Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cancer Res ; 78(13): 3589-3603, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29691253

ABSTRACT

Natural killer (NK) cells express receptors specific for MHC class I (MHC-I) molecules involved in "missing-self" recognition of cancer and virus-infected cells. Here we elucidate the role of MHC-I-independent NKR-P1B:Clr-b interactions in the detection of oncogenic transformation by NK cells. Ras oncogene overexpression was found to promote a real-time loss of Clr-b on mouse fibroblasts and leukemia cells, mediated in part via the Raf/MEK/ERK and PI3K pathways. Ras-driven Clr-b downregulation occurred at the level of the Clrb (Clec2d) promoter, nascent Clr-b transcripts, and cell surface Clr-b protein, in turn promoting missing-self recognition via the NKR-P1B inhibitory receptor. Both Ras- and c-Myc-mediated Clr-b loss selectively augmented cytotoxicity of oncogene-transformed leukemia cells by NKR-P1B+ NK cells in vitro and enhanced rejection by WT mice in vivo Interestingly, genetic ablation of either one (Clr-b+/-) or two Clr-b alleles (Clr-b-/-) enhanced survival of Eµ-cMyc transgenic mice in a primary lymphoma model despite preferential rejection of Clr-b-/- hematopoietic cells previously observed following adoptive transfer into naïve wild-type mice in vivo Collectively, these findings suggest that the inhibitory NKR-P1B:Clr-b axis plays a beneficial role in innate detection of oncogenic transformation via NK-cell-mediated cancer immune surveillance, in addition to a pathologic role in the immune escape of primary lymphoma cells in Eµ-cMyc mice in vivo These results provide a model for the human NKR-P1A:LLT1 system in cancer immunosurveillance in patients with lymphoma and suggest it may represent a target for immune checkpoint therapy.Significance: A mouse model shows that an MHC-independent NK-cell recognition axis enables the detection of leukemia cells, with implications for a novel immune checkpoint therapy target in human lymphoma. Cancer Res; 78(13); 3589-603. ©2018 AACR.


Subject(s)
Immunologic Surveillance , Killer Cells, Natural/immunology , Lectins, C-Type/metabolism , Lymphoma/immunology , NK Cell Lectin-Like Receptor Subfamily B/metabolism , Receptors, Cell Surface/metabolism , Receptors, Immunologic/metabolism , Signal Transduction/immunology , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/immunology , Disease Models, Animal , Down-Regulation , HEK293 Cells , Humans , Lectins, C-Type/immunology , Lymphoma/genetics , Lymphoma/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , NIH 3T3 Cells , NK Cell Lectin-Like Receptor Subfamily B/immunology , Receptors, Cell Surface/immunology , Receptors, Immunologic/immunology
2.
Food Chem Toxicol ; 116(Pt A): 32-41, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29454867

ABSTRACT

Microbial-based cleaning products (MBCPs) contain bacteria and chemical constituents. They are used in consumer applications such as odor reduction, unclogging drains, and surface cleaning. To determine the capacity of a model MBCP to contribute to acute allergic lung inflammation, a two-week repeated exposure regimen was used. Mice were exposed by endotracheal instillation to saline alone, MBCP alone, house dust mites (HDM) alone, or sequentially (i.e., MBCP followed by HDM, HDM followed by MBCP, or HDM + MBCP followed by HDM). Both whole MBCP and acellular MBCP filtrate were investigated, and showed minimal differences in the endpoints examined. HDM exposure caused pulmonary perivascular inflammation, bronchiolar mucous cell metaplasia, elevated bronchoalveolar lavage fluid (BALF) eosinophils, and HDM-specific IgG1. For MBCP, notable changes were associated with sequential exposures. MBCP/HDM caused elevated TH2 cytokines in BALF, and elevated neutrophils, eosinophils and IL-5 in peripheral blood. Co-administration of MBCP and HDM followed by HDM resulted in elevated blood and BALF eosinophils and HDM-specific IgE and IgG1. These results demonstrated that acellular MBCP filtrate, and not bacteria within MBCPs, potentiated the acute allergic inflammation to HDM. This methodology could be extended to investigate chronic allergic inflammation and inflammatory potential of other MBCPs and biotechnology products with complex compositions.


Subject(s)
Biological Factors/immunology , Detergents/adverse effects , Pneumonia/immunology , Pyroglyphidae/immunology , Animals , Biological Factors/adverse effects , Biological Factors/analysis , Bronchoalveolar Lavage Fluid/immunology , Detergents/chemistry , Disease Models, Animal , Eosinophils/immunology , Female , Humans , Immunoglobulin E/immunology , Lung/immunology , Mice , Mice, Inbred BALB C , Pneumonia/etiology , Quality Control
3.
Food Chem Toxicol ; 110: 395-401, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29107027

ABSTRACT

The chemical amaranth (AM) is permitted as a colouring agent in a variety of foods. Safety was established based on chronic rodent studies. AM and its metabolite naphthionic acid (NA) can be absorbed through the intestine, exposing circulating immune cells including splenocytes. An AM feeding study in rats demonstrated an increase in blood lymphocytes. Yet, in contrast, AM inhibited the delayed-type hypersensitivity reaction to antigen. DO11.10 mice express a T Cell Receptor specific for ovalbumin323-339 peptide (OVAp) presented by I-Ad MHCII. DO11.10 splenocytes were cultured to evaluate mechanisms by which AM and NA modulate immune cell function in vitro. Exposure to OVAp alone for 72 h induced cell proliferation, and combination with 2 or 20 µg/mL AM increased IFN-γ. Cytotoxicity was evident at higher concentrations of AM (200 and 2000 µg/mL) and NA (2000 µg/mL) in combination with OVAp, as both cell number and cytokine secretion decreased. At 200 µg/mL AM with OVAp, immunotoxicity gene expression was modified and OVAp-specific KJ1-26+ CD28+ cells became enriched. The equivalent dose of NA did not modify those parameters. Using an antigen-specific model in vitro, lower concentrations of AM potentiated pro-inflammatory cytokine production, and higher concentrations of AM and NA demonstrated cytotoxicity.


Subject(s)
Amaranth Dye/pharmacology , Food Coloring Agents/pharmacology , Immunologic Factors/pharmacology , Spleen/immunology , Animals , Cells, Cultured , Female , Interferon-gamma/genetics , Interferon-gamma/immunology , Lymphocytes/drug effects , Lymphocytes/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , Naphthalenesulfonates/pharmacology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Spleen/cytology , Spleen/drug effects
4.
J Innate Immun ; 9(4): 343-358, 2017.
Article in English | MEDLINE | ID: mdl-28288457

ABSTRACT

Natural killer (NK) cells are innate lymphocytes that aid in self-nonself discrimination by recognizing cells undergoing pathological alterations. The NKR-P1B inhibitory receptor recognizes Clr-b, a self-encoded marker of cell health downregulated during viral infection. Here, we show that Clr-b loss during mouse cytomegalovirus (MCMV) infection is predicated by a loss of Clr-b (Clec2d) promoter activity and nascent transcripts, driven in part by MCMV ie3 (M122) activity. In contrast, uninfected bystander cells near MCMV-infected fibroblasts reciprocally upregulate Clr-b expression due to paracrine type-I interferon (IFN) signaling. Exposure of fibroblasts to type-I IFN augments Clec2d promoter activity and nascent Clr-b transcripts, dependent upon a cluster of IRF3/7/9 motifs located ∼200 bp upstream of the transcriptional start site. Cells deficient in type-I IFN signaling components revealed IRF9 and STAT1 as key transcription factors involved in Clr-b upregulation. In chromatin immunoprecipitation experiments, the Clec2d IRF cluster recruited STAT2 upon IFN-α exposure, confirming the involvement of ISGF3 (IRF9/STAT1/STAT2) in positively regulating the Clec2d promoter. These findings demonstrate that Clr-b is an IFN-stimulated gene on healthy bystander cells, in addition to a missing-self marker on MCMV-infected cells, and thereby enhances the dynamic range of innate self-nonself discrimination by NK cells.


Subject(s)
Fibroblasts/physiology , Herpesviridae Infections/immunology , Killer Cells, Natural/immunology , Lectins, C-Type/metabolism , Muromegalovirus/immunology , NK Cell Lectin-Like Receptor Subfamily B/metabolism , Receptors, Immunologic/metabolism , STAT1 Transcription Factor/genetics , Animals , Cytotoxicity, Immunologic , Immunity, Innate , Interferon Type I/metabolism , Interferon-Stimulated Gene Factor 3, gamma Subunit/genetics , Lectins, C-Type/genetics , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , NIH 3T3 Cells , Paracrine Communication , Promoter Regions, Genetic/genetics , Receptors, Immunologic/genetics , STAT2 Transcription Factor/genetics
5.
Nanotoxicology ; 10(10): 1422-1430, 2016 12.
Article in English | MEDLINE | ID: mdl-27534448

ABSTRACT

Humans could become exposed to carbon black nanoparticles (CBNPs) in consumer products or an occupational setting. In rodent models, acute respiratory, subcutaneous, and direct immune cell exposure to CBNPs has been shown to enhance allergic sensitization to co-administered ovalbumin (OVA) protein from chicken egg. However, little is known about the effects of ingested CBNPs on immunological responses and oral tolerance to food antigens. We hypothesized that ingestion of CBNPs would enhance the development of food allergy to OVA. Allergy prone DO11.10 mice were orally exposed to CBNPs every second day for 2 weeks (total dose 10.8 (LOW) or 108 µg (HI)), with and without (±) co-administered OVA. Systemic immune parameters were measured at necropsy. Exposure to OVA resulted in significant increases in serum anti-OVA IgG1, anti-OVA IgM, and anti-OVA IgA antibodies relative to vehicle control. Immunophenotyping revealed a reduction in the number of OVA-specific CD4+ T helper cells upon OVA ± CBNPHI treatment in the spleen. Yet, secretion of the allergy-associated Th2 cytokines IL-4, IL-9, and IL-13 was greater in OVA323-339 peptide-pulsed splenocytes from OVA + CBNPHI-treated mice compared with control. Transcriptome analysis at necropsy of splenocytes from OVA + CBNPHI dose mice compared with OVA mice revealed increases in the allergy associated genes Il4 and Stat6 and decreases in Csf3r and Retnlg. Although oral exposure to high-dose CBNPs did not impact OVA-specific antibody production relative to OVA, we did observe increased expression of genes and cytokines associated with allergy in peripheral splenocytes. This work suggests that CBNP gastrointestinal exposure may potentiate allergy pathways.


Subject(s)
Food Hypersensitivity , Nanoparticles/toxicity , Ovalbumin/immunology , Receptors, Antigen, T-Cell/genetics , Soot/toxicity , Administration, Oral , Animals , Cytokines/metabolism , Female , Food Hypersensitivity/etiology , Food Hypersensitivity/genetics , Food Hypersensitivity/immunology , Immunoglobulins/blood , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , Ovalbumin/genetics , Spleen/drug effects , Spleen/immunology , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology , Th2 Cells/drug effects , Th2 Cells/immunology
6.
J Immunol ; 194(6): 2909-18, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25681346

ABSTRACT

MHC-I-specific receptors play a vital role in NK cell-mediated "missing-self" recognition, which contributes to NK cell activation. In contrast, MHC-independent NK recognition mechanisms are less well characterized. In this study, we investigated the role of NKR-P1B:Clr-b (Klrb1:Clec2d) interactions in determining the outcome of murine hematopoietic cell transplantation in vivo. Using a competitive transplant assay, we show that Clr-b(-/-) bone marrow (BM) cells were selectively rejected by wild-type B6 recipients, to a similar extent as H-2D(b-/-) MHC-I-deficient BM cells. Selective rejection of Clr-b(-/-) BM cells was mitigated by NK depletion of recipient mice. Competitive rejection of Clr-b(-/-) BM cells also occurred in allogeneic transplant recipients, where it was reversed by selective depletion of NKR-P1B(hi) NK cells, leaving the remaining NKR-P1B(lo) NK subset and MHC-I-dependent missing-self recognition intact. Moreover, competitive rejection of Clr-b(-/-) hematopoietic cells was abrogated in Nkrp1b-deficient recipients, which lack the receptor for Clr-b. Of interest, similar to MHC-I-deficient NK cells, Clr-b(-/-) NK cells were hyporesponsive to both NK1.1 (NKR-P1C)-stimulated and IL-12/18 cytokine-primed IFN-γ production. These findings support a unique and nonredundant role for NKR-P1B:Clr-b interactions in missing-self recognition of normal hematopoietic cells and suggest that optimal BM transplant success relies on MHC-independent tolerance mechanisms. These findings provide a model for human NKR-P1A:LLT1 (KLRB1:CLEC2D) interactions in human hematopoietic cell transplants.


Subject(s)
Bone Marrow Transplantation/methods , Killer Cells, Natural/immunology , Lectins, C-Type/immunology , Membrane Proteins/immunology , NK Cell Lectin-Like Receptor Subfamily B/immunology , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Line, Tumor , Flow Cytometry , Gene Expression/immunology , Graft Rejection/genetics , Graft Rejection/immunology , Hematopoietic Stem Cell Transplantation/methods , Histocompatibility Antigen H-2D/genetics , Histocompatibility Antigen H-2D/immunology , Histocompatibility Antigen H-2D/metabolism , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , Killer Cells, Natural/metabolism , Lectins, C-Type/deficiency , Lectins, C-Type/genetics , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , NK Cell Lectin-Like Receptor Subfamily B/deficiency , NK Cell Lectin-Like Receptor Subfamily B/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Homologous
7.
Toxicol Sci ; 138(2): 322-32, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24449417

ABSTRACT

Agglomerated carbon black nanoparticles (CBNPs) administered via respiratory or subcutaneous routes have been shown to promote allergic sensitization to coadministered ovalbumin (OVA) protein in rodents. In the present study, we aimed to model and elucidate the mechanism of this adjuvanticity using an in vitro assay based on T cell sensitization to ovalbumin323₋339 peptide (OVA(p)). CBNP base particles of 22 and 39 nm were characterized and termed CBNP22 and CBNP39 powders. Splenic leukocytes derived from transgenic DO11.10 mice were exposed to suspensions of media alone, concanavalin A mitogen, CBNP agglomerates smaller than 220 nm, OVA(p) alone, OVA(p) + anti-CD28 costimulant, OVA(p) + cyclosporin A immunosuppressant, or OVA(p) + CBNPs. Samples were analyzed at 72 h post-exposure. Proliferation rate, a marker of cellular mitosis, was assessed. Polymerase chain reaction arrays were used to assess genes involved in allergic response pathways. The mitogen control, costimulatory control, and immunosuppressive control chemicals modified the T helper cell proliferation rate. CBNP22 mildly reduced proliferation at 12 µg/ml, but CBNP39 did not. Gene expression analysis of cells treated with OVA(p) showed that coincubation with 12 µg/ml CBNP22 enhanced gene expression of interleukin-4 (IL-4), IL-10, and IL-13, all allergy-associated Th2 cytokines. Coincubation of OVA(p) with 12 µg/ml CBNP39 significantly enhanced IL-13 gene expression concurrent with downregulation of the Th1-associated transcription factor Stat4. IL-4 and IL-13 protein secretion reflected the mRNA trends. The changes were consistently higher in cells exposed to CBNP22 than CBNP39, suggesting that smaller particle size, higher surface area, and higher purity were associated with the direct adjuvant effect on Th2 cells in this genetically susceptible model of OVA allergy.


Subject(s)
Adjuvants, Immunologic/toxicity , Hypersensitivity/immunology , Nanoparticles/chemistry , Ovalbumin/immunology , Soot/toxicity , Th2 Cells/drug effects , Adjuvants, Immunologic/chemistry , Animals , Cell Proliferation/drug effects , Cells, Cultured , Cytokines/genetics , Cytokines/immunology , Female , Flow Cytometry , Hypersensitivity/etiology , Male , Mice , Mice, Inbred Strains , Microscopy, Electron, Scanning , Particle Size , Porosity , Primary Cell Culture , Soot/chemistry , Surface Properties , Th2 Cells/immunology
8.
Cancer Res ; 70(18): 7102-13, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20823164

ABSTRACT

Natural killer (NK) cells can recognize and kill tumor cells lacking "self" markers, such as class I MHC, but the basis for this recognition is not completely understood. NKR-P1 receptors are members of the C-type lectin-related NK receptor superfamily that are conserved from rodents to humans. Identification of Clr ligands for the NKR-P1 receptors has facilitated functional analysis of MHC-independent target cell recognition by NK cells. One receptor-ligand pair, NKR-P1B:Clr-b, can mediate "missing-self" recognition of tumor and infected cells, but the role of this axis in sensing stressed cells remains unknown. Here, we show that Clr-b is rapidly downregulated in cells undergoing genotoxic and cellular stress at the level of both RNA and surface protein. Stress-mediated loss of Clr-b on leukemia cells enhanced cytotoxicity mediated by NKR-P1B(+) NK cells. Notably, Clr-b downregulation was coordinated functionally with stress-mediated upregulation of NKG2D ligands (but not class I MHC). Our findings highlight a unique role for the MHC-independent NKR-P1B:Clr-b missing-self axis in recognition of stressed cells, and provide evidence of two independent levels of Clr-b regulation in stressed cells.


Subject(s)
DNA Damage/immunology , Killer Cells, Natural/immunology , Animals , Aphidicolin/pharmacology , Ataxia Telangiectasia Mutated Proteins , Bleomycin/pharmacology , Cell Cycle Proteins/metabolism , Cisplatin/pharmacology , DNA Damage/drug effects , DNA-Binding Proteins/metabolism , Down-Regulation , Killer Cells, Natural/drug effects , Lectins, C-Type/biosynthesis , Lectins, C-Type/immunology , Membrane Proteins/biosynthesis , Membrane Proteins/immunology , Mice , NIH 3T3 Cells , Protein Serine-Threonine Kinases/metabolism , Purines/pharmacology , Receptors, Immunologic/immunology , Roscovitine , Tumor Suppressor Proteins/metabolism
9.
Semin Immunol ; 20(6): 321-30, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18595730

ABSTRACT

The Ly49 and Nkrp1 loci encode structurally and functionally related cell surface proteins that positively or negatively regulate natural killer (NK) cell-mediated cytotoxicity and cytokine production. Yet despite their clear relatedness and genetic linkage within the NK gene complex (NKC), these two multi-gene families have adopted dissimilar evolutionary strategies. The Ly49 genes are extremely polymorphic and evolutionarily dynamic, with distinct gene numbers, remarkable allelic diversity, and varying MHC-I-ligand specificities and affinities among different murine haplotypes. In contrast, the Nkrp1 genes have opted for overall conservation of genomic organization, sequences, and ligand specificities, with only limited and focused allelic polymorphism. Possible selection pressures driving such varied evolution of the two gene families may include disequilibrium from ligand co-inheritance, pathogen immunoevasin strategies, flexibility in host counter-evolution mechanisms, and the prevalence and dynamics of inherent repetitive elements.


Subject(s)
Evolution, Molecular , NK Cell Lectin-Like Receptor Subfamily A/genetics , NK Cell Lectin-Like Receptor Subfamily B/genetics , Animals , Genetic Variation/immunology , Haplotypes/genetics , Humans , Interspersed Repetitive Sequences/genetics , Models, Genetic , Selection, Genetic
10.
Immunity ; 26(5): 617-27, 2007 May.
Article in English | MEDLINE | ID: mdl-17462921

ABSTRACT

Cytomegaloviruses are known to encode several gene products that function to subvert MHC-dependent immune recognition. Here we characterize a rat cytomegalovirus (RCMV) C-type lectin-like (RCTL) gene product with homology to the Clr ligands for the NKR-P1 receptors. RCMV infection rapidly extinguished host Clr-b expression, thereby sensitizing infected cells to killing by natural killer (NK) cells. However, the RCTL protein functioned as a decoy ligand to protect infected cells from NK killing via direct interaction with the NKR-P1B inhibitory receptor. In vivo, an RCTL mutant virus displayed diminished virulence in an NK-dependent and strain-specific manner, suggesting that host NKR-P1 polymorphisms have evolved to avert the viral decoy mechanism while maintaining Clr-b recognition to preserve self tolerance. These findings reveal a unique strategy adopted by cytomegaloviruses to evade MHC-independent self-nonself discrimination. The existence of lectin-like genes in several poxviruses suggests that this may represent a common theme for viral evasion of innate immunity.


Subject(s)
Immunity, Innate/immunology , Muromegalovirus/immunology , Receptors, Calcitonin/metabolism , Receptors, Immunologic/metabolism , Virus Internalization , Alleles , Amino Acid Sequence , Animals , Animals, Genetically Modified , Calcitonin Receptor-Like Protein , Cell Line , Female , Gene Expression Regulation , Genome, Viral/genetics , Humans , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Ligands , Molecular Sequence Data , Muromegalovirus/genetics , Muromegalovirus/pathogenicity , Phylogeny , Rats , Receptors, Calcitonin/chemistry , Receptors, Calcitonin/genetics , Sequence Alignment , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...