Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Leukoc Biol ; 100(5): 1011-1025, 2016 11.
Article in English | MEDLINE | ID: mdl-27531927

ABSTRACT

Skeletal muscle regeneration requires coordination between dynamic cellular populations and tissue microenvironments. Macrophages, recruited via CCR2, are essential for regeneration; however, the contribution of macrophages and the role of CCR2 on nonhematopoietic cells has not been defined. In addition, aging and sex interactions in regeneration and sarcopenia are unclear. Muscle regeneration was measured in young (3-6 mo), middle (11-15 mo), old (24-32 mo) male and female CCR2-/- mice. Whereas age-related muscle atrophy/sarcopenia was present, regenerated myofiber cross-sectional area (CSA) in CCR2-/- mice was comparably impaired across all ages and sexes, with increased adipocyte area compared with wild-type (WT) mice. CCR2-/- mice myofibers achieved approximately one third of baseline CSA even 84 d after injury. Regenerated CSA and clearance of necrotic tissue were dependent on bone marrow-derived cellular expression of CCR2. Myogenic progenitor cells isolated from WT and CCR2-/- mice exhibited comparable proliferation and differentiation capacity. The most striking cellular anomaly in injured muscle of CCR2-/- mice was markedly decreased macrophages, with a predominance of Ly6C- anti-inflammatory monocytes/macrophages. Ablation of proinflammatory TLR signaling did not affect muscle regeneration or resolution of necrosis. Of interest, many proinflammatory, proangiogenic, and chemotactic cytokines were markedly elevated in injured muscle of CCR2-/- relative to WT mice despite impairments in macrophage recruitment. Collectively, these results suggest that CCR2 on bone marrow-derived cells, likely macrophages, were essential to muscle regeneration independent of TLR signaling, aging, and sex. Decreased proinflammatory monocytes/macrophages actually promoted a proinflammatory microenvironment, which suggests that inflammaging was present in young CCR2-/- mice.


Subject(s)
Macrophages/physiology , Muscle, Skeletal/physiology , Myositis/physiopathology , Receptors, CCR2/deficiency , Regeneration/physiology , Adaptor Proteins, Vesicular Transport/deficiency , Aging/immunology , Animals , Body Weight , Cell Cycle , Cell Division , Cytokines/blood , Female , Inflammation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Monocytes/physiology , Muscle Development , Muscle, Skeletal/injuries , Myeloid Differentiation Factor 88/deficiency , Myoblasts/pathology , Necrosis , Radiation Chimera , Receptors, CCR2/physiology , Sarcopenia/physiopathology , Specific Pathogen-Free Organisms
2.
PLoS Pathog ; 10(1): e1003848, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24391503

ABSTRACT

Pathogen-associated molecular patterns (PAMPs) trigger host immune response by activating pattern recognition receptors like toll-like receptors (TLRs). However, the mechanism whereby several pathogens, including viruses, activate TLRs via a non-PAMP mechanism is unclear. Endogenous "inflammatory mediators" called damage-associated molecular patterns (DAMPs) have been implicated in regulating immune response and inflammation. However, the role of DAMPs in inflammation/immunity during virus infection has not been studied. We have identified a DAMP molecule, S100A9 (also known as Calgranulin B or MRP-14), as an endogenous non-PAMP activator of TLR signaling during influenza A virus (IAV) infection. S100A9 was released from undamaged IAV-infected cells and extracellular S100A9 acted as a critical host-derived molecular pattern to regulate inflammatory response outcome and disease during infection by exaggerating pro-inflammatory response, cell-death and virus pathogenesis. Genetic studies showed that the DDX21-TRIF signaling pathway is required for S100A9 gene expression/production during infection. Furthermore, the inflammatory activity of extracellular S100A9 was mediated by activation of the TLR4-MyD88 pathway. Our studies have thus, underscored the role of a DAMP molecule (i.e. extracellular S100A9) in regulating virus-associated inflammation and uncovered a previously unknown function of the DDX21-TRIF-S100A9-TLR4-MyD88 signaling network in regulating inflammation during infection.


Subject(s)
Adaptor Proteins, Vesicular Transport/immunology , Calgranulin B/immunology , DEAD-box RNA Helicases/immunology , Influenza A Virus, H1N1 Subtype/immunology , Myeloid Differentiation Factor 88/immunology , Orthomyxoviridae Infections/immunology , Signal Transduction/immunology , Toll-Like Receptor 4/immunology , Adaptor Proteins, Vesicular Transport/genetics , Animals , Calgranulin B/genetics , DEAD-box RNA Helicases/genetics , Dogs , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Inflammation/virology , Madin Darby Canine Kidney Cells , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/pathology , Signal Transduction/genetics , Toll-Like Receptor 4/genetics
3.
Exp Gerontol ; 47(7): 507-18, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22548913

ABSTRACT

Alveolar macrophages (AMs) are the first immune cells to respond to an invading pathogen and coordinate the inflammatory response within the lungs. Studies suggest that macrophages exhibit age-related deficiencies in Toll-like receptor (TLR) function; however, the impact of this dysfunction during pneumonia, the leading cause of infectious death in the elderly, and the underlying mechanisms responsible remain unclear. We examined disease severity in young, mature, and aged BALB/cBy mice following intratracheal infection with the Gram-positive bacteria Streptococcus pneumoniae (Spn). Both mature and aged mice failed to clear bacteria and as a result had increased mortality, tissue damage and vascular leakage. Early production of TNFα, IL-1ß, and IL-6 during pneumonia declined with age and was associated with an inability of isolated AMs to respond to pneumococcal cell wall (CW) and ethanol-killed Spn ex vivo. Total levels of TLR1 were unaffected by age and TLR2 surface expression was slightly yet significantly increased on aged AMs suggesting that intracellular TLR signaling defects were responsible for the age-related decline in cytokine responsiveness. Following infection of isolated AMs with live Spn, a significant age-related decline in TLR2-induced phosphorylation of p65 NFκB, JNK and p38 MAPK, and an increase in ERK phosphorylation was observed by immunoblotting. These data are the first to demonstrate that TLR2-dependent recognition of Spn by aged AMs is impaired and is associated with a delayed pro-inflammatory cytokine response in vivo along with enhanced susceptibility to pneumococcal pneumonia.


Subject(s)
Aging/immunology , Cytokines/biosynthesis , Macrophages, Alveolar/immunology , Pneumonia, Pneumococcal/immunology , Toll-Like Receptor 2/metabolism , Animals , Cells, Cultured , Colony Count, Microbial , Disease Susceptibility , Female , Interleukin-6/biosynthesis , MAP Kinase Kinase 4/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Pneumonia, Pneumococcal/metabolism , Signal Transduction/immunology , Toll-Like Receptor 2/immunology , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , p38 Mitogen-Activated Protein Kinases/metabolism
4.
J Immunol ; 188(11): 5604-11, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22529298

ABSTRACT

TLR signaling is critical for early host defense against pathogens, but the contributions of mast cell TLR-mediated mechanisms and subsequent effector functions during pulmonary infection are largely unknown. We have previously demonstrated that mast cells, through the production of IL-4, effectively control Francisella tularensis replication. In this study, the highly human virulent strain of F. tularensis SCHU S4 and the live vaccine strain were used to investigate the contribution of mast cell/TLR regulation of Francisella. Mast cells required TLR2 for effective bacterial killing, regulation of the hydrolytic enzyme cathepsin L, and for coordination and trafficking of MHC class II and lysosomal-associated membrane protein 2. Infected TLR2(-/-) mast cells, in contrast to wild-type and TLR4(-/-) cells, lacked detectable IL-4 and displayed increased cell death with a 2-3 log increase of F. tularensis replication, but could be rescued with rIL-4 treatment. Importantly, MHC class II and lysosomal-associated membrane protein 2 localization with labeled F. tularensis in the lungs was greater in wild-type than in TLR2(-/-) mice. These results provide evidence for the important effector contribution of mast cells and TLR2-mediated signaling on early innate processes in the lung following pulmonary F. tularensis infection and provide additional insight into possible mechanisms by which intracellular pathogens modulate respiratory immune defenses.


Subject(s)
Francisella tularensis/growth & development , Francisella tularensis/immunology , Mast Cells/immunology , Mast Cells/metabolism , Signal Transduction/immunology , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 2/physiology , Animals , Cell Death/genetics , Cell Death/immunology , Interleukin-4/deficiency , Mast Cells/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Transport/genetics , Protein Transport/immunology , Signal Transduction/genetics , Toll-Like Receptor 4/physiology , Tularemia/immunology , Tularemia/microbiology , Tularemia/prevention & control
5.
PLoS One ; 7(1): e29695, 2012.
Article in English | MEDLINE | ID: mdl-22295065

ABSTRACT

Human respiratory syncytial virus (RSV) constitute highly pathogenic virus that cause severe respiratory diseases in newborn, children, elderly and immuno-compromised individuals. Airway inflammation is a critical regulator of disease outcome in RSV infected hosts. Although "controlled" inflammation is required for virus clearance, aberrant and exaggerated inflammation during RSV infection results in development of inflammatory diseases like pneumonia and bronchiolitis. Interleukin-1ß (IL-1ß) plays an important role in inflammation by orchestrating the pro-inflammatory response. IL-1ß is synthesized as an immature pro-IL-1ß form. It is cleaved by activated caspase-1 to yield mature IL-1ß that is secreted extracellularly. Activation of caspase-1 is mediated by a multi-protein complex known as the inflammasome. Although RSV infection results in IL-1ß release, the mechanism is unknown. Here in, we have characterized the mechanism of IL-1ß secretion following RSV infection. Our study revealed that NLRP3/ASC inflammasome activation is crucial for IL-1ß production during RSV infection. Further studies illustrated that prior to inflammasome formation; the "first signal" constitutes activation of toll-like receptor-2 (TLR2)/MyD88/NF-κB pathway. TLR2/MyD88/NF-κB signaling is required for pro-IL-1ß and NLRP3 gene expression during RSV infection. Following expression of these genes, two "second signals" are essential for triggering inflammasome activation. Intracellular reactive oxygen species (ROS) and potassium (K(+)) efflux due to stimulation of ATP-sensitive ion channel promote inflammasome activation following RSV infection. Thus, our studies have underscored the requirement of TLR2/MyD88/NF-κB pathway (first signal) and ROS/potassium efflux (second signal) for NLRP3/ASC inflammasome formation, leading to caspase-1 activation and subsequent IL-1ß release during RSV infection.


Subject(s)
Inflammasomes/metabolism , Potassium/metabolism , Reactive Oxygen Species/metabolism , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Virus Infections/pathology , Respiratory Syncytial Viruses/pathogenicity , Signal Transduction , Animals , CARD Signaling Adaptor Proteins , Carrier Proteins/genetics , Carrier Proteins/metabolism , Caspase 1/metabolism , Cell Line , Cytoskeletal Proteins/metabolism , Enzyme Activation , Gene Expression Regulation , Humans , Interleukin-1beta/biosynthesis , Interleukin-1beta/metabolism , Intracellular Space/metabolism , KATP Channels/metabolism , Mice , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Protein Precursors/genetics , Respiratory Syncytial Virus Infections/genetics , Toll-Like Receptor 2/metabolism
6.
J Immunol ; 185(12): 7562-72, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21098227

ABSTRACT

An inadequate innate immune response appears to contribute to the virulence of Francisella tularensis following pulmonary infection. Studies in mice suggest that this poor response results from suppression of proinflammatory cytokine production early during infection, but the mechanisms involved are not understood. PI3K is known to regulate proinflammatory cytokine expression, but its exact role (positive versus negative) is controversial. We sought to clarify the role of PI3K in regulating proinflammatory signaling and cytokine production during infection with F. tularensis live vaccine strain (LVS). In this study, we demonstrate that the induction of TNF and IL-6 expression by LVS in mouse bone marrow-derived macrophages was markedly enhanced when PI3K activity was inhibited by either of the well-known chemical inhibitors, wortmannin or LY294002. The enhanced cytokine expression was accompanied by enhanced activation of p38 MAPK and ERK1/2, both of which were critical for LVS-induced expression of TNF and IL-6. LVS-induced MAPK activation and cytokine production were TLR2- and MyD88- dependent. PI3K/Akt activation was MyD88-dependent, but was surprisingly TLR2-independent. LVS infection also rapidly induced MAPK phosphatase-1 (MKP-1) expression; PI3K and TLR2 signaling were required. Peak levels of MKP-1 correlated closely with the decline in p38 MAPK and ERK1/2 phosphorylation. These data suggest that infection by LVS restrains the TLR2-triggered proinflammatory response via parallel activation of PI3K, leading to enhanced MKP-1 expression, accelerated deactivation of MAPKs, and suppression of proinflammatory cytokine production. This TLR2-independent inhibitory pathway may be an important mechanism by which Francisella suppresses the host's innate immune response.


Subject(s)
Bacterial Vaccines/immunology , Francisella tularensis/immunology , Immunity, Innate/immunology , Phosphatidylinositol 3-Kinases/immunology , Signal Transduction/immunology , Toll-Like Receptor 2/immunology , Tularemia/immunology , Androstadienes/pharmacology , Animals , Bacterial Vaccines/genetics , Bacterial Vaccines/metabolism , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Chromones/pharmacology , Dual Specificity Phosphatase 1/genetics , Dual Specificity Phosphatase 1/immunology , Dual Specificity Phosphatase 1/metabolism , Enzyme Inhibitors/pharmacology , Francisella tularensis/metabolism , Immunity, Innate/drug effects , Immunity, Innate/genetics , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Interleukin-6/genetics , Interleukin-6/immunology , Interleukin-6/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Mutant Strains , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/immunology , Mitogen-Activated Protein Kinase 3/metabolism , Morpholines/pharmacology , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Myeloid Differentiation Factor 88/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Phosphorylation/genetics , Phosphorylation/immunology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Tularemia/genetics , Tularemia/metabolism , Tularemia/prevention & control , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
7.
PLoS One ; 4(11): e7920, 2009 Nov 20.
Article in English | MEDLINE | ID: mdl-19936231

ABSTRACT

BACKGROUND: Francisella tularensis is the causative agent of tularemia and is classified as a Category A select agent. Recent studies have implicated TLR2 as a critical element in the host protective response to F. tularensis infection, but questions remain about whether TLR2 signaling dominates the response in all circumstances and with all species of Francisella and whether F. tularensis PAMPs are predominantly recognized by TLR2/TLR1 or TLR2/TLR6. To address these questions, we have explored the role of Toll-like receptors (TLRs) in the host response to infections with F. tularensis Live Vaccine Strain (LVS) and F. tularensis subspecies (subsp.) novicida in vivo. METHODOLOGY/PRINCIPAL FINDINGS: C57BL/6 (B6) control mice and TLR- or MyD88-deficient mice were infected intranasally (i.n.) or intradermally (i.d.) with F. tularensis LVS or with F. tularensis subsp. novicida. B6 mice survived >21 days following infection with LVS by both routes and survival of TLR1(-/-), TLR4(-/-), and TLR6(-/-) mice infected i.n. with LVS was equivalent to controls. Survival of TLR2(-/-) and MyD88(-/-) mice, however, was significantly reduced compared to B6 mice, regardless of the route of infection or the subspecies of F. tularensis. TLR2(-/-) and MyD88(-/-) mice also showed increased bacterial burdens in lungs, liver, and spleen compared to controls following i.n. infection. Primary macrophages from MyD88(-/-) and TLR2(-/-) mice were significantly impaired in the ability to secrete TNF and other pro-inflammatory cytokines upon ex vivo infection with LVS. TNF expression was also impaired in vivo as demonstrated by analysis of bronchoalveolar lavage fluid and by in situ immunofluorescent staining. CONCLUSIONS/SIGNIFICANCE: We conclude from these studies that TLR2 and MyD88, but not TLR4, play critical roles in the innate immune response to F. tularensis infection regardless of the route of infection or the subspecies. Moreover, signaling through TLR2 does not depend exclusively on TLR1 or TLR6 during F. tularensis LVS infection.


Subject(s)
Francisella tularensis/metabolism , Myeloid Differentiation Factor 88/genetics , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics , Toll-Like Receptors/metabolism , Tularemia/metabolism , Animals , Bronchoalveolar Lavage , Inflammation , Lung/microbiology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Differentiation Factor 88/metabolism , Proteasome Endopeptidase Complex , Signal Transduction , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism
8.
Vaccine ; 27(41): 5554-61, 2009 Sep 18.
Article in English | MEDLINE | ID: mdl-19651173

ABSTRACT

Francisella tularensis, an intracellular Gram-negative bacterium, is the causative agent of tularemia and a potential bioweapon. Currently, there is no licensed vaccine against this organism. We have characterized the efficacy of a defined F. tularensis subsp. novicida mutant (DeltaiglB) as a live attenuated vaccine against pneumonic tularemia. Replication of the iglB mutant (KKF235) in murine macrophages was significantly lower than the wild type novicida strain U112, and exhibited an LD(50) greater than 10(6)-fold (>10(7)CFU vs <10CFU) in an intranasal challenge model. Mice immunized with KKF235 intranasally or orally induced robust antigen-specific splenic IFN-gamma recall responses, as well as the production of systemic and mucosal antibodies. Intranasal vaccination with KKF235 protected mice from subsequent homotypic challenge with U112 as well as heterotypic challenge with F. tularensis subsp. holarctica (LVS). Moreover, protected animals also exhibited minimal pathological changes compared with mock-vaccinated and challenged animals. The protection conferred by KKF235 vaccination was shown to be highly dependent on endogenous IFN-gamma production. Most significantly, oral immunization with KKF235 protected mice from a highly lethal subsp. tularensis (SCHU S4) pulmonary challenge. Collectively, these results further suggest the feasibility of using defined pathogenicity island mutants as live vaccine candidates against pneumonic tularemia.


Subject(s)
Bacterial Vaccines/immunology , Francisella tularensis/immunology , Gene Deletion , Genomic Islands , Tularemia/prevention & control , Administration, Oral , Animals , Antibodies, Bacterial/analysis , Antibodies, Bacterial/blood , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/genetics , Cell Line , Colony Count, Microbial , Female , Francisella tularensis/genetics , Francisella tularensis/pathogenicity , Genes, Bacterial , Interferon-gamma/metabolism , Leukocytes, Mononuclear/immunology , Macrophages/microbiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microbial Viability , Spleen/immunology , Survival Analysis , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology
9.
J Immunol ; 183(2): 1291-300, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19542374

ABSTRACT

We found that IL-17, a signature cytokine of Th17, was produced early in the innate immunity phase after an intranasal infection with the obligate intracellular pathogen Chlamydia muridarum. The airway IL-17, which peaked at 48 h after infection, was dependent on live chlamydial organism replication and MyD88-mediated signaling pathways. Treatment with antibiotics or knockout of the MyD88 gene, but not Toll/IL receptor domain-containing adapter-inducing IFN-beta, can block the early IL-17 production. Treatment of mice with an anti-IL-17-neutralizing mAb enhanced growth of chlamydial organisms in the lung, dissemination to other organs, and decreased mouse survival, whereas treatment with an isotype-matched control IgG had no effect. Although IL-17 did not directly affect chlamydial growth in cell culture, it enhanced the production of other inflammatory cytokines and chemokines by Chlamydia-infected cells and promoted neutrophil infiltration in mouse airways during chlamydial infection, which may contribute to the antichlamydial effect of IL-17. These observations suggest that an early IL-17 response as an innate immunity component plays an important role in initiating host defense against infection with intracellular bacterial pathogens in the airway.


Subject(s)
Chlamydia muridarum , Interleukin-17/immunology , Myeloid Differentiation Factor 88/physiology , Respiratory Tract Infections/immunology , Animals , Chlamydia muridarum/growth & development , Cytokines/biosynthesis , Immunity, Innate , Interleukin-17/biosynthesis , Mice , Myeloid Differentiation Factor 88/immunology , Neutrophil Infiltration , Survival Rate , Time Factors
10.
Proc Natl Acad Sci U S A ; 105(27): 9313-8, 2008 Jul 08.
Article in English | MEDLINE | ID: mdl-18591675

ABSTRACT

Francisella tularensis is an intracellular, Gram-negative bacterium that is the causative agent of pulmonary tularemia. The pathogenesis and mechanisms related to innate resistance against F. tularensis are not completely understood. Mast cells are strategically positioned within mucosal tissues, the major interface with the external environment, to initiate innate responses at the site of infection. Mast cell numbers in the cervical lymph nodes and the lungs progressively increased as early as 48 h after intranasal F. tularensis live vaccine strain (LVS) challenge. We established a primary bone marrow-derived mast cell-macrophage coculture system and found that mast cells significantly inhibit F. tularensis LVS uptake and growth within macrophages. Importantly, mice deficient in either mast cells or IL-4 receptor displayed greater susceptibility to the infection when compared with corresponding wild-type animals. Contact-dependent events and secreted products including IL-4 from mast cells, and IL-4 production from other cellular sources, appear to mediate the observed protective effects. These results demonstrate a previously unrecognized role for mast cells and IL-4 and provide a new dimension to our understanding of the innate immune mechanisms involved in controlling intramacrophage Francisella replication.


Subject(s)
Contact Inhibition , DNA Replication , Francisella tularensis/immunology , Interleukin-4/metabolism , Macrophages/immunology , Macrophages/microbiology , Mast Cells/cytology , Administration, Intranasal , Animals , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/immunology , Cell Adhesion , Immunity, Innate/immunology , Intracellular Space/microbiology , Lung/immunology , Lung/microbiology , Lung/pathology , Mast Cells/immunology , Mast Cells/microbiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Tularemia/immunology , Tularemia/microbiology
11.
Immunol Cell Biol ; 86(6): 515-22, 2008.
Article in English | MEDLINE | ID: mdl-18427567

ABSTRACT

We have previously demonstrated the protective efficacy of intranasal vaccination with a defined Francisella tularensis subsp. novicida DeltaiglC mutant (KKF24) against pulmonary F. novicida U112 challenge. In this study, we further characterized the mechanisms of KKF24-induced immunity. Intranasally vaccinated KKF24 C57BL/6 major histocompatibility class (MHC) class II-/- mice produced minimal antigen-specific interferon (IFN)-gamma and serum antibodies and were highly susceptible (0% survival) to F. novicida challenge, compared to MHC class I-/- or wild-type mice (both 100% survival). Protective immunity could be transferred by immune serum into recipient wild type, but not IFN-gamma-/- mice. The protective effect of KKF24 vaccination against the respiratory F. novicida U112 challenge was not abrogated by anti-CD4 neutralizing antibody treatment and was not conferred by adoptive transfer of KKF24-specific CD4+ T cells. The protective effect of antibody was partially dependent upon Fc receptor-mediated clearance. Taken together, our data indicate that CD4+ T cells are required for priming, but not during the effector phase, of anti-KKF24 antibody-mediated IFN-gamma-dependent immunity against pulmonary F. novicida infection.


Subject(s)
Antibodies, Bacterial/physiology , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/immunology , CD4-Positive T-Lymphocytes/immunology , Interferon-gamma/immunology , Lung/microbiology , Tularemia/immunology , Tularemia/prevention & control , Adoptive Transfer , Animals , Enzyme-Linked Immunosorbent Assay , Francisella tularensis/genetics , Francisella tularensis/immunology , Genes, MHC Class I/physiology , Genes, MHC Class II/physiology , Immunity, Cellular , Immunization , Lymphocyte Depletion , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Receptors, IgG/physiology , Spleen/cytology , Spleen/immunology , Spleen/metabolism , Survival Rate , Vaccination
12.
Int Immunol ; 16(12): 1741-9, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15504762

ABSTRACT

Both germline transcription and switch recombination of heavy chain genes are likely to be regulated by cis elements binding transcription factors in the promoter regions of germline immunoglobulin genes. To identify cis-acting elements important in germline transcription of the murine gamma1 heavy chain gene, we have used a transgenic approach. Seventeen kb gamma1 immunoglobulin transgenes with mutations in three NF-kappaB sites in the gamma1 proximal promoter, a putative CD40 response element, are expressed well. Compared to wild-type transgenes, there is no deficiency in the expression of the transgenes with mutations of the three NF-kappaB sites after induction of splenic B cells with IL-4 alone, CD40L, or CD40L + IL-4. There may be a small reduction in the response of these mutant transgenes after induction with LPS + IL-4. We also prepared transgenes that were truncated at -150 (rather than -2100) and therefore included the wild-type Stat6 binding site at -123 and the three wild-type NF-kappaB sites. Nevertheless, gamma1 germline transcripts were not expressed from these transgenes. We conclude that the three proximal NF-kappaB sites are dispensable for expression of gamma1 germline transcripts under most conditions. However, cis-acting elements distal to -150 must be critical to this transcription.


Subject(s)
CD40 Ligand/physiology , Gene Expression Regulation , Immunoglobulin gamma-Chains/genetics , NF-kappa B/physiology , Response Elements/physiology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CD40 Ligand/genetics , CD40 Ligand/pharmacology , Gene Dosage , Germ Cells/metabolism , Interleukin-4/pharmacology , Lipopolysaccharides/pharmacology , Mice , Mice, Transgenic , Mutation/genetics , NF-kappa B/genetics , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/physiology , Response Elements/genetics , STAT6 Transcription Factor , Trans-Activators/genetics , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...