyzed mESC-differentiated fibroblasts (mESC-FBs), including plastic adherence, clonogenic growth, MSC marker expression, tri-lineage differentiation potential, and the capacity to express immunomodulators. RESULTS: Although previously characterized as mESC-differentiated fibroblasts (mESC-FBs), these cells exhibit major properties of Ad-MSCs. However, mESC-FBs also display unique features inherited from ESCs, including robust expansion capacity, senescence resistance, and attenuated innate immunity. In particular, mESC-FBs are insensitive to bacterial endotoxin (lipopolysaccharide, LPS) and do not express LPS-induced inflammatory molecules, in contrast to bone marrow (BM)-MSCs. We further demonstrate that mESC-FBs are resistant to the cytotoxicity associated with inflammatory cytokines, bacterial endotoxins (LPS and heat-killed bacteria), and macrophage-mediated inflammation. CONCLUSIONS: While it remains to be determined how the unique properties of mESC-FBs will affect their immunoregulatory activity under an in vivo condition, our findings demonstrate that ESCs could be used as an alternative source to generate a new class of ESC-MSCs with unique features potentially useful in regenerative medicine.


Subject(s)
Cell Differentiation/drug effects , Fibroblasts/drug effects , Mesenchymal Stem Cells/drug effects , Mouse Embryonic Stem Cells/drug effects , Tretinoin/pharmacology , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Biomarkers/metabolism , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/immunology , Cell Adhesion/drug effects , Cell Proliferation/drug effects , Coculture Techniques , Collagen Type II/genetics , Collagen Type II/immunology , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/immunology , Fibroblasts/cytology , Fibroblasts/immunology , Gene Expression , Humans , Immunity, Innate , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/immunology , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/immunology , PPAR gamma/genetics , PPAR gamma/immunology , RAW 264.7 Cells , Regenerative Medicine/methods , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/immunology
6.
J Immunol ; 201(6): 1692-1704, 2018 09 15.
Article in English | MEDLINE | ID: mdl-30061199

ABSTRACT

The C/EBPα transcription factor is required for myelopoiesis, with prior observations suggesting additional contributions to B lymphopoiesis. Cebpa expression is evident in common lymphoid progenitor (CLP) and preproB cells but is absent in proB and preB cells. We previously observed that marrow lacking the Cebpa +37 kb enhancer is impaired in producing B cells upon competitive transplantation. Additionally, a Cebpa enhancer/promoter-hCD4 transgene is expressed in B/myeloid CFU. Extending these findings, pan-hematopoietic murine Cebpa enhancer deletion using Mx1-Cre leads to expanded CLP, fewer preproB cells, markedly reduced proB and preB cells, and reduced mature B cells, without affecting T cell numbers. In contrast, enhancer deletion at the proB stage using Mb1-Cre does not impair B cell maturation. Further evaluation of CLP reveals that the Cebpa transgene is expressed almost exclusively in Flt3+ multipotent CLP versus B cell-restricted Flt3- CLP. In vitro, hCD4+ preproB cells produce both B and myeloid cells, whereas hCD4- preproB cells only produce B cells. Additionally, a subset of hCD4- preproB cells express high levels of RAG1-GFP, as seen also in proB cells. Global gene expression analysis indicates that hCD4+ preproB cells express proliferative pathways, whereas B cell development and signal transduction pathways predominate in hCD4- preproB cells. Consistent with these changes, Cebpa enhancer-deleted preproB cells downmodulate cell cycle pathways while upregulating B cell signaling pathways. Collectively, these findings indicate that C/EBPα is required for Flt3+ CLP maturation into preproB cells and then for proliferative Cebpaint B/myeloid preproB cells to progress to Cebpalo B cell-restricted preproB cells and finally to Cebpaneg proB cells.


Subject(s)
CCAAT-Enhancer-Binding Proteins/immunology , Cell Differentiation/immunology , Lymphopoiesis/immunology , Myeloid Progenitor Cells/immunology , Precursor Cells, B-Lymphoid/immunology , Animals , CCAAT-Enhancer-Binding Proteins/genetics , CD4 Antigens/genetics , CD4 Antigens/immunology , Cell Differentiation/genetics , Humans , Lymphopoiesis/genetics , Mice , Mice, Transgenic , Myeloid Progenitor Cells/cytology , Precursor Cells, B-Lymphoid/cytology
7.
Exp Hematol ; 66: 63-78.e13, 2018 10.
Article in English | MEDLINE | ID: mdl-30031847

ABSTRACT

TRIBBLES pseudokinases (TRIB1, TRIB2, and TRIB3) are important regulators of normal and malignant hemopoiesis. The relative abundance of each TRIBBLES family member may be important for distinct oncogenic or tumor suppressor functions. We map the expression profiles of TRIB1, TRIB2, and TRIB3 in human and murine hemopoietic stem, progenitor and mature cells, and in human leukemia datasets. Our data show that TRIB1-TRIB2 have an inverse expression relationship in normal hemopoiesis, whereas TRIB1-TRIB3 have a positive correlation. We reveal that TRIB3 expression is high in the dormant hemopoietic stem cell (HSC) population, implicating a novel role for TRIB3 in stem cell quiescence. These analyses support a non-redundant role for each TRIBBLES member during normal hemopoietic differentiation. We show that TRIB1-TRIB2 display a significant negative correlation in myelodysplastic syndrome and acute myeloid leukemia (AML) subtypes, but not in acute lymphoid leukemia. This inverse relationship is specific to certain subtypes of AML. A positive correlation exists in different leukemia subtypes between TRIB1-TRIB3. The TRIB1-TRIB2 and TRIB1-TRIB3 correlations are consistent with a correlative relationship with C/EBP transcription factor family members. Our results have implications for the development of strategies to therapeutically target these genes in different types of leukemia.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/genetics , Cell Cycle Proteins/genetics , Gene Expression Regulation, Leukemic , Hematopoiesis/genetics , Intracellular Signaling Peptides and Proteins/genetics , Leukemia, Myeloid, Acute/genetics , Myelodysplastic Syndromes/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Repressor Proteins/genetics , Animals , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/immunology , Calcium-Calmodulin-Dependent Protein Kinases/immunology , Cell Cycle Proteins/immunology , Cell Differentiation , Cell Lineage/genetics , Cell Lineage/immunology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/pathology , Chromosome Aberrations , Databases, Genetic , Datasets as Topic , Hematopoiesis/immunology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/pathology , Humans , Intracellular Signaling Peptides and Proteins/immunology , Karyotype , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/pathology , Mice , Myelodysplastic Syndromes/immunology , Myelodysplastic Syndromes/pathology , Protein Isoforms/genetics , Protein Isoforms/immunology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Repressor Proteins/immunology
8.
Eur J Haematol ; 101(1): 86-94, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29624746

ABSTRACT

OBJECTIVES: Acute myeloid leukemia (AML) with hyperleukocytosis (HL) is intuitively thought as a unique group with dismal prognosis. However, comprehensive studies regarding the genetic landscape and clinical outcome in this group of patients are limited. METHODS: A total of 693 newly diagnosed de novo non-M3 AML patients were consecutively enrolled. We compared relevant mutations in 20 genes between AML patients with or without HL and exposed their prognostic implications. RESULTS: Hyperleukocytosis, defined as initial white blood cell counts above 50 000/µL, occurred in 28.9% of AML patients. HL patients had higher incidences of FLT3-ITD, NPM1, DNMT3A, CEBPA, and TET2 mutations. Multivariate analysis demonstrated that HL was an independent poor prognostic factor for overall survival and disease-free survival in total patients, those with intermediate-risk cytogenetics and normal karyotype irrespective of genetic alterations. Intriguingly, HL predicted poor survival in CEBPA double mutated, NPM1 + /FLT3-ITD- and NPM1-/FLT3-ITD- patients. Further, HL patients who received allogeneic hematopoietic stem cell transplantation (allo-HSCT) in first complete remission (CR) had a significantly longer overall survival and disease-free survival than those without allo-HSCT. CONCLUSIONS: Hyperleukocytosis is an independent poor prognostic factor irrespective of cytogenetics and mutation status. Allo-HSCT in first CR seems to ameliorate the poor prognostic impact of HL.


Subject(s)
Gene Expression Regulation, Leukemic , Hematopoietic Stem Cell Transplantation , Leukemia, Myeloid, Acute/diagnosis , Leukocytosis/diagnosis , Mutation , Adolescent , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/immunology , Cohort Studies , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/immunology , DNA Methyltransferase 3A , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Dioxygenases , Female , Humans , Karyotyping , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/therapy , Leukocytosis/genetics , Leukocytosis/mortality , Leukocytosis/therapy , Male , Middle Aged , Multivariate Analysis , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Nucleophosmin , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/immunology , Remission Induction , Risk Factors , Survival Analysis , Transplantation, Homologous , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/immunology
9.
Blood ; 131(10): 1081-1093, 2018 03 08.
Article in English | MEDLINE | ID: mdl-29343483

ABSTRACT

Members of the NR4A subfamily of nuclear receptors have complex, overlapping roles during hematopoietic cell development and also function as tumor suppressors of hematologic malignancies. We previously identified NR4A1 and NR4A3 (NR4A1/3) as functionally redundant suppressors of acute myeloid leukemia (AML) development. However, their role in hematopoietic stem cell (HSC) homeostasis remains to be disclosed. Using a conditional Nr4a1/Nr4a3 knockout mouse (CDKO), we show that codepletion of NR4A1/3 promotes acute changes in HSC homeostasis including loss of HSC quiescence, accumulation of oxidative stress, and DNA damage while maintaining stem cell regenerative and differentiation capacity. Molecular profiling of CDKO HSCs revealed widespread upregulation of genetic programs governing cell cycle and inflammation and an aberrant activation of the interferon and NF-κB signaling pathways in the absence of stimuli. Mechanistically, we demonstrate that NR4A1/3 restrict HSC proliferation in part through activation of a C/EBPα-driven antiproliferative network by directly binding to a hematopoietic-specific Cebpa enhancer and activating Cebpa transcription. In addition, NR4A1/3 occupy the regulatory regions of NF-κB-regulated inflammatory cytokines, antagonizing the activation of NF-κB signaling. Taken together, our results reveal a novel coordinate control of HSC quiescence by NR4A1/3 through direct activation of C/EBPα and suppression of activation of NF-κB-driven proliferative inflammatory responses.


Subject(s)
CCAAT-Enhancer-Binding Proteins/immunology , Cell Proliferation , DNA-Binding Proteins/immunology , Hematopoietic Stem Cells/immunology , Nerve Tissue Proteins/immunology , Nuclear Receptor Subfamily 4, Group A, Member 1/immunology , Receptors, Steroid/immunology , Receptors, Thyroid Hormone/immunology , Signal Transduction/immunology , Animals , CCAAT-Enhancer-Binding Proteins/genetics , DNA Damage , DNA-Binding Proteins/genetics , Enhancer Elements, Genetic/immunology , Hematopoietic Stem Cells/pathology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Oxidative Stress/genetics , Oxidative Stress/immunology , Receptors, Steroid/genetics , Receptors, Thyroid Hormone/genetics , Signal Transduction/genetics , Transcription, Genetic/immunology
10.
Clin Calcium ; 27(6): 845-850, 2017.
Article in Japanese | MEDLINE | ID: mdl-28536323

ABSTRACT

More than 100 years ago, macrophages was discovered. So far, we thought that these are no subtype of macrophages in out body. However, recent studies of these cell types indicate that more than two types of macrophages are existed, and each cell types has inherent functions. In this review, we describe the functional diversity of various type of disorder-specific macrophages and its differentiation mechanism.


Subject(s)
Macrophages/immunology , Animals , CCAAT-Enhancer-Binding Proteins/immunology , Cell Differentiation , Fibrosis/immunology , Humans , Stem Cells/immunology
11.
J Pineal Res ; 62(3)2017 Apr.
Article in English | MEDLINE | ID: mdl-28152213

ABSTRACT

Melatonin affects a variety of physiological processes including circadian rhythms, cellular redox status, and immune function. Importantly, melatonin significantly influences T-cell-mediated immune responses, which are crucial to protect mammals against cancers and infections, but are associated with pathogenesis of many autoimmune diseases. This review focuses on our current understanding of the significance of melatonin in T-cell biology and the beneficial effects of melatonin in T-cell response-based diseases. In addition to expressing both membrane and nuclear receptors for melatonin, T cells have the four enzymes required for the synthesis of melatonin and produce high levels of melatonin. Meanwhile, melatonin is highly effective in modulating T-cell activation and differentiation, especially for Th17 and Treg cells, and also memory T cells. Mechanistically, the influence of melatonin in T-cell biology is associated with membrane and nuclear receptors as well as receptor-independent pathways, for example, via calcineurin. Several cell signaling pathways, including ERK1/2-C/EBPα, are involved in the regulatory roles of melatonin in T-cell biology. Through modulation in T-cell responses, melatonin exerts beneficial effects in various inflammatory diseases, such as type 1 diabetes, systemic lupus erythematosus, and multiple sclerosis. These findings highlight the importance of melatonin signaling in T-cell fate determination, and T cell-based immune pathologies.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Lupus Erythematosus, Systemic/immunology , MAP Kinase Signaling System/immunology , Multiple Sclerosis/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Animals , CCAAT-Enhancer-Binding Proteins/immunology , Diabetes Mellitus, Type 1/pathology , Humans , Lupus Erythematosus, Systemic/pathology , Mitogen-Activated Protein Kinase 1/immunology , Mitogen-Activated Protein Kinase 3/immunology , Multiple Sclerosis/pathology , T-Lymphocytes, Regulatory/pathology , Th17 Cells/pathology
12.
Stem Cell Reports ; 6(6): 940-956, 2016 06 14.
Article in English | MEDLINE | ID: mdl-27264973

ABSTRACT

Toll-like receptor 4 (TLR4) plays a central role in host responses to bacterial infection, but the precise mechanism(s) by which its downstream signaling components coordinate the bone marrow response to sepsis is poorly understood. Using mice deficient in TLR4 downstream adapters MYD88 or TRIF, we demonstrate that both cell-autonomous and non-cell-autonomous MYD88 activation are major causes of myelosuppression during sepsis, while having a modest impact on hematopoietic stem cell (HSC) functions. In contrast, cell-intrinsic TRIF activation severely compromises HSC self-renewal without directly affecting myeloid cells. Lipopolysaccharide-induced activation of MYD88 or TRIF contributes to cell-cycle activation of HSC and induces rapid and permanent changes in transcriptional programs, as indicated by persistent downregulation of Spi1 and CebpA expression after transplantation. Thus, distinct mechanisms downstream of TLR4 signaling mediate myelosuppression and HSC exhaustion during sepsis through unique effects of MyD88 and TRIF.


Subject(s)
Adaptor Proteins, Vesicular Transport/immunology , Hematopoietic Stem Cells/pathology , Myeloid Cells/pathology , Myeloid Differentiation Factor 88/immunology , Sepsis/immunology , Adaptor Proteins, Vesicular Transport/deficiency , Adaptor Proteins, Vesicular Transport/genetics , Animals , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/immunology , Cell Cycle , Disease Models, Animal , Gene Expression Regulation , Hematopoietic Stem Cells/immunology , Lipopolysaccharides , Mice , Mice, Knockout , Myeloid Cells/immunology , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/immunology , Sepsis/genetics , Sepsis/pathology , Signal Transduction , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Trans-Activators/genetics , Trans-Activators/immunology , Transcription, Genetic
13.
Cell Rep ; 14(7): 1581-1589, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26876169

ABSTRACT

Pathogens attack host cells by deploying toxins that perturb core host processes. Recent findings from the nematode C. elegans and other metazoans indicate that surveillance or "effector-triggered" pathways monitor functioning of these core processes and mount protective responses when they are perturbed. Despite a growing number of examples of surveillance immunity, the signaling components remain poorly defined. Here, we show that CEBP-2, the C. elegans ortholog of mammalian CCAAT-enhancer-binding protein gamma, is a key player in surveillance immunity. We show that CEBP-2 acts together with the bZIP transcription factor ZIP-2 in the protective response to translational block by P. aeruginosa Exotoxin A as well as perturbations of other processes. CEBP-2 serves to limit pathogen burden, promote survival upon P. aeruginosa infection, and also promote survival upon Exotoxin A exposure. These findings may have broad implications for the mechanisms by which animals sense pathogenic attack and mount protective responses.


Subject(s)
CCAAT-Enhancer-Binding Proteins/immunology , Caenorhabditis elegans Proteins/immunology , Caenorhabditis elegans/immunology , Death-Associated Protein Kinases/immunology , Host-Pathogen Interactions , Immunologic Surveillance , Pseudomonas aeruginosa/growth & development , ADP Ribose Transferases/biosynthesis , ADP Ribose Transferases/immunology , Animals , Bacterial Toxins/biosynthesis , Bacterial Toxins/immunology , CCAAT-Enhancer-Binding Proteins/antagonists & inhibitors , CCAAT-Enhancer-Binding Proteins/genetics , Caenorhabditis elegans/genetics , Caenorhabditis elegans/microbiology , Caenorhabditis elegans Proteins/antagonists & inhibitors , Caenorhabditis elegans Proteins/genetics , Death-Associated Protein Kinases/antagonists & inhibitors , Death-Associated Protein Kinases/genetics , Exotoxins/biosynthesis , Exotoxins/immunology , Gene Expression Regulation/immunology , Immunity, Innate , Pseudomonas aeruginosa/immunology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Survival Rate , Virulence Factors/biosynthesis , Virulence Factors/immunology , Pseudomonas aeruginosa Exotoxin A
14.
Sci Rep ; 6: 21502, 2016 Feb 11.
Article in English | MEDLINE | ID: mdl-26865111

ABSTRACT

Bacterial endophthalmitis remains a devastating inflammatory condition associated with permanent vision loss. Hence, assessing the host response in this disease may provide new targets for intervention. Using a mouse model of Staphylococcus aureus (SA) endophthalmitis and performing retinal transcriptome analysis, we discovered progressive changes in the expression of 1,234 genes. Gene ontology (GO) and pathway analyses revealed the major pathways impacted in endophthalmitis includes: metabolism, inflammatory/immune, antimicrobial, cell trafficking, and lipid biosynthesis. Among the immune/inflammation pathways, JAK/Stat and IL-17A signaling were the most significantly affected. Interactive network-based analyses identified 13 focus hub genes (IL-6, IL-1ß, CXCL2, STAT3, NUPR1, Jun, CSF1, CYR61, CEBPB, IGF-1, EGFR1, SPP1, and TGM2) within these important pathways. The expression of hub genes confirmed by qRT-PCR, ELISA (IL-6, IL-1ß, and CXCL2), and Western blot or immunostaining (CEBP, STAT3, NUPR1, and IGF1) showed strong correlation with transcriptome data. Since TLR2 plays an important role in SA endophthalmitis, counter regulation analysis of TLR2 ligand pretreated retina or the use of retinas from TLR2 knockout mice showed the down-regulation of inflammatory regulatory genes. Collectively, our study provides, for the first time, a comprehensive analysis of the transcriptomic response and identifies key pathways regulating retinal innate responses in staphylococcal endophthalmitis.


Subject(s)
Endophthalmitis/genetics , Gene Regulatory Networks , Host-Pathogen Interactions , Staphylococcal Infections/genetics , Toll-Like Receptor 2/genetics , Transcriptome , Animals , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/immunology , Cytokines/genetics , Cytokines/immunology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Disease Models, Animal , Endophthalmitis/immunology , Endophthalmitis/microbiology , Endophthalmitis/pathology , Female , Gene Expression Profiling , Gene Expression Regulation , Gene Ontology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Annotation , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Principal Component Analysis , Retina/immunology , Retina/microbiology , Retina/pathology , Signal Transduction , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology , Staphylococcus aureus/pathogenicity , Staphylococcus aureus/physiology , Systems Biology , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 2/immunology
15.
J Clin Invest ; 126(2): 732-44, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26752645

ABSTRACT

The immunoregulatory cytokine macrophage migration inhibitory factor (MIF) is encoded in a functionally polymorphic locus that is linked to the susceptibility of autoimmune and infectious diseases. The MIF promoter contains a 4-nucleotide microsatellite polymorphism (-794 CATT) that repeats 5 to 8 times in the locus, with greater numbers of repeats associated with higher mRNA levels. Because there is no information about the transcriptional regulation of these common alleles, we used oligonucleotide affinity chromatography and liquid chromatography-mass spectrometry to identify nuclear proteins that interact with the -794 CATT5-8 site. An analysis of monocyte nuclear lysates revealed that the transcription factor ICBP90 (also known as UHRF1) is the major protein interacting with the MIF microsatellite. We found that ICBP90 is essential for MIF transcription from monocytes/macrophages, B and T lymphocytes, and synovial fibroblasts, and TLR-induced MIF transcription is regulated in an ICBP90- and -794 CATT5-8 length-dependent manner. Whole-genome transcription analysis of ICBP90 shRNA-treated rheumatoid synoviocytes uncovered a subset of proinflammatory and immune response genes that overlapped with those regulated by MIF shRNA. In addition, the expression levels of ICBP90 and MIF were correlated in joint synovia from patients with rheumatoid arthritis. These findings identify ICBP90 as a key regulator of MIF transcription and provide functional insight into the regulation of the polymorphic MIF locus.


Subject(s)
Arthritis, Rheumatoid/metabolism , CCAAT-Enhancer-Binding Proteins/metabolism , Genetic Loci , Genetic Predisposition to Disease , Intramolecular Oxidoreductases/biosynthesis , Macrophage Migration-Inhibitory Factors/biosynthesis , Microsatellite Repeats , Polymorphism, Genetic , Promoter Regions, Genetic , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/immunology , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/immunology , Humans , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/immunology , Jurkat Cells , Macrophage Migration-Inhibitory Factors/genetics , Macrophage Migration-Inhibitory Factors/immunology , Transcription, Genetic/genetics , Transcription, Genetic/immunology , Ubiquitin-Protein Ligases
16.
Leukemia ; 30(3): 674-82, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26500142

ABSTRACT

B cells have been shown to be refractory to reprogramming and B-cell-derived induced pluripotent stem cells (iPSC) have only been generated from murine B cells engineered to carry doxycycline-inducible Oct4, Sox2, Klf4 and Myc (OSKM) cassette in every tissue and from EBV/SV40LT-immortalized lymphoblastoid cell lines. Here, we show for the first time that freshly isolated non-cultured human cord blood (CB)- and peripheral blood (PB)-derived CD19+CD20+ B cells can be reprogrammed to iPSCs carrying complete VDJH immunoglobulin (Ig) gene monoclonal rearrangements using non-integrative tetracistronic, but not monocistronic, OSKM-expressing Sendai Virus. Co-expression of C/EBPα with OSKM facilitates iPSC generation from both CB- and PB-derived B cells. We also demonstrate that myeloid cells are much easier to reprogram than B and T lymphocytes. Differentiation potential back into the cell type of their origin of B-cell-, T-cell-, myeloid- and fibroblast-iPSCs is not skewed, suggesting that their differentiation does not seem influenced by 'epigenetic memory'. Our data reflect the actual cell-autonomous reprogramming capacity of human primary B cells because biased reprogramming was avoided by using freshly isolated primary cells, not exposed to cytokine cocktails favoring proliferation, differentiation or survival. The ability to reprogram CB/PB-derived primary human B cells offers an unprecedented opportunity for studying developmental B lymphopoiesis and modeling B-cell malignancies.


Subject(s)
B-Lymphocytes/metabolism , CCAAT-Enhancer-Binding Proteins/genetics , Cellular Reprogramming/genetics , Fetal Blood/metabolism , Induced Pluripotent Stem Cells/metabolism , Leukocytes, Mononuclear/metabolism , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Base Sequence , CCAAT-Enhancer-Binding Proteins/immunology , Cell Differentiation , Cell Separation , Cellular Reprogramming/immunology , Fetal Blood/cytology , Fetal Blood/immunology , Gene Expression , Genetic Vectors , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/immunology , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/immunology , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , Molecular Sequence Data , Myeloid Cells/cytology , Myeloid Cells/immunology , Myeloid Cells/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/immunology , Primary Cell Culture , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/immunology , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/immunology , Sendai virus/genetics , V(D)J Recombination/immunology
17.
J Immunol ; 195(1): 80-6, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26019275

ABSTRACT

Neutrophil-specific granule deficiency (SGD) is a rare autosomal recessive primary immunodeficiency characterized by neutrophil dysfunction, bilobed neutrophil nuclei and lack of neutrophil-specific granules. Defects in a myeloid-specific transcription factor, CCAAT/enhancer binding protein-ε (C/EBPε), have been identified in two cases in which homozygous frameshift mutations led to loss of the leucine zipper domain. In this study, we report a 55-y-old woman affected with SGD caused by a novel homozygous 2-aa deletion (ΔRS) in the leucine zipper domain of the C/EBPε gene. The patient showed characteristic neutrophil abnormalities and recurrent skin infections; however, there was no history of deep organ infections. Biochemical analysis revealed that, in contrast to the two frameshift mutations, the ΔRS mutant maintained normal cellular localization, DNA-binding activity, and dimerization, and all three mutants exhibited marked reduction in transcriptional activity. The ΔRS mutant was defective in its association with Gata1 and PU.1, as well as aberrant cooperative transcriptional activation of eosinophil major basic protein. Thus, the ΔRS likely impairs protein-protein interaction with other transcription factors, resulting in a loss of transcriptional activation. These results further support the importance of the leucine zipper domain of C/EBPε for its essential function, and indicate that multiple molecular mechanisms lead to SGD.


Subject(s)
Base Sequence , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/immunology , Lactoferrin/deficiency , Leukocyte Disorders/genetics , Neutrophils/immunology , Sequence Deletion , Adult , Cytoplasmic Granules/immunology , Cytoplasmic Granules/pathology , Eosinophil Major Basic Protein/genetics , Eosinophil Major Basic Protein/immunology , Female , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/immunology , Gene Expression Regulation , Homozygote , Humans , Lactoferrin/genetics , Lactoferrin/immunology , Leukocyte Disorders/immunology , Leukocyte Disorders/pathology , Male , Middle Aged , Molecular Sequence Data , Neutrophils/pathology , Protein Binding , Protein Structure, Tertiary , Proteoglycans/genetics , Proteoglycans/immunology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/immunology , Signal Transduction , Trans-Activators/genetics , Trans-Activators/immunology , Transcription, Genetic
18.
J Immunol ; 195(1): 217-26, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26019273

ABSTRACT

Proper regulation of microbial-induced cytokines is critical to intestinal immune homeostasis. Acute stimulation of nucleotide-binding oligomerization domain 2 (NOD2), the Crohn's disease-associated sensor of bacterial peptidoglycan, induces cytokines. However, chronic NOD2 stimulation in macrophages decreases cytokines upon pattern recognition receptor (PRR) restimulation; cytokine attenuation to PRR stimulation is similarly observed in intestinal macrophages. The role for the transcriptional repressors Twist1 and Twist2 in regulating PRR-induced cytokine outcomes is poorly understood and has not been reported for NOD2. We found that Twist1 and Twist2 were required for optimal cytokine downregulation during acute and, particularly, chronic NOD2 stimulation of human macrophages. Consistently, Twist1 and Twist2 expression was increased after chronic NOD2 stimulation; this increased expression was IL-10 and TGF-ß dependent. Although Twist1 and Twist2 did not coregulate each other's expression, they cooperated to enhance binding to cytokine promoters after chronic NOD2 stimulation. Moreover, Twist1 and Twist2 contributed to enhance expression and promoter binding of the proinflammatory inhibitor c-Maf and the transcriptional repressor Bmi1. Restoring c-Maf and Bmi1 expression in Twist-deficient macrophages restored NOD2-induced cytokine downregulation. Furthermore, with chronic NOD2 stimulation, Twist1 and Twist2 contributed to the decreased expression and cytokine promoter binding of the transcriptional activators activating transcription factor 4, C/EBPα, Runx1, and Runx2. Knockdown of these transcriptional activators in Twist-deficient macrophages restored cytokine downregulation after chronic NOD2 stimulation. Finally, NOD2 synergized with additional PRRs to increase Twist1 and Twist2 expression and Twist-dependent pathways. Therefore, after chronic NOD2 stimulation Twist1 and Twist2 coordinate the regulation of both transcriptional activators and repressors, thereby mediating optimal cytokine downregulation.


Subject(s)
Macrophage Activation/drug effects , Macrophages/immunology , Nod2 Signaling Adaptor Protein/immunology , Nuclear Proteins/immunology , Repressor Proteins/immunology , Twist-Related Protein 1/immunology , Acetylmuramyl-Alanyl-Isoglutamine/pharmacology , Activating Transcription Factor 4/antagonists & inhibitors , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/immunology , Antibodies, Neutralizing/pharmacology , CCAAT-Enhancer-Binding Proteins/antagonists & inhibitors , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/immunology , Core Binding Factor Alpha 1 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/immunology , Core Binding Factor Alpha 2 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/immunology , Gene Expression Regulation , Humans , Interleukin-10/antagonists & inhibitors , Interleukin-10/genetics , Interleukin-10/immunology , Macrophages/cytology , Macrophages/drug effects , Nod2 Signaling Adaptor Protein/agonists , Nod2 Signaling Adaptor Protein/genetics , Nuclear Proteins/genetics , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/immunology , Primary Cell Culture , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins c-maf/genetics , Proto-Oncogene Proteins c-maf/immunology , RNA, Small Interfering/genetics , RNA, Small Interfering/immunology , Repressor Proteins/genetics , Signal Transduction , Transcription, Genetic , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/immunology , Twist-Related Protein 1/genetics
19.
Int Immunol ; 27(8): 381-91, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25840438

ABSTRACT

Periodontitis is caused by multi-bacterial infection and Aggregatibacter actinomycetemcomitans and Enterococcus faecalis are closely associated with inflammatory periodontal diseases. Although lipopolysaccharide (LPS) of A. actinomycetemcomitans (Aa.LPS) and lipoteichoic acid of E. faecalis (Ef.LTA) are considered to be major virulence factors evoking inflammatory responses, their combinatorial effect on the induction of chemokines has not been investigated. In this study, we investigated the interaction between Aa.LPS and Ef.LTA on IL-8 expression in human periodontal ligament (PDL) cells. Aa.LPS, but not Ef.LTA, substantially induced IL-8 expression at the protein and mRNA levels. Interestingly, Ef.LTA suppressed Aa.LPS-induced IL-8 expression without affecting the binding of Aa.LPS to Toll-like receptor (TLR) 4. Ef.LTA reduced Aa.LPS-induced phosphorylation of mitogen-activated protein kinases, including ERK, JNK and p38 kinase. Furthermore, Ef.LTA inhibited the Aa.LPS-induced transcriptional activities of the activating protein 1, CCAAT/enhancer-binding protein and nuclear factor-kappa B transcription factors, all of which are known to regulate IL-8 gene expression. Ef.LTA augmented the expression of IL-1 receptor-associated kinase-M (IRAK-M), a negative regulator of TLR intracellular signaling pathways, in the presence of Aa.LPS at both the mRNA and protein levels. Small interfering RNA silencing IRAK-M reversed the attenuation of Aa.LPS-induced IL-8 expression by Ef.LTA. Collectively, these results suggest that Ef.LTA down-regulates Aa.LPS-induced IL-8 expression in human PDL cells through up-regulation of the negative regulator IRAK-M.


Subject(s)
Aggregatibacter actinomycetemcomitans/chemistry , Enterococcus faecalis/chemistry , Fibroblasts/drug effects , Interleukin-8/genetics , Lipopolysaccharides/pharmacology , Osteoblasts/drug effects , Teichoic Acids/pharmacology , Adult , Aggregatibacter actinomycetemcomitans/metabolism , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/immunology , Enterococcus faecalis/metabolism , Female , Fibroblasts/cytology , Fibroblasts/immunology , Gene Expression Regulation , Humans , Interleukin-1 Receptor-Associated Kinases/antagonists & inhibitors , Interleukin-1 Receptor-Associated Kinases/genetics , Interleukin-1 Receptor-Associated Kinases/immunology , Interleukin-8/agonists , Interleukin-8/antagonists & inhibitors , Interleukin-8/immunology , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/immunology , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/immunology , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/immunology , Osteoblasts/cytology , Osteoblasts/immunology , Periodontal Ligament/cytology , Periodontal Ligament/drug effects , Periodontal Ligament/immunology , Phosphorylation , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Transcription Factor AP-1/genetics , Transcription Factor AP-1/immunology , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/immunology
20.
J Immunol ; 193(7): 3769-78, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25172497

ABSTRACT

Increasing evidence suggests that the novel anti-inflammatory and proresolving mediators such as the resolvins play an important role during inflammation. However, the functions of these lipid mediators in immune complex-induced lung injury remain unknown. In this study, we determined the role of aspirin-triggered resolvin D1 (AT-RvD1) and its metabolically stable analog, 17R-hydroxy-19-para-fluorophenoxy-resolvin D1 methyl ester (p-RvD1), in IgG immune complex-induced inflammatory responses in myeloid cells and injury in the lung. We show that lung vascular permeability in the AT-RvD1- or p-RvD1-treated mice was significantly reduced when compared with values in mice receiving control vesicle during the injury. Furthermore, i.v. administration of either AT-RvD1 or p-RvD1 caused significant decreases in the bronchoalveolar lavage fluid contents of neutrophils, inflammatory cytokines, and chemokines. Of interest, AT-RvD1 or p-RvD1 significantly reduced bronchoalveolar lavage fluid complement C5a level. By EMSA, we demonstrate that IgG immune complex-induced activation of NF-κB and C/EBPß transcription factors in the lung was significantly inhibited by AT-RvD1 and p-RvD1. Moreover, AT-RvD1 dramatically mitigates IgG immune complex-induced NF-κB and C/EBP activity in alveolar macrophages. Also, secretion of TNF-α, IL-6, keratinocyte cell-derived chemokine, and MIP-1α from IgG immune complex-stimulated alveolar macrophages or neutrophils was significantly decreased by AT-RvD1. These results suggest a new approach to the blocking of immune complex-induced inflammation.


Subject(s)
Acute Lung Injury , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antigen-Antibody Complex/immunology , Aspirin/pharmacology , Complement C5a/immunology , Immunoglobulin G/immunology , Pneumonia , Acute Lung Injury/chemically induced , Acute Lung Injury/immunology , Acute Lung Injury/pathology , Acute Lung Injury/prevention & control , Animals , Bronchoalveolar Lavage Fluid/immunology , CCAAT-Enhancer-Binding Proteins/immunology , Cell Line , Chemokine CCL3/immunology , Cytokines/immunology , Docosahexaenoic Acids , Macrophages, Alveolar/immunology , Macrophages, Alveolar/pathology , Mice , NF-kappa B/immunology , Neutrophils/immunology , Neutrophils/pathology , Pneumonia/chemically induced , Pneumonia/immunology , Pneumonia/pathology , Pneumonia/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL