Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters











Database
Language
Publication year range
1.
J Leukoc Biol ; 105(6): 1285-1296, 2019 06.
Article in English | MEDLINE | ID: mdl-30821868

ABSTRACT

It is now evident that NK cells kill bacteria, fungi, and parasites in addition to tumor and virus-infected cells. In addition to a number of recent publications that have identified the receptors and ligands, and mechanisms of cytotoxicity, new insights are reflected in the reports from researchers all over the world at the 17th Meeting of the Society for Natural Immunity held in San Antonio, TX, USA from May 28 through June 1, 2018. We will provide an overview of the field and discuss how the presentations at the meeting might shape our knowledge and future directions in the field.


Subject(s)
Bacteria/immunology , Fungi/immunology , Immunity, Cellular , Killer Cells, Natural/immunology , Viruses/immunology , Animals , Congresses as Topic , Humans , Immunity, Innate , Societies, Scientific , Texas
2.
Cell Host Microbe ; 23(1): 121-133.e4, 2018 Jan 10.
Article in English | MEDLINE | ID: mdl-29290576

ABSTRACT

Candida albicans bloodstream infection causes fungal septicaemia and death in over half of afflicted patients. Polymorphonuclear leukocytes (PMN) mediate defense against invasive candidiasis, but their role in protection versus tissue injury and sepsis is unclear. We observe PMN intravascular swarming and subsequent clustering in response to C. albicans yeast in a lethal septic mouse and human pulmonary circulation model. Live C. albicans sequester to the endothelium and are immediately captured by complement-dependent PMN chemotaxis, which is required for host survival. However, complement activation also leads to Leukotriene B4 (LTB4)-mediated intravascular PMN clustering and occlusion, resulting in capillaritis with pulmonary hemorrhage and hypoxemia. This clustering is unique to fungi and triggered by fungal cell wall components. PMN clustering is absent in mice lacking LTB4-receptor, and capillaritis is attenuated upon pharmacological LTB4 blockade without affecting phagocytosis. Therefore, therapeutically disrupting infection-induced capillaritis may limit organ injury without impairing host defense during fungal sepsis.


Subject(s)
Arterial Occlusive Diseases/microbiology , Candida albicans/immunology , Candidiasis/immunology , Leukotriene B4/immunology , Neutrophil Infiltration/immunology , Neutrophils/immunology , Sepsis/immunology , Animals , Arterial Occlusive Diseases/immunology , Candidiasis/microbiology , Candidiasis/pathology , Cells, Cultured , Female , Humans , Lung/blood supply , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Sepsis/microbiology , Sepsis/pathology
3.
mBio ; 7(4)2016 08 23.
Article in English | MEDLINE | ID: mdl-27555306

ABSTRACT

UNLABELLED: Cryptococcus neoformans is a pathogenic yeast and a leading cause of life-threatening meningitis in AIDS patients. Natural killer (NK) cells are important immune effector cells that directly recognize and kill C. neoformans via a perforin-dependent cytotoxic mechanism. We previously showed that NK cells from HIV-infected patients have aberrant anticryptococcal killing and that interleukin-12 (IL-12) restores the activity at least partially through restoration of NKp30. However, the mechanisms causing this defect or how IL-12 restores the function was unknown. By examining the sequential steps in NK cell killing of Cryptococcus, we found that NK cells from HIV-infected patients had defective binding of NK cells to C. neoformans Moreover, those NK cells that bound to C. neoformans failed to polarize perforin-containing granules to the microbial synapse compared to healthy controls, suggesting that binding was insufficient to restore a defect in perforin polarization. We also identified lower expression of intracellular perforin and defective perforin release from NK cells of HIV-infected patients in response to C. neoformans Importantly, treatment of NK cells from HIV-infected patients with IL-12 reversed the multiple defects in binding, granule polarization, perforin content, and perforin release and restored anticryptococcal activity. Thus, there are multiple defects in the cytolytic machinery of NK cells from HIV-infected patients, which cumulatively result in defective NK cell anticryptococcal activity, and each of these defects can be reversed with IL-12. IMPORTANCE: The mechanisms by which NK cells bind directly to pathogens and deploy their deadly cytolytic machinery during microbial host defense are only beginning to be elucidated. With the goal of understanding this process, we used NK cells from HIV-infected patients, which were known to have a defect in killing of Cryptococcus neoformans Taking advantage of previous studies that had shown that IL-12 restored killing, we used the cytokine as a gain-of-function approach to define the relevance of multiple steps in the recognition and cytolytic pathway. We demonstrated that NK cells from HIV-infected patients failed to kill Cryptococcus due to defects in perforin expression, granule polarization, and release of perforin. Additionally, IL-12 restored recognition of C. neoformans through binding of the NK-activating receptor NKp30. These observations identify important mechanisms used by NK cells to kill microbes and determine that defects in NK cells from HIV-infected patients are reversible.


Subject(s)
Cryptococcosis/immunology , Cryptococcus neoformans/immunology , HIV Infections/complications , Interleukin-12/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/microbiology , Cell Adhesion , Cells, Cultured , Cytoplasmic Granules/metabolism , Humans , Perforin/metabolism
4.
J Biol Chem ; 291(13): 6912-22, 2016 Mar 25.
Article in English | MEDLINE | ID: mdl-26867574

ABSTRACT

The activity of Rac in leukocytes is essential for immunity. However, its role in NK cell-mediated anti-microbial signaling remains unclear. In this study, we investigated the role of Rac in NK cell mediated anti-cryptococcal killing. We found thatCryptococcus neoformansindependently activates both Rac and SFK pathways in NK cells, and unlike in tumor killing,Cryptococcusinitiated a novel Rac → PI3K → Erk cytotoxicity cascade. Remarkably, Rac was not required for conjugate formation, despite its essential role in NK cytotoxicity againstC. neoformans Taken together, our data show that, unlike observations with tumor cells, NK cells use a novel Rac cytotoxicity pathway in conjunction with SFK, to killC. neoformans.


Subject(s)
Class Ia Phosphatidylinositol 3-Kinase/immunology , Cryptococcus neoformans/physiology , Cytotoxicity, Immunologic , Killer Cells, Natural/immunology , rac GTP-Binding Proteins/immunology , rac1 GTP-Binding Protein/immunology , src-Family Kinases/immunology , Cell Line, Tumor , Class Ia Phosphatidylinositol 3-Kinase/genetics , Gene Expression Regulation , Host-Pathogen Interactions , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/microbiology , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/immunology , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/immunology , Phosphorylation/drug effects , Primary Cell Culture , Pyrones/pharmacology , Quinolines/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , rac GTP-Binding Proteins/antagonists & inhibitors , rac GTP-Binding Proteins/genetics , rac1 GTP-Binding Protein/antagonists & inhibitors , rac1 GTP-Binding Protein/genetics , src-Family Kinases/genetics , RAC2 GTP-Binding Protein
5.
J Immunol ; 196(3): 1259-71, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26740109

ABSTRACT

Cryptococcus gattii is an emerging fungal pathogen on the west coast of Canada and the United States that causes a potentially fatal infection in otherwise healthy individuals. In previous investigations of the mechanisms by which C. gattii might subvert cell-mediated immunity, we found that C. gattii failed to induce dendritic cell (DC) maturation, leading to defective T cell responses. However, the virulence factor and the mechanisms of evasion of DC maturation remain unknown. The cryptococcal polysaccharide capsule is a leading candidate because of its antiphagocytic properties. Consequently, we asked if the capsule of C. gattii was involved in evasion of DC maturation. We constructed an acapsular strain of C. gattii through CAP59 gene deletion by homologous integration. Encapsulated C. gattii failed to induce human monocyte-derived DC maturation and T cell proliferation, whereas the acapsular mutant induced both processes. Surprisingly, encapsulation impaired DC maturation independent of its effect on phagocytosis. Indeed, DC maturation required extracellular receptor signaling that was dependent on TNF-α and p38 MAPK, but not ERK activation, and the cryptococcal capsule blocked this extracellular recognition. Although the capsule impaired phagocytosis that led to pH-dependent serine-, threonine-, and cysteine-sensitive protease-dependent Ag processing, it was insufficient to impair T cell responses. In summary, C. gattii affects two independent processes, leading to DC maturation and Ag processing. The polysaccharide capsule masked extracellular detection and reduced phagocytosis that was required for DC maturation and Ag processing, respectively. However, the T cell response was fully restored by inducing DC maturation.


Subject(s)
Antigen Presentation/immunology , Cryptococcosis/immunology , Cryptococcus gattii/immunology , Dendritic Cells/immunology , Fungal Capsules/immunology , Immune Evasion/immunology , Blotting, Western , Cell Proliferation , Humans , Lymphocyte Activation/immunology , T-Lymphocytes/immunology
6.
Cell Host Microbe ; 14(4): 387-97, 2013 Oct 16.
Article in English | MEDLINE | ID: mdl-24139398

ABSTRACT

Natural killer (NK) cells are a subset of immune effectors that directly bind and kill fungi via a perforin-dependent mechanism. The receptor mediating this activity and its potential role in disease remain unknown. Using an unbiased approach, we determined that NKp30 is responsible for recognition and killing of the fungal pathogens Cryptococcus and Candida. NKp30 was required for NK cell-fungal conjugate formation, phosphatidylinositol 3-kinase (PI3K) signaling, and perforin release. Because fungal infections are a leading cause of death in AIDS patients, we examined NKp30 expression in HIV-infected patients. NK cells from these patients had diminished NKp30 expression, defective perforin release, and blunted microbicidal activity. Surprisingly, interleukin-12 (IL-12) restored NKp30 expression and fungal killing. Thus, the NKp30 receptor plays a critical role in NK cell antifungal cytotoxicity, and diminished expression of NKp30 is responsible for defective antifungal activity of NK cells from HIV-infected patients, which can be corrected with IL-12.


Subject(s)
Candida/immunology , Cryptococcus/immunology , HIV Infections/immunology , Host-Pathogen Interactions , Immune Tolerance , Killer Cells, Natural/immunology , Natural Cytotoxicity Triggering Receptor 3/biosynthesis , Cells, Cultured , Down-Regulation , Fungi , Humans , Microbial Viability/drug effects , Perforin/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL