Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
J Immunol ; 209(2): 346-353, 2022 07 15.
Article in English | MEDLINE | ID: mdl-35750336

ABSTRACT

Our recent data demonstrate a critical role of the RIG-I-like receptor family in regulating antifungal immunity against Aspergillus fumigatus in a murine model. However, the importance of this pathway in humans and the cell types that use this innate immune receptor family to detect A. fumigatus remain unresolved. In this study, using patients who underwent hematopoietic stem cell transplantation, we demonstrate that a polymorphism in human MAVS present in the donor genome was associated with the incidence of invasive pulmonary aspergillosis. Moreover, in a separate cohort of confirmed invasive pulmonary aspergillosis patients, polymorphisms in the IFIH1 gene alter the inflammatory response, including IFN-responsive chemokines. Returning to our murine model, we now demonstrate that CD11c+ Siglec F+ alveolar macrophages require Mavs expression to maintain host resistance against A. fumigatus. Our data support the role of MAVS signaling in mediating antifungal immunity in both mice and humans at least in part through the role of MAVS-dependent signaling in alveolar macrophages.


Subject(s)
Aspergillus fumigatus , Invasive Pulmonary Aspergillosis , Animals , Antifungal Agents , Disease Models, Animal , Humans , Macrophages, Alveolar , Mice
2.
J Immunol ; 205(11): 3058-3070, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33087405

ABSTRACT

RIG-I-like receptors (RLR) are cytosolic RNA sensors that signal through the MAVS adaptor to activate IFN responses against viruses. Whether the RLR family has broader effects on host immunity against other pathogen families remains to be fully explored. In this study, we demonstrate that MDA5/MAVS signaling was essential for host resistance against pulmonary Aspergillus fumigatus challenge through the regulation of antifungal leukocyte responses in mice. Activation of MDA5/MAVS signaling was driven by dsRNA from live A. fumigatus serving as a key vitality-sensing pattern recognition receptor. Interestingly, induction of type I IFNs after A. fumigatus challenge was only partially dependent on MDA5/MAVS signaling, whereas type III IFN expression was entirely dependent on MDA5/MAVS signaling. Ultimately, type I and III IFN signaling drove the expression of CXCL10. Furthermore, the MDA5/MAVS-dependent IFN response was critical for the induction of optimal antifungal neutrophil killing of A. fumigatus spores. In conclusion, our data broaden the role of the RLR family to include a role in regulating antifungal immunity against A. fumigatus.


Subject(s)
Aspergillus fumigatus/immunology , Interferon-Induced Helicase, IFIH1/immunology , Interferon-Induced Helicase, IFIH1/metabolism , Pathogen-Associated Molecular Pattern Molecules/immunology , Pathogen-Associated Molecular Pattern Molecules/metabolism , Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Chemokine CXCL10/immunology , Chemokine CXCL10/metabolism , Female , Interferons/immunology , Interferons/metabolism , Male , Mice , Mice, Inbred C57BL , Receptors, Pattern Recognition/immunology , Receptors, Pattern Recognition/metabolism , Signal Transduction/immunology
4.
PLoS Pathog ; 13(4): e1006340, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28423062

ABSTRACT

Aspergillus fumigatus is responsible for a disproportionate number of invasive mycosis cases relative to other common filamentous fungi. While many fungal factors critical for infection establishment are known, genes essential for disease persistence and progression are ill defined. We propose that fungal factors that promote navigation of the rapidly changing nutrient and structural landscape characteristic of disease progression represent untapped clinically relevant therapeutic targets. To this end, we find that A. fumigatus requires a carbon catabolite repression (CCR) mediated genetic network to support in vivo fungal fitness and disease progression. While CCR as mediated by the transcriptional repressor CreA is not required for pulmonary infection establishment, loss of CCR inhibits fungal metabolic plasticity and the ability to thrive in the dynamic infection microenvironment. Our results suggest a model whereby CCR in an environmental filamentous fungus is dispensable for initiation of pulmonary infection but essential for infection maintenance and disease progression. Conceptually, we argue these data provide a foundation for additional studies on fungal factors required to support fungal fitness and disease progression and term such genes and factors, DPFs (disease progression factors).


Subject(s)
Aspergillosis/microbiology , Aspergillus fumigatus/genetics , Carbon/metabolism , Catabolite Repression , Fungal Proteins/metabolism , Gene Regulatory Networks , Aspergillosis/pathology , Aspergillus fumigatus/physiology , Disease Progression , Fungal Proteins/genetics , Gene Expression Regulation, Fungal/drug effects , Models, Biological , Repressor Proteins/genetics , Repressor Proteins/metabolism , Stress, Physiological
5.
J Infect Dis ; 218(5): 809-813, 2018 07 24.
Article in English | MEDLINE | ID: mdl-29668950

ABSTRACT

Staphylococcus aureus is a predominant cause of fatal pneumonia following influenza A virus (IAV) infection. Herein we investigate the influence of antecedent IAV infection on S. aureus virulence gene expression. Using a murine model, comparing the USA300 and USA300ΔsaeR/S strains, we demonstrate that S. aureus pathogenesis following IAV infection is SaeR/S dependent. Furthermore, we show that IAV modulates the lung environment to rapidly up-regulate S. aureus virulence factors containing the SaeR-binding domain. Data demonstrate that the pathogen response to IAV infection impacts host outcome and provides evidence that the ability of S. aureus to sense and respond to the lung environment determines severity of pneumonia.


Subject(s)
Bacterial Proteins/metabolism , Orthomyxoviridae Infections/complications , Pneumonia, Staphylococcal/immunology , Protein Kinases/metabolism , Staphylococcus aureus/pathogenicity , Transcription Factors/metabolism , Animals , Bacterial Proteins/genetics , Disease Models, Animal , Female , Gene Deletion , Male , Mice, Inbred BALB C , Orthomyxoviridae Infections/pathology , Pneumonia, Staphylococcal/genetics , Protein Kinases/genetics , Staphylococcus aureus/genetics , Transcription Factors/genetics
6.
Infect Immun ; 85(12)2017 12.
Article in English | MEDLINE | ID: mdl-28947643

ABSTRACT

Heterogeneity among Aspergillus fumigatus isolates results in unique virulence potential and inflammatory responses. How these isolates drive specific immune responses and how this affects fungally induced lung damage and disease outcome are unresolved. We demonstrate that the highly virulent CEA10 strain is able to rapidly germinate within the immunocompetent lung environment, inducing greater lung damage, vascular leakage, and interleukin 1α (IL-1α) release than the low-virulence Af293 strain, which germinates with a lower frequency in this environment. Importantly, the clearance of CEA10 was consequently dependent on IL-1α, in contrast to Af293. The release of IL-1α occurred by a caspase 1/11- and P2XR7-independent mechanism but was dependent on calpain activity. Our finding that early fungal conidium germination drives greater lung damage and IL-1α-dependent inflammation is supported by three independent experimental lines. First, pregermination of Af293 prior to in vivo challenge drives greater lung damage and an IL-1α-dependent neutrophil response. Second, the more virulent EVOL20 strain, derived from Af293, is able to germinate in the airways, leading to enhanced lung damage and IL-1α-dependent inflammation and fungal clearance. Third, primary environmental A. fumigatus isolates that rapidly germinate under airway conditions follow the same trend toward IL-1α dependency. Our data support the hypothesis that A. fumigatus phenotypic variation significantly contributes to disease outcomes.


Subject(s)
Aspergillosis/immunology , Aspergillus fumigatus/immunology , Aspergillus fumigatus/pathogenicity , Interleukin-1alpha/immunology , Lung/immunology , Animals , Cells, Cultured , Immunocompetence , Inflammation , Lung/microbiology , Macrophages/immunology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Spores, Fungal/immunology , Spores, Fungal/pathogenicity , Virulence
7.
PLoS Pathog ; 11(1): e1004625, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25629406

ABSTRACT

Aspergillus fumigatus is a mold that causes severe pulmonary infections. Our knowledge of how A. fumigatus growth is controlled in the respiratory tract is developing, but still limited. Alveolar macrophages, lung resident macrophages, and airway epithelial cells constitute the first lines of defense against inhaled A. fumigatus conidia. Subsequently, neutrophils and inflammatory CCR2+ monocytes are recruited to the respiratory tract to prevent fungal growth. However, the mechanism of neutrophil and macrophage recruitment to the respiratory tract after A. fumigatus exposure remains an area of ongoing investigation. Here we show that A. fumigatus pulmonary challenge induces expression of the inflammasome-dependent cytokines IL-1ß and IL-18 within the first 12 hours, while IL-1α expression continually increases over at least the first 48 hours. Strikingly, Il1r1-deficient mice are highly susceptible to pulmonary A. fumigatus challenge exemplified by robust fungal proliferation in the lung parenchyma. Enhanced susceptibility of Il1r1-deficient mice correlated with defects in leukocyte recruitment and anti-fungal activity. Importantly, IL-1α rather than IL-1ß was crucial for optimal leukocyte recruitment. IL-1α signaling enhanced the production of CXCL1. Moreover, CCR2+ monocytes are required for optimal early IL-1α and CXCL1 expression in the lungs, as selective depletion of these cells resulted in their diminished expression, which in turn regulated the early accumulation of neutrophils in the lung after A. fumigatus challenge. Enhancement of pulmonary neutrophil recruitment and anti-fungal activity by CXCL1 treatment could limit fungal growth in the absence of IL-1α signaling. In contrast to the role of IL-1α in neutrophil recruitment, the inflammasome and IL-1ß were only essential for optimal activation of anti-fungal activity of macrophages. As such, Pycard-deficient mice are mildly susceptible to A. fumigatus infection. Taken together, our data reveal central, non-redundant roles for IL-1α and IL-1ß in controlling A. fumigatus infection in the murine lung.


Subject(s)
Aspergillus fumigatus/immunology , Chemotaxis, Leukocyte , Interleukin-1alpha/physiology , Pulmonary Aspergillosis/immunology , Animals , Bronchial Provocation Tests , Cells, Cultured , Chemotaxis, Leukocyte/genetics , Chemotaxis, Leukocyte/immunology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Aspergillosis/genetics , Signal Transduction/genetics , Signal Transduction/immunology
8.
Immunity ; 28(6): 859-69, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18499487

ABSTRACT

Through genetic recombination, the adaptive immune system generates a diverse T cell repertoire allowing recognition of a vast spectrum of foreign antigens. Any given CD8+ T cell specificity is thought to be rare, but none have been directly quantified. Here, major histocompatibility complex tetramer and magnetic-bead technology were coupled to quantitate naive antigen-specific CD8+ T cells and the early response to infection. Among six specificities measured, the number of naive antigen-specific precursors ranged from approximately 80 to 1200 cells/mouse. After vesicular stomatitis virus infection, the antigen-specific CD8+ T cell response occurred in discrete phases: prolonged activation of a subset of cells over the first 72 hr followed by a rapid proliferative burst. Naive precursor frequency altered response kinetics and regulated immunodominance, as well as the time required for the responding population to shift toward CD62L(hi) memory cells. Thus, initial endogenous precursor frequencies were surprisingly diverse and not only regulated initial immune response characteristics but also controlled memory CD8+ T cell lineage decisions.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Precursor Cells, T-Lymphoid/immunology , Rhabdoviridae Infections/immunology , Vesicular stomatitis Indiana virus/immunology , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation , Histocompatibility Antigens Class I/immunology , L-Selectin/genetics , L-Selectin/immunology , L-Selectin/metabolism , Lymphocyte Activation , Lymphocyte Count , Mice , Mice, Inbred C57BL , Mice, Transgenic , Precursor Cells, T-Lymphoid/cytology , Precursor Cells, T-Lymphoid/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rhabdoviridae Infections/virology
9.
PLoS Pathog ; 10(9): e1004378, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25255025

ABSTRACT

Hypoxia inducible factor 1α (HIF1α) is the mammalian transcriptional factor that controls metabolism, survival, and innate immunity in response to inflammation and low oxygen. Previous work established that generation of hypoxic microenvironments occurs within the lung during infection with the human fungal pathogen Aspergillus fumigatus. Here we demonstrate that A. fumigatus stabilizes HIF1α protein early after pulmonary challenge that is inhibited by treatment of mice with the steroid triamcinolone. Utilizing myeloid deficient HIF1α mice, we observed that HIF1α is required for survival and fungal clearance early following pulmonary challenge with A. fumigatus. Unlike previously reported research with bacterial pathogens, HIF1α deficient neutrophils and macrophages were surprisingly not defective in fungal conidial killing. The increase in susceptibility of the myeloid deficient HIF1α mice to A. fumigatus was in part due to decreased early production of the chemokine CXCL1 (KC) and increased neutrophil apoptosis at the site of infection, resulting in decreased neutrophil numbers in the lung. Addition of recombinant CXCL1 restored neutrophil survival and numbers, murine survival, and fungal clearance. These results suggest that there are unique HIF1α mediated mechanisms employed by the host for protection and defense against fungal pathogen growth and invasion in the lung. Additionally, this work supports the strategy of exploring HIF1α as a therapeutic target in specific immunosuppressed populations with fungal infections.


Subject(s)
Aspergillus fumigatus/immunology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunity, Innate/immunology , Lung/immunology , Myeloid Cells/immunology , Neutrophils/immunology , Pulmonary Aspergillosis/prevention & control , Animals , Apoptosis , Blotting, Western , Cell Movement , Cell Proliferation , Cells, Cultured , Chemokine CXCL1/genetics , Chemokine CXCL1/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immunoenzyme Techniques , Inflammation/immunology , Inflammation/metabolism , Inflammation/microbiology , Lung/metabolism , Lung/microbiology , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Mice , Myeloid Cells/metabolism , Myeloid Cells/microbiology , Neutrophils/metabolism , Neutrophils/microbiology , Pulmonary Aspergillosis/immunology , Pulmonary Aspergillosis/metabolism , Pulmonary Aspergillosis/microbiology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
10.
Cytokine ; 84: 63-73, 2016 08.
Article in English | MEDLINE | ID: mdl-27253487

ABSTRACT

Invasive aspergillosis (IA) remains a devastating disease in immune compromised patients despite significant advances in our understanding of fungal virulence and host defense mechanisms. In this review, we summarize important research advances in the fight against IA with particular focus on early events in the interactions between Aspergillus fumigatus and the host that occur in the respiratory tract. Advances in understanding mechanisms of immune effector cell recruitment, antifungal effector mechanisms, and how the dynamic host-fungal interaction alters the local microenvironment to effect outcomes are highlighted. These advances illustrate exciting new therapeutic opportunities, but also emphasize the importance of understanding each unique fungus-host interaction for improving patient outcomes.


Subject(s)
Aspergillosis/immunology , Aspergillus fumigatus/immunology , Animals , Antifungal Agents/pharmacology , Antifungal Agents/therapeutic use , Aspergillosis/drug therapy , Aspergillus fumigatus/drug effects , Host-Pathogen Interactions/immunology , Humans , Precision Medicine
11.
J Immunol ; 190(9): 4676-84, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23526820

ABSTRACT

Influenza A virus (IAV) is a major respiratory pathogen of both humans and animals. The lung is protected from pathogens by alveolar epithelial cells, tissue-resident alveolar macrophages, dendritic cells, and mast cells. The role of alveolar epithelial cells, endothelial cells, and alveolar macrophages during IAV infection has been studied previously. In this study, we address the role of mast cells during IAV infection. Respiratory infection with A/WSN/33 causes significant disease and immunopathology in C57BL/6 mice but not in B6.Cg-Kit(W-sh) mice, which lack mast cells. During in vitro coculture, A/WSN/33 caused mast cells to release histamine, secrete cytokines and chemokines, and produce leukotrienes. Moreover, when mast cells were infected with IAV, the virus did not replicate within mast cells. Importantly, human H1N1, H3N2, and influenza B virus isolates also could activate mast cells in vitro. Mast cell production of cytokines and chemokines occurs in a RIG-I/MAVS-dependent mechanism; in contrast, histamine production occurred through a RIG-I/MAVS-independent mechanism. Our data highlight that, following IAV infection, the response of mast cells is controlled by multiple receptors. In conclusion, we identified a unique inflammatory cascade activated during IAV infection that could potentially be targeted to limit morbidity following IAV infection.


Subject(s)
DEAD-box RNA Helicases/metabolism , Inflammation/immunology , Influenza, Human/immunology , Mast Cells/immunology , Orthomyxoviridae Infections/immunology , Animals , Chemokines/immunology , Chemokines/metabolism , DEAD-box RNA Helicases/immunology , Histamine/immunology , Histamine/metabolism , Humans , Inflammation/metabolism , Inflammation/virology , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza, Human/metabolism , Influenza, Human/virology , Leukotrienes/immunology , Leukotrienes/metabolism , Lung/immunology , Lung/metabolism , Lung/virology , Mast Cells/metabolism , Mast Cells/virology , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/virology , STAT6 Transcription Factor/immunology , STAT6 Transcription Factor/metabolism
12.
J Immunol ; 188(6): 2483-7, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22327071

ABSTRACT

IL-15 plays a multifaceted role in immune homeostasis, but the unreliability of IL-15 detection has stymied exploration of IL-15 regulation in vivo. To visualize IL-15 expression, we created a transgenic mouse expressing emerald-GFP (EmGFP) under IL-15 promoter control. EmGFP/IL-15 was prevalent in innate cells including dendritic cells (DCs), macrophages, and monocytes. However, DC subsets expressed varying levels of EmGFP/IL-15 with CD8(+) DCs constitutively expressing EmGFP/IL-15 and CD8(-) DCs expressing low EmGFP/IL-15 levels. Virus infection resulted in IL-15 upregulation in both subsets. By crossing the transgenic mice to mice deficient in specific elements of innate signaling, we found a cell-intrinsic dependency of DCs and Ly6C(+) monocytes on IFN-α receptor expression for EmGFP/IL-15 upregulation after vesicular stomatitis virus infection. In contrast, myeloid cells did not require the expression of MyD88 to upregulate EmGFP/IL-15 expression. These findings provide evidence of previously unappreciated regulation of IL-15 expression in myeloid lineages during homeostasis and following infection.


Subject(s)
Dendritic Cells/metabolism , Interleukin-15/biosynthesis , Signal Transduction/immunology , Animals , Cell Separation , Dendritic Cells/immunology , Flow Cytometry , Interleukin-15/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Myeloid Differentiation Factor 88/immunology , Myeloid Differentiation Factor 88/metabolism , Receptor, Interferon alpha-beta/immunology , Receptor, Interferon alpha-beta/metabolism , Vesicular Stomatitis/immunology
13.
Immunol Rev ; 235(1): 206-18, 2010 May.
Article in English | MEDLINE | ID: mdl-20536565

ABSTRACT

The control of the differentiation pathways followed by responding CD8(+) T cells to produce protective memory cells has been intensely studied. Recent developments have identified heterogeneity at the effector cytotoxic T-lymphocyte level within which a bona fide memory cell precursor has emerged. The challenge now is to identify the cellular and molecular factors that control this developmental pathway. This review considers aspects of the regulation of the induction of effectors, the transition of effectors to memory cells, and the dynamics of the memory population.


Subject(s)
Cytotoxicity, Immunologic , Immunologic Memory , Lymphocyte Activation , T-Lymphocytes, Cytotoxic/immunology , Animals , Cell Differentiation , Cell Proliferation , Communicable Diseases/immunology , Cytokines/metabolism , Humans , Signal Transduction
14.
J Immunol ; 187(10): 4967-78, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21987662

ABSTRACT

In response to infection, CD8(+) T cells integrate multiple signals and undergo an exponential increase in cell numbers. Simultaneously, a dynamic differentiation process occurs, resulting in the formation of short-lived effector cells (SLECs; CD127(low)KLRG1(high)) and memory precursor effector cells (CD127(high)KLRG1(low)) from an early effector cell that is CD127(low)KLRG1(low) in phenotype. CD8(+) T cell differentiation during vesicular stomatitis virus infection differed significantly than during Listeria monocytogenes infection with a substantial reduction in early effector cell differentiation into SLECs. SLEC generation was dependent on Ebi3 expression. Furthermore, SLEC differentiation during vesicular stomatitis virus infection was enhanced by administration of CpG-DNA, through an IL-12-dependent mechanism. Moreover, CpG-DNA treatment enhanced effector CD8(+) T cell functionality and memory subset distribution, but in an IL-12-independent manner. Population dynamics were dramatically different during secondary CD8(+) T cell responses, with a much greater accumulation of SLECs and the appearance of a significant number of CD127(high)KLRG1(high) memory cells, both of which were intrinsic to the memory CD8(+) T cell. These subsets persisted for several months but were less effective in recall than memory precursor effector cells. Thus, our data shed light on how varying the context of T cell priming alters downstream effector and memory CD8(+) T cell differentiation.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Differentiation/immunology , Listeriosis/immunology , Vesicular Stomatitis/immunology , Vesicular Stomatitis/pathology , Animals , Cytotoxicity, Immunologic , Female , Immunologic Memory , Inflammation/microbiology , Inflammation/virology , Listeriosis/pathology , Lymphocyte Activation/immunology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Ovalbumin/administration & dosage , Ovalbumin/immunology
15.
Proc Natl Acad Sci U S A ; 107(1): 193-8, 2010 Jan 05.
Article in English | MEDLINE | ID: mdl-19966302

ABSTRACT

Both CD4(+) T cell help and IL-2 have been postulated to "program" activated CD8(+) T cells for memory cell development. However, the linkage between these two signals has not been well elucidated. Here we have studied effector and memory CD8(+) T cell differentiation following infection with three pathogens (Listeria monocytogenes, vesicular stomatitis virus, and vaccinia virus) in the absence of both CD4(+) T cells and IL-2 signaling. We found that expression of CD25 on antigen-specific CD8(+) T cells peaked 3-4 days after initial priming and was dependent on CD4(+) T cell help, likely through a CD28:CD80/86 mediated pathway. CD4(+) T cell or CD25-deficiency led to normal early effector CD8(+) T cell differentiation, but a subsequent lack of accumulation of CD8(+) T cells resulting in overall decreased memory cell generation. Interestingly, in both primary and recall responses KLRG1(high) CD127(low) short-lived effector cells were drastically diminished in the absence of IL-2 signaling, although memory precursors remained intact. In contrast to previous reports, upon secondary antigen encounter CD25-deficient CD8(+) T cells were capable of undergoing robust expansion, but short-lived effector development was again impaired. Thus, these results demonstrated that CD4(+) T cell help and IL-2 signaling were linked via CD25 up-regulation, which controls the expansion and differentiation of antigen-specific effector CD8(+) T cells, rather than "programming" memory cell traits.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immune System/immunology , Interleukin-2 Receptor alpha Subunit/immunology , Adoptive Transfer , Animals , Cell Differentiation/immunology , Immunologic Memory/immunology , Interleukin-2/immunology , Listeria monocytogenes/immunology , Mice , Mice, Knockout , Signal Transduction/physiology , Vaccinia virus/immunology , Vesiculovirus/immunology
16.
Front Allergy ; 4: 1098474, 2023.
Article in English | MEDLINE | ID: mdl-37168500

ABSTRACT

Background: Mast cells are the major effector cell type for IgE-mediated allergic reactions. Recent studies revealed a role for mast cells in orchestrating the host response to viral infections. Objective: We studied the relationship between FcεRI (high-affinity IgE receptor) and RIG-I-like receptor (RLR)-mediated antiviral signaling pathways. Methods: Mast cells (BMMCs) were cultured from bone marrow cells from mice deficient in MAVS or other RLR signaling molecules. MAVS expression was restored by retroviral transduction of MAVS-deficient BMMCs. These cells were stimulated with IgE and antigen and their activation (degranulation and cytokine production/secretion) was quantified. FcεRI-mediated signaling events such as protein phosphorylation and Ca2+ flux were analyzed by western blotting and enzyme assays. WT and mutant mice as well as mast cell-deficient KitW-sh/W-sh mice engrafted with BMMCs were subjected to passive cutaneous anaphylaxis. Results: Unexpectedly, we found that mast cells devoid of the adaptor molecule MAVS exhibit dramatically increased cytokine production upon FcεRI stimulation, despite near-normal degranulation. Consistent with these observations, MAVS inhibited tyrosine phosphorylation, thus catalytic activity of Syk kinase, the key signaling molecule for FcεRI-mediated mast cell activation. By contrast, mast cells deficient in RIG-I, MDA5 or IRF3, which are antiviral receptor and signaling molecules upstream or downstream of MAVS, exhibited reduced or normal mast cell activation. MAVS-deficient mice showed enhanced late-phase responses in passive cutaneous anaphylaxis. Conclusion: This study demonstrates that the adaptor MAVS in the RLR innate immune pathway uniquely intersects with the adaptive immune FcεRI signaling pathway.

17.
J Immunol ; 185(1): 263-72, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20519649

ABSTRACT

The CD8(+) T cell response to infection is characterized by the appearance of short-lived (CD127(low) killer cell lectin-like receptor G 1-high) and memory-precursor (CD127(high) killer cell lectin-like receptor G 1-low) effector cells. How and when central-memory T (T(CM); CD62L(high) CCR7(+)) cell and effector-memory T(T(EM); CD62L(low) CCR7(-)) cell subsets are established remains unclear. We now show that the T(CM) cell lineage represents an early developmental branchpoint during the CD8(+) T cell response to infection. Central-memory CD8(+) T cells could be identified prior to the peak of the CD8(+) T cell response and were enriched in lymphoid organs. Moreover, the kinetics and magnitude of T(CM) cell development were dependent on the infectious agent. Furthermore, the extent of early Ag availability, which regulated programmed death-1 and CD25 expression levels, controlled the T(CM)/T(EM) cell lineage decision ultimately through IL-2 and IL-15 signaling levels. These observations identify key early signals that help establish the T(CM)/T(EM) cell dichotomy and provide the means to manipulate memory lineage choices.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Immunologic Memory , Lymphocyte Activation/immunology , Signal Transduction/immunology , T-Lymphocyte Subsets/immunology , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/microbiology , Cell Lineage/genetics , Cell Lineage/immunology , Female , Gene Expression Regulation/immunology , L-Selectin/biosynthesis , L-Selectin/genetics , Listeriosis/immunology , Listeriosis/metabolism , Listeriosis/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Radiation Chimera/immunology , T-Lymphocyte Subsets/microbiology , T-Lymphocyte Subsets/pathology , Vesicular Stomatitis/immunology , Vesicular Stomatitis/metabolism , Vesicular Stomatitis/pathology
18.
mBio ; 13(6): e0285422, 2022 12 20.
Article in English | MEDLINE | ID: mdl-36377895

ABSTRACT

Aspergillus fumigatus is a human fungal pathogen that is most often avirulent in immunecompetent individuals because the innate immune system is efficient at eliminating fungal conidia. However, recent clinical observations have shown that severe influenza A virus (IAV) infection can lead to secondary A. fumigatus infections with high mortality. Little is currently known about how IAV infection alters the innate antifungal immune response. Here, we established a murine model of IAV-induced A. fumigatus (IAV-Af) superinfection by inoculating mice with IAV followed 6 days later by A. fumigatus conidia challenge. We observed increased mortality in the IAV-Af-superinfected mice compared to mice challenged with either IAV or A. fumigatus alone. A. fumigatus conidia were able to germinate and establish a biofilm in the lungs of the IAV-Af superinfection group, which was not seen following fungal challenge alone. While we did not observe any differences in inflammatory cell recruitment in the IAV-Af superinfection group compared to single-infection controls, we observed defects in Aspergillus conidial uptake and killing by both neutrophils and monocytes after IAV infection. pHrodo Green zymosan bioparticle (pHrodo-zymosan) and CM-H2DCFDA [5-(and-6)-chloromethyl-2',7'-dichlorodihydrofluorescein diacetate] staining, indicators of phagolysosome maturation and reactive oxygen species (ROS) production, respectively, revealed that the fungal killing defect was due in part to reduced phagolysosome maturation. Collectively, our data demonstrate that the ability of neutrophils and monocytes to kill and clear Aspergillus conidia is strongly reduced in the pulmonary environment of an IAV-infected lung, which leads to invasive pulmonary aspergillosis and increased overall mortality in our mouse model, recapitulating what is observed clinically in humans. IMPORTANCE Influenza A virus (IAV) is a common respiratory virus that causes seasonal illness in humans, but can cause pandemics and severe infection in certain patients. Since the emergence of the 2009 H1N1 pandemic strains, there has been an increase in clinical reports of IAV-infected patients in the intensive care unit (ICU) developing secondary pulmonary aspergillosis. These cases of flu-Aspergillus superinfections are associated with worse clinical outcomes than secondary bacterial infections in the setting of IAV. To date, we have a limited understanding of the cause(s) of secondary fungal infections in immunocompetent hosts. IAV-induced modulation of cytokine production and innate immune cellular function generates a unique immune environment in the lung, which could make the host vulnerable to a secondary fungal infection. Our work shows that defects in phagolysosome maturation in neutrophils and monocytes after IAV infection impair the ability of these cells to kill A. fumigatus, thus leading to increased fungal germination and growth and subsequent invasive aspergillosis. Our work lays a foundation for future mechanistic studies examining the exact immune modulatory events occurring in the respiratory tract after viral infection leading to secondary fungal infections.


Subject(s)
Aspergillosis , Influenza A Virus, H1N1 Subtype , Superinfection , Humans , Animals , Mice , Aspergillus fumigatus , Spores, Fungal , Zymosan , Aspergillosis/microbiology , Aspergillus
19.
Int Immunol ; 22(8): 619-25, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20504887

ABSTRACT

Understanding the regulation of the CD8(+) T-cell response and how protective memory cells are generated has been intensely studied. It is now appreciated that a naive CD8(+) T cell requires at least three signals to mount an effective immune response: (i) TCR triggering, (ii) co-stimulation and (iii) inflammatory cytokines. Only recently have we begun to understand the molecular integration of those signals and how early events regulate the fate decisions of the responding CD8(+) T cells. This review will discuss the recent findings about both the extracellular and intracellular factors that regulate the destiny of responding CD8(+) T cells.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation , Animals , Humans , Immunologic Memory , Models, Biological
20.
Front Immunol ; 12: 675294, 2021.
Article in English | MEDLINE | ID: mdl-34322116

ABSTRACT

Aspergillus fumigatus airway infections are associated with increased rates of hospitalizations and declining lung function in patients with chronic lung disease. While the pathogenesis of invasive A. fumigatus infections is well studied, little is known about the development and progression of airway infections. Previous studies have demonstrated a critical role for the IL-1 cytokines, IL-1α and IL-1ß in enhancing pulmonary neutrophil recruitment during invasive aspergillosis. Here we use a mouse model of A. fumigatus airway infection to study the role of these IL-1 cytokines in immunocompetent mice. In the absence of IL-1 receptor signaling, mice exhibited reduced numbers of viable pulmonary neutrophils and increased levels of neutrophil apoptosis during fungal airway infection. Impaired neutrophil viability in these mice was associated with reduced pulmonary and systemic levels of G-CSF, and treatment with G-CSF restored both neutrophil viability and resistance to A. fumigatus airway infection. Taken together, these data demonstrate that IL-1 dependent G-CSF production plays a key role for host resistance to A. fumigatus airway infection through suppressing neutrophil apoptosis at the site of infection.


Subject(s)
Aspergillosis/immunology , Aspergillus fumigatus/pathogenicity , Lung/immunology , Neutrophils/physiology , Pulmonary Aspergillosis/immunology , Receptors, Interleukin-1/physiology , Animals , Apoptosis/immunology , Chemokines/metabolism , Granulocyte Colony-Stimulating Factor/metabolism , Humans , Interleukin-1alpha , Interleukin-1beta , Lung/pathology , Macrophages , Mice , Mice, Inbred C57BL , Neutrophil Infiltration , Neutrophils/immunology
SELECTION OF CITATIONS
SEARCH DETAIL