Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Front Immunol ; 14: 1227175, 2023.
Article in English | MEDLINE | ID: mdl-38094305

ABSTRACT

Single-cell RNA sequencing (scRNA-seq) is the state-of-the-art approach to study transcriptomic signatures in individual cells in respiratory health and disease. However, classical scRNA-seq approaches provide no spatial information and are performed using either bronchoalveolar lavage fluid (BAL) or lung single cell suspensions to assess transcript levels in airway and tissue immune cells, respectively. Herein we describe a simple method to simultaneously characterize transcriptomic features of airway, lung parenchymal and intravascular immune cells based on differential in vivo labeling with barcoded antibodies. In addition to gaining basic spatial information, this approach allows for direct comparison of cells within different anatomical compartments. Furthermore, this method provides a time- and cost-effective alternative to classical scRNA-seq where lung and BAL samples are processed individually, reducing animal and reagent use. We demonstrate the feasibility of this approach in a preclinical mouse model of bacterial lung infection comparing airway, parenchymal and vasculature neutrophils early after infection.


Subject(s)
Lung , Pneumonia , Mice , Animals , Bronchoalveolar Lavage Fluid , Gene Expression Profiling , Neutrophils
2.
Cell ; 186(3): 621-645.e33, 2023 02 02.
Article in English | MEDLINE | ID: mdl-36736301

ABSTRACT

Inborn errors of human IFN-γ-dependent macrophagic immunity underlie mycobacterial diseases, whereas inborn errors of IFN-α/ß-dependent intrinsic immunity underlie viral diseases. Both types of IFNs induce the transcription factor IRF1. We describe unrelated children with inherited complete IRF1 deficiency and early-onset, multiple, life-threatening diseases caused by weakly virulent mycobacteria and related intramacrophagic pathogens. These children have no history of severe viral disease, despite exposure to many viruses, including SARS-CoV-2, which is life-threatening in individuals with impaired IFN-α/ß immunity. In leukocytes or fibroblasts stimulated in vitro, IRF1-dependent responses to IFN-γ are, both quantitatively and qualitatively, much stronger than those to IFN-α/ß. Moreover, IRF1-deficient mononuclear phagocytes do not control mycobacteria and related pathogens normally when stimulated with IFN-γ. By contrast, IFN-α/ß-dependent intrinsic immunity to nine viruses, including SARS-CoV-2, is almost normal in IRF1-deficient fibroblasts. Human IRF1 is essential for IFN-γ-dependent macrophagic immunity to mycobacteria, but largely redundant for IFN-α/ß-dependent antiviral immunity.


Subject(s)
COVID-19 , Mycobacterium , Child , Humans , Interferon-gamma , SARS-CoV-2 , Interferon-alpha , Interferon Regulatory Factor-1
3.
Life Sci Alliance ; 6(4)2023 04.
Article in English | MEDLINE | ID: mdl-36754569

ABSTRACT

ICOS is a T-cell costimulatory receptor critical for Tfh cell generation and function. However, the role of ICOS in Tfr cell differentiation remains unclear. Using Foxp3-Cre-mediated ICOS knockout (ICOS FC) mice, we show that ICOS deficiency in Treg-lineage cells drastically reduces the number of Tfr cells during GC reactions but has a minimal impact on conventional Treg cells. Single-cell transcriptome analysis of Foxp3+ cells at an early stage of the GC reaction suggests that ICOS normally inhibits Klf2 expression to promote follicular features including Bcl6 up-regulation. Furthermore, ICOS costimulation promotes nuclear localization of NFAT2, a known driver of CXCR5 expression. Notably, ICOS FC mice had an unaltered overall GC B-cell output but showed signs of expanded autoreactive B cells along with elevated autoantibody titers. Thus, our study demonstrates that ICOS costimulation is critical for Tfr cell differentiation and highlights the importance of Tfr cells in maintaining humoral immune tolerance during GC reactions.


Subject(s)
Germinal Center , T-Lymphocytes, Regulatory , Animals , Mice , T-Lymphocytes, Regulatory/metabolism , B-Lymphocytes , Cell Differentiation/genetics , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Inducible T-Cell Co-Stimulator Protein/genetics , Inducible T-Cell Co-Stimulator Protein/metabolism
4.
Antibiotics (Basel) ; 11(4)2022 Apr 01.
Article in English | MEDLINE | ID: mdl-35453224

ABSTRACT

Acute and chronic lower airway disease still represent a major cause of morbidity and mortality on a global scale. With the steady rise of multidrug-resistant respiratory pathogens, such as Pseudomonas aeruginosa and Klebsiella pneumoniae, we are rapidly approaching the advent of a post-antibiotic era. In addition, potentially detrimental novel variants of respiratory viruses continuously emerge with the most prominent recent example being severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). To this end, alternative preventive and therapeutic intervention strategies will be critical to combat airway infections in the future. Chronic respiratory diseases are associated with alterations in the lung and gut microbiome, which is thought to contribute to disease progression and increased susceptibility to infection with respiratory pathogens. In this review we will focus on how modulating and harnessing the microbiome may pose a novel strategy to prevent and treat pulmonary infections as well as chronic respiratory disease.

5.
Front Immunol ; 12: 664218, 2021.
Article in English | MEDLINE | ID: mdl-34867937

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) play a key role in the initiation and orchestration of early type 2 immune responses. Upon tissue damage, ILC2s are activated by alarmins such as IL-33 and rapidly secrete large amounts of type 2 signature cytokines. ILC2 activation is governed by a network of transcriptional regulators including nuclear factor (NF)-κB family transcription factors. While it is known that activating IL-33 receptor signaling results in downstream NF-κB activation, the underlying molecular mechanisms remain elusive. Here, we found that the NF-κB subunit c-Rel is required to mount effective innate pulmonary type 2 immune responses. IL-33-mediated activation of ILC2s in vitro as well as in vivo was found to induce c-Rel mRNA and protein expression. In addition, we demonstrate that IL-33-mediated activation of ILC2s leads to nuclear translocation of c-Rel in pulmonary ILC2s. Although c-Rel was found to be a critical mediator of innate pulmonary type 2 immune responses, ILC2-intrinsic deficiency of c-Rel did not have an impact on the developmental capacity of ILC2s nor affected homeostatic numbers of lung-resident ILC2s at steady state. Moreover, we demonstrate that ILC2-intrinsic deficiency of c-Rel alters the capacity of ILC2s to upregulate the expression of ICOSL and OX40L, key stimulatory receptors, and the expression of type 2 signature cytokines IL-5, IL-9, IL-13, and granulocyte-macrophage colony-stimulating factor (GM-CSF). Collectively, our data using Rel-/- mice suggest that c-Rel promotes acute ILC2-driven allergic airway inflammation and suggest that c-Rel may contribute to the pathophysiology of ILC2-mediated allergic airway disease. It thereby represents a promising target for the treatment of allergic asthma, and evaluating the effect of established c-Rel inhibitors in this context would be of great clinical interest.


Subject(s)
Immunity, Innate , Lung/immunology , Lymphocyte Subsets/immunology , Proto-Oncogene Proteins c-rel/immunology , Animals , Asthma/immunology , Asthma/pathology , Disease Models, Animal , Female , Gene Expression , In Vitro Techniques , Interleukin-33/immunology , Lung/pathology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Proto-Oncogene Proteins c-rel/deficiency , Proto-Oncogene Proteins c-rel/genetics
6.
Cancer Res ; 81(14): 3876-3889, 2021 07 15.
Article in English | MEDLINE | ID: mdl-33975880

ABSTRACT

Breast cancer diagnosed within 10 years following childbirth is defined as postpartum breast cancer (PPBC) and is highly metastatic. Interactions between immune cells and other stromal cells within the involuting mammary gland are fundamental in facilitating an aggressive tumor phenotype. The MNK1/2-eIF4E axis promotes translation of prometastatic mRNAs in tumor cells, but its role in modulating the function of nontumor cells in the PPBC microenvironment has not been explored. Here, we used a combination of in vivo PPBC models and in vitro assays to study the effects of inactivation of the MNK1/2-eIF4E axis on the protumor function of select cells of the tumor microenvironment. PPBC mice deficient for phospho-eIF4E (eIF4ES209A) were protected against lung metastasis and exhibited differences in the tumor and lung immune microenvironment compared with wild-type mice. Moreover, the expression of fibroblast-derived IL33, an alarmin known to induce invasion, was repressed upon MNK1/2-eIF4E axis inhibition. Imaging mass cytometry on PPBC and non-PPBC patient samples indicated that human PPBC contains phospho-eIF4E high-expressing tumor cells and CD8+ T cells displaying markers of an activated dysfunctional phenotype. Finally, inhibition of MNK1/2 combined with anti-PD-1 therapy blocked lung metastasis of PPBC. These findings implicate the involvement of the MNK1/2-eIF4E axis during PPBC metastasis and suggest a promising immunomodulatory route to enhance the efficacy of immunotherapy by blocking phospho-eIF4E. SIGNIFICANCE: This study investigates the MNK1/2-eIF4E signaling axis in tumor and stromal cells in metastatic breast cancer and reveals that MNK1/2 inhibition suppresses metastasis and sensitizes tumors to anti-PD-1 immunotherapy.


Subject(s)
Breast Neoplasms/drug therapy , Eukaryotic Initiation Factor-4E/therapeutic use , Immunosuppression Therapy/methods , Animals , Disease Models, Animal , Eukaryotic Initiation Factor-4E/pharmacology , Female , Humans , Mice , Neoplasm Metastasis , Postpartum Period
7.
J Allergy Clin Immunol ; 147(6): 2330-2342, 2021 06.
Article in English | MEDLINE | ID: mdl-33453289

ABSTRACT

BACKGROUND: Lung nociceptor neurons amplify immune cell activity and mucus metaplasia in response to an inhaled allergen challenge in sensitized mice. OBJECTIVE: We sought to identify the cellular mechanisms by which these sensory neurons are activated subsequent to allergen exposure. METHODS: We used calcium microscopy and electrophysiologic recording to assess whether vagal neurons directly respond to the model allergen ovalbumin (OVA). Next, we generated the first nociceptor-specific FcεR1γ knockdown (TRPV1Cre::FcεR1γfl/fl) mice to assess whether this targeted invalidation would affect the severity of allergic inflammation in response to allergen challenges. RESULTS: Lung-innervating jugular nodose complex ganglion neurons express the high-affinity IgE receptor FcεR1, the levels of which increase in OVA-sensitized mice. FcεR1γ-expressing vagal nociceptor neurons respond directly to OVA complexed with IgE with depolarization, action potential firing, calcium influx, and neuropeptide release. Activation of vagal neurons by IgE-allergen immune complexes, through the release of substance P from their peripheral terminals, directly amplifies TH2 cell influx and polarization in the airways. Allergic airway inflammation is decreased in TRPV1Cre::FcεR1γfl/fl mice and in FcεR1α-/- mice into which bone marrow has been transplanted. Finally, increased in vivo circulating levels of IgE following allergen sensitization enhances the responsiveness of FcεR1 to immune complexes in both mouse jugular nodose complex ganglion neurons and human induced pluripotent stem cell-derived nociceptors. CONCLUSIONS: Allergen sensitization triggers a feedforward inflammatory loop between IgE-producing plasma cells, FcεR1-expressing vagal sensory neurons, and TH2 cells, which helps to both initiate and amplify allergic airway inflammation. These data highlight a novel target for reducing allergy, namely, FcεR1γ expressed by nociceptors.


Subject(s)
Gene Expression , Hypersensitivity/immunology , Hypersensitivity/metabolism , Receptors, IgE/genetics , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Allergens/immunology , Animals , Calcium/metabolism , Disease Models, Animal , Disease Susceptibility/immunology , Genetic Predisposition to Disease , Hypersensitivity/genetics , Hypersensitivity/pathology , Mice , Mice, Knockout , Neurons/immunology , Neurons/metabolism , Nociceptors/metabolism , Ovalbumin/adverse effects , Ovalbumin/immunology , Receptors, IgE/metabolism , Respiratory Mucosa/pathology , Substance P/metabolism , Vagus Nerve
8.
Sci Immunol ; 4(36)2019 06 07.
Article in English | MEDLINE | ID: mdl-31175178

ABSTRACT

CCL8-CCR8 axis and collagen-I regulate ILC2 cells during lung inflammation.


Subject(s)
Lymphocytes , Pneumonia , Animals , Chemokine CCL8 , Cues , Humans , Immunity, Innate , Lung , Mice
9.
Front Immunol ; 10: 930, 2019.
Article in English | MEDLINE | ID: mdl-31231357

ABSTRACT

Millions of people worldwide are suffering from allergic inflammatory airway disorders. These conditions are regarded as a consequence of multiple imbalanced immune events resulting in an inadequate response with the exact underlying mechanisms still being a subject of ongoing research. Several cell populations have been proposed to be involved but it is becoming increasingly evident that group 2 innate lymphoid cells (ILC2s) play a key role in the initiation and orchestration of respiratory allergic inflammation. ILC2s are important mediators of inflammation but also tissue remodeling by secreting large amounts of signature cytokines within a short time period. Thereby, ILC2s instruct innate but also adaptive immune responses. Here, we will discuss the recent literature on allergic inflammation of the respiratory tract with a focus on ILC2 biology. Furthermore, we will highlight different therapeutic strategies to treat pulmonary allergic inflammation and their potential influence on ILC2 function as well as discuss the perspective of using human ILC2s for diagnostic purposes.


Subject(s)
Lymphocytes/immunology , Respiratory Hypersensitivity/immunology , Animals , Humans
10.
Front Immunol ; 9: 840, 2018.
Article in English | MEDLINE | ID: mdl-29760695

ABSTRACT

Group 2 innate lymphoid cells (ILC2) represent an evolutionary rather old but only recently identified member of the family of innate lymphoid cells and have received much attention since their detailed description in 2010. They can orchestrate innate as well as adaptive immune responses as they interact with and influence several immune and non-immune cell populations. Moreover, ILC2 are able to rapidly secrete large amounts of type 2 cytokines that can contribute to protective but also detrimental host immune responses depending on timing, location, and physiological context. Interestingly, ILC2, despite their scarcity, are the dominant innate lymphoid cell population in the lung, indicating a key role as first responders and amplifiers upon immune challenge at this site. In addition, the recently described tissue residency of ILC2 further underlines the importance of their respective microenvironment. In this review, we provide an overview of lung physiology including a description of the most prominent pulmonary resident cells together with a review of known and potential ILC2 interactions within this unique environment. We will further outline recent observations regarding pulmonary ILC2 during immune challenge including respiratory infections and discuss different models and approaches to study ILC2 biology in the lung.


Subject(s)
Homeostasis , Immunity, Innate , Lung/immunology , Lymphocytes/immunology , Animals , Cellular Microenvironment , Cytokines/immunology , Humans , Mice , Th2 Cells
11.
J Immunol ; 198(1): 71-81, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27881703

ABSTRACT

The regulatory properties of B cells have been studied in autoimmune diseases; however, their role in allergic diseases is poorly understood. We demonstrate that Semaphorin 4C (Sema4C), an axonal guidance molecule, plays a crucial role in B cell regulatory function. Mice deficient in Sema4C exhibited increased airway inflammation after allergen exposure, with massive eosinophilic lung infiltrates and increased Th2 cytokines. This phenotype was reproduced by mixed bone marrow chimeric mice with Sema4C deficient only in B cells, indicating that B lymphocytes were the key cells affected by the absence of Sema4C expression in allergic inflammation. We determined that Sema4C-deficient CD19+CD138+ cells exhibited decreased IL-10 and increased IL-4 expression in vivo and in vitro. Adoptive transfer of Sema4c-/- CD19+CD138+ cells induced marked pulmonary inflammation, eosinophilia, and increased bronchoalveolar lavage fluid IL-4 and IL-5, whereas adoptive transfer of wild-type CD19+CD138+IL-10+ cells dramatically decreased allergic airway inflammation in wild-type and Sema4c-/- mice. This study identifies a novel pathway by which Th2-mediated immune responses are regulated. It highlights the importance of plasma cells as regulatory cells in allergic inflammation and suggests that CD138+ B cells contribute to cytokine balance and are important for maintenance of immune homeostasis in allergic airways disease. Furthermore, we demonstrate that Sema4C is critical for optimal regulatory cytokine production in CD138+ B cells.


Subject(s)
B-Lymphocyte Subsets/immunology , Plasma Cells/immunology , Respiratory Hypersensitivity/immunology , Semaphorins/immunology , Adoptive Transfer , Animals , Blotting, Western , Cytokines/biosynthesis , Flow Cytometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumonia/immunology , Syndecan-1/immunology
12.
J Leukoc Biol ; 101(3): 643-654, 2017 03.
Article in English | MEDLINE | ID: mdl-27895164

ABSTRACT

MYSM1 is a chromatin-binding histone deubiquitinase. MYSM1 mutations in humans result in lymphopenia whereas loss of Mysm1 in mice causes severe hematopoietic abnormalities, including an early arrest in B cell development. However, it remains unknown whether MYSM1 is required at later checkpoints in B cell development or for B cell-mediated immune responses. We analyzed conditional mouse models Mysm1fl/flTg.mb1-cre, Mysm1fl/flTg.CD19-cre, and Mysm1fl/flTg.CD21-cre with inactivation of Mysm1 at prepro-B, pre-B, and follicular B cell stages of development. We show that loss of Mysm1 at the prepro-B cell stage in Mysm1fl/flTg.mb1-cre mice results in impaired B cell differentiation, with an ∼2-fold reduction in B cell numbers in the lymphoid organs. Mysm1fl/flTg.mb1-cre B cells also showed increased expression of activation markers and impaired survival and proliferation. In contrast, Mysm1 was largely dispensable from the pre-B cell stage onward, with Mysm1fl/flTg.CD19-cre and Mysm1fl/flTg.CD21-cre mice showing no alterations in B cell numbers and largely normal responses to stimulation. MYSM1, therefore, has an essential role in B cell lineage specification but is dispensable at later stages of development. Importantly, MYSM1 activity at the prepro-B cell stage of development is important for the normal programming of B cell responses to stimulation once they complete their maturation process.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Cycle Checkpoints , Cell Differentiation , Cell Lineage , Endopeptidases/metabolism , Immunity, Cellular , Animals , Biomarkers/metabolism , Cell Cycle Checkpoints/immunology , Cell Differentiation/immunology , Cell Proliferation , Cell Survival , Immunity, Humoral , Immunoglobulin Class Switching , Integrases/metabolism , Lymphocyte Activation/immunology , Mice, Inbred C57BL , Mice, Knockout , Receptors, Complement 3d/metabolism , Trans-Activators , Ubiquitin-Specific Proteases
13.
Front Immunol ; 7: 564, 2016.
Article in English | MEDLINE | ID: mdl-27994594

ABSTRACT

Elicitation of type I interferon (IFN-I) has been shown to both enhance and impair cell-mediated immune responses in acute and persistent viral infections, respectively. Here, we show that, in addition to its effect on T cells, IFN-I drives impairment of specific antibody responses through interaction with B cells in the acute phase of lymphocytic choriomeningitis virus (LCMV) infection. This impairment was limited to the T cell-dependent B cell response and was associated with disruption of B cell follicles, development of hypergammaglobulinemia (HGG), and expansion of the T follicular helper cell population. Antigen-specific antibody responses were restored by ablation of IFN-I signaling through antibody-mediated IFN-I receptor blockade and B cell-specific IFN-I receptor knockout. Importantly, IFN-I receptor deficiency in B cells also accelerated the development of LCMV neutralizing antibodies and alleviated HGG. These results provide a potential therapeutic target toward efficient treatment measures that limit immunopathology in persistent viral infections.

14.
Nat Immunol ; 17(1): 65-75, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26595887

ABSTRACT

Viral respiratory tract infections are the main causative agents of the onset of infection-induced asthma and asthma exacerbations that remain mechanistically unexplained. Here we found that deficiency in signaling via type I interferon receptor led to deregulated activation of group 2 innate lymphoid cells (ILC2 cells) and infection-associated type 2 immunopathology. Type I interferons directly and negatively regulated mouse and human ILC2 cells in a manner dependent on the transcriptional activator ISGF3 that led to altered cytokine production, cell proliferation and increased cell death. In addition, interferon-γ (IFN-γ) and interleukin 27 (IL-27) altered ILC2 function dependent on the transcription factor STAT1. These results demonstrate that type I and type II interferons, together with IL-27, regulate ILC2 cells to restrict type 2 immunopathology.


Subject(s)
Immunity, Innate/immunology , Interferon Type I/immunology , Lymphocytes/immunology , Respiratory Tract Infections/immunology , Animals , Cytokines/biosynthesis , Cytokines/immunology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/immunology , Real-Time Polymerase Chain Reaction , Respiratory Tract Infections/pathology
15.
Infect Immun ; 83(12): 4541-54, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26371125

ABSTRACT

The genetic basis of natural susceptibility to progressive Cryptococcus neoformans infection is not well understood. Using C57BL/6 and CBA/J inbred mice, we previously identified three chromosomal regions associated with C. neoformans susceptibility (Cnes1, Cnes2, and Cnes3). To validate and characterize the role of Cnes2 during the host response, we constructed a congenic strain on the C57BL/6 background (B6.CBA-Cnes2). Phenotypic analysis of B6.CBA-Cnes2 mice 35 days after C. neoformans infection showed a significant reduction of fungal burden in the lungs and spleen with higher pulmonary expression of gamma interferon (IFN-γ) and interleukin-12 (IL-12), lower expression of IL-4, IL-5, and IL-13, and an absence of airway epithelial mucus production compared to that in C57BL/6 mice. Multiparameter flow cytometry of infected lungs also showed a significantly higher number of neutrophils, exudate macrophages, CD11b(+) dendritic cells, and CD4(+) cells in B6.CBA-Cnes2 than in C57BL/6 mice. The activation state of recruited macrophages and dendritic cells was also significantly increased in B6.CBA-Cnes2 mice. Taken together, these findings demonstrate that the Cnes2 interval is a potent regulator of host defense, immune responsiveness, and differential Th1/Th2 polarization following C. neoformans infection.


Subject(s)
Chromosomes, Mammalian/immunology , Cryptococcosis/genetics , Cryptococcus neoformans/immunology , Genetic Loci/immunology , Genetic Predisposition to Disease , Immunity, Innate , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/microbiology , CD4-Positive T-Lymphocytes/pathology , Chromosomes, Mammalian/chemistry , Crosses, Genetic , Cryptococcosis/immunology , Cryptococcosis/microbiology , Cryptococcosis/pathology , Dendritic Cells/immunology , Dendritic Cells/microbiology , Dendritic Cells/pathology , Gene Expression , Host-Pathogen Interactions , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-12/genetics , Interleukin-12/immunology , Interleukin-13/genetics , Interleukin-13/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Interleukin-5/genetics , Interleukin-5/immunology , Macrophages/immunology , Macrophages/microbiology , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Neutrophils/immunology , Neutrophils/microbiology , Neutrophils/pathology , Phenotype , Th1-Th2 Balance
16.
J Immunol ; 192(6): 2787-99, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24534531

ABSTRACT

Innate immune responses provoke the accumulation of leukocytes at sites of inflammation. In addition to monocytes and granulocytes, B cells also participate in antimicrobial innate immune responses; however, the mechanisms for accumulation of B cells to sites of inflammation are not well understood. To study B cell accumulation following systemic inflammation, we used a model synthetic ligand that stimulates a specific pattern recognition molecule, nucleotide-binding oligomerization domain-containing protein 1 (Nod1). Upon exposure to Nod1 agonists, both B cells and neutrophils rapidly accumulate within the spleen, and dendritic cells migrate into the periarterial lymphoid sheath. Nod1 stimulation led to a marked increase in several chemokines within the spleen, including CXCL13, CCL2, and CCL20. Whereas the lymphotoxin pathway was critical for the induction of the B cell chemoattractant CXCL13 in response to Nod1 agonists, B cell accumulation within the spleen following Nod1-induced systemic inflammation was independent of the lymphotoxin pathway. In contrast, a CCR6/CCL20 chemokine loop instructed rapid increase of B cells in the spleen in response to systemic administration of Nod1 agonists in a TNF-α-dependent manner. Moreover, CCR6 was required to regulate Nod1-mediated B cell responses. These results reveal a novel mechanism of B cells during inflammation and shed light on how B cells participate in innate immune responses to microbial stimulation.


Subject(s)
B-Lymphocytes/immunology , Chemokine CCL20/immunology , Nod1 Signaling Adaptor Protein/immunology , Receptors, CCR6/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Transplantation/methods , Cell Line , Cells, Cultured , Chemokine CCL20/metabolism , Diaminopimelic Acid/analogs & derivatives , Diaminopimelic Acid/pharmacology , Female , Flow Cytometry , Lymphocyte Count , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Lymphoid Tissue/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/metabolism , Nod1 Signaling Adaptor Protein/genetics , Nod1 Signaling Adaptor Protein/metabolism , Receptors, CCR6/genetics , Receptors, CCR6/metabolism , Spleen/cytology , Spleen/immunology , Spleen/metabolism , Transplantation Chimera/blood , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...