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










Publication year range
1.
Front Immunol ; 11: 1959, 2020.
Article in English | MEDLINE | ID: mdl-32849655

ABSTRACT

The lung is the vital target organ of coronavirus disease 2019 (COVID-19) caused by infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In the majority of patients the most active virus replication seems to be found in the upper respiratory tract, severe cases however suffer from SARS-like disease associated with virus replication in lung tissues. Due to the current lack of suitable anti-viral drugs the induction of protective immunity such as neutralizing antibodies in the lung is the key aim of the only alternative approach-the development and application of SARS-CoV-2 vaccines. However, past experience from experimental animals, livestock, and humans showed that induction of immunity in the lung is limited following application of vaccines at peripheral sides such as skin or muscles. Based on several considerations we therefore propose here to consider the application of a Modified Vaccinia virus Ankara (MVA)-based vaccine to mucosal surfaces of the respiratory tract as a favorable approach to combat COVID-19.


Subject(s)
Betacoronavirus/chemistry , Coronavirus Infections/prevention & control , Pandemics/prevention & control , Pneumonia, Viral/prevention & control , Spike Glycoprotein, Coronavirus/immunology , Vaccinia virus/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/immunology , Administration, Mucosal , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Bronchi/immunology , COVID-19 , Coronavirus Infections/virology , Humans , Immunoglobulin A/metabolism , Lymphoid Tissue/immunology , Plasma Cells/immunology , Pneumonia, Viral/virology , Respiratory Mucosa/drug effects , Respiratory Mucosa/immunology , SARS-CoV-2 , T-Lymphocytes/immunology , Vaccination , Vaccines, Attenuated/immunology
2.
Front Immunol ; 10: 1082, 2019.
Article in English | MEDLINE | ID: mdl-31156635

ABSTRACT

Implant associated infections represent a serious health burden in clinics since some microorganisms are able to colonize biological surfaces or surfaces of indwelling medical devices and form biofilms. Biofilms represent communities of microorganisms attached to hydrated surfaces and enclosed in self-produced extracellular matrix. This renders them resistant to exogenous assaults like antibiotics or immune effector mechanisms. Little is known regarding the role of the immune system in the formation of biofilms during implant associated infections, largely due to the lack of suitable mouse models. Here we use colonized osmotic pumps in mice to study the interaction of an activated immune system with biofilm-forming Staphylococcus aureus encoding Gaussia luciferase. This approach permits biofilm formation on the osmotic pumps in living animals. It also allows the continuous supply of soluble immune cell activating agents, such as cytokines to study their effect on biofilm formation in vivo. Using non-invasive imaging of the bioluminescent signal emitted by the lux expressing bacteria for quantification of bacterial load in conjunction with light and electron microscopy, we observed that pump-supplied pro-inflammatory cytokine IL-1ß strongly increased biofilm formation along with a massive influx of neutrophils adjacent to the biofilm-coated pumps. Thus, our data demonstrate that immune defense mechanisms can augment biofilm formation.


Subject(s)
Biofilms/growth & development , Inflammation/immunology , Infusion Pumps, Implantable/microbiology , Interleukin-1beta/metabolism , Neutrophils/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/physiology , Animals , Bacterial Load , Female , Humans , Immune System Diseases , Interleukin-1beta/immunology , Leukocyte Disorders , Mice , Mice, Inbred C57BL , Models, Animal
3.
Cell Rep ; 23(3): 783-795, 2018 Apr 17.
Article in English | MEDLINE | ID: mdl-29669284

ABSTRACT

The processes underlying the development and maintenance of tertiary lymphoid organs are incompletely understood. Using a Ccr7 knockout/knockin approach, we show that spontaneous bronchus-associated lymphoid tissue (BALT) formation can be caused by CCR7-mediated migration defects of dendritic cells (DCs) in the lung. Plt/plt mice that lack the CCR7 ligands CCL19 and CCL21-serine do not form BALT spontaneously because lung-expressed CCL21-leucine presumably suffices to maintain steady-state DC egress. However, plt/plt mice are highly susceptible to modified vaccinia virus infection, showing enhanced recruitment of immune cells as well as alterations in CCR7-ligand-mediated lymphocyte egress from the lungs, leading to dramatically enhanced BALT. Furthermore, we identify two independent BALT homing routes for blood-derived lymphocytes. One is HEV mediated and depends on CCR7 and L-selectin, while the second route is via the lung parenchyma and is independent of these molecules. Together, these data provide insights into CCR7/CCR7-ligand-orchestrated aspects in BALT formation.


Subject(s)
Bronchi/cytology , Chemokine CCL19/metabolism , Chemokine CCL21/metabolism , Lymphocytes/immunology , Receptors, CCR7/metabolism , Animals , Antibodies, Monoclonal/immunology , Bone Marrow Cells/cytology , Chemokine CCL19/deficiency , Chemokine CCL19/genetics , Dendritic Cells/cytology , Dendritic Cells/metabolism , L-Selectin/immunology , L-Selectin/metabolism , Ligands , Lung/cytology , Lung/immunology , Lung/metabolism , Lymphocytes/cytology , Lymphocytes/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Receptors, CCR7/deficiency , Receptors, CCR7/genetics , Vaccinia virus/physiology
4.
Cell Mol Immunol ; 15(10): 875-887, 2018 10.
Article in English | MEDLINE | ID: mdl-29429996

ABSTRACT

Bronchus-associated lymphoid tissue (BALT) develops at unpredictable locations around lung bronchi following pulmonary inflammation. The formation and composition of BALT have primarily been investigated by immunohistology that, due to the size of the invested organ, is usually restricted to a limited number of histological sections. To assess the entire BALT of the lung, other approaches are urgently needed. Here, we introduce a novel light sheet microscopy-based approach for assessing lymphoid tissue in the lung. Using antibody staining of whole lung lobes and optical clearing by organic solvents, we present a method that allows in-depth visualization of the entire bronchial tree, the lymphatic vasculature and the immune cell composition of the induced BALT. Furthermore, three-dimensional analysis of the entire lung allows the qualitative and quantitative enumeration of the induced BALT. Using this approach, we show that a single intranasal application of the replication-deficient poxvirus MVA induces BALT that constitutes up to 8% of the entire lung volume in mice deficient in CCR7, in contrast to wild type mice (WT). Furthermore, BALT induced by heat-inactivated E. coli is dominated by a pronounced T cell infiltration in Cxcr5-deficient mice, in contrast to WT mice.


Subject(s)
Bronchi , Lung , Lymphoid Tissue , Animals , Bronchi/cytology , Bronchi/immunology , Lung/cytology , Lung/immunology , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Mice , Mice, Knockout , Microscopy
5.
Methods Mol Biol ; 1559: 185-198, 2017.
Article in English | MEDLINE | ID: mdl-28063045

ABSTRACT

Bronchus-associated lymphoid tissue (BALT) forms spontaneously in the lung after pulmonary infection and has been identified as a highly organized lymphoid structure supporting the efficient priming of T cells in the lung. To explore the mechanisms and instructive signals controlling BALT neogenesis we used both, a single dose of vaccinia virus MVA and repeated inhalations of heat-inactivated Pseudomonas aeruginosa (P. aeruginosa). Intranasal administration of both pathogens induces highly organized BALT but distinct pathways and molecules are used to promote the development of BALT. Here, we describe the induction and phenotype of the distinct types of BALT as well as the immunofluorescence microscopy-based analysis of the induced lymphoid tissue in the lung.


Subject(s)
Bronchi/pathology , Lymphoid Tissue/pathology , Microscopy, Fluorescence/methods , Microtomy/methods , Pseudomonas aeruginosa/immunology , Vaccinia virus/immunology , Administration, Intranasal , Animals , Biomarkers/metabolism , Bronchi/immunology , Bronchi/microbiology , Bronchi/virology , Chemokine CXCL12/genetics , Chemokine CXCL12/immunology , Chemokine CXCL13/genetics , Chemokine CXCL13/immunology , Gene Expression , Hot Temperature , Interleukin-17/deficiency , Interleukin-17/genetics , Interleukin-17/immunology , Lymphoid Tissue/immunology , Lymphoid Tissue/microbiology , Lymphoid Tissue/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Virus Inactivation
6.
J Exp Med ; 211(4): 643-51, 2014 Apr 07.
Article in English | MEDLINE | ID: mdl-24663215

ABSTRACT

Ectopic lymphoid tissue, such as bronchus-associated lymphoid tissue (BALT) in the lung, develops spontaneously at sites of chronic inflammation or during infection. The molecular mechanisms underlying the neogenesis of such tertiary lymphoid tissue are still poorly understood. We show that the type of inflammation-inducing pathogen determines which key factors are required for the formation and maturation of BALT. Thus, a single intranasal administration of the poxvirus modified vaccinia virus Ankara (MVA) is sufficient to induce highly organized BALT with densely packed B cell follicles containing a network of CXCL13-expressing follicular DCs (FDCs), as well as CXCL12-producing follicular stromal cells. In contrast, mice treated with P. aeruginosa (P.a.) develop BALT but B cell follicles lack FDCs while still harboring CXCL12-positive follicular stromal cells. Furthermore, in IL-17-deficient mice, P.a.-induced BALT largely lacks B cells as well as CXCL12-expressing stromal cells, and only loose infiltrates of T cells are present. We show that Toll-like receptor pathways are required for BALT induction by P.a., but not MVA, and provide evidence that IL-17 drives the differentiation of lung stroma toward podoplanin-positive CXCL12-expressing cells that allow follicle formation even in the absence of FDCs. Taken together, our results identify distinct pathogen-dependent induction and maturation pathways for BALT formation.


Subject(s)
B-Lymphocytes/immunology , Bronchi/pathology , Cell Differentiation , Chemokine CXCL12/metabolism , Dendritic Cells, Follicular/cytology , Interleukin-17/metabolism , Lymphoid Tissue/immunology , Adaptor Proteins, Vesicular Transport/metabolism , Animals , B-Lymphocytes/cytology , Cell Differentiation/immunology , Chick Embryo , Dendritic Cells, Follicular/immunology , Lymphoid Tissue/microbiology , Lymphoid Tissue/pathology , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/metabolism , Pseudomonas Infections/immunology , Pseudomonas Infections/microbiology , Pseudomonas Infections/pathology , Pseudomonas aeruginosa/physiology , Receptors, CXCR4/metabolism , Signal Transduction , Stromal Cells/metabolism , Up-Regulation
7.
Eur J Immunol ; 44(5): 1320-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24500801

ABSTRACT

αß T-cell development and selection proceed while thymocytes successively migrate through distinct regions of the thymus. For γδ T cells, the interplay of intrathymic migration and cell differentiation is less well understood. Here, we crossed C-C chemokine receptor (CCR)7-deficient (Ccr7(-/-) ) and CCR9-deficient mice (Ccr9(-/-) ) to mice with a TcrdH2BeGFP reporter background to investigate the impact of thymic localization on γδ T-cell development. γδ T-cell frequencies and numbers were decreased in CCR7-deficient and increased in CCR9-deficient mice. Transfer of CCR7- or CCR9-deficient BM into irradiated C57BL/6 WT recipients reproduced these phenotypes, pointing toward cell-intrinsic migration defects. Monitoring recent thymic emigrants by intrathymic labeling allowed us to identify decreased thymic γδ T-cell output in CCR7-deficient mice. In vitro, CCR7-deficient precursors showed normal γδ T-cell development. Immunohistology revealed that CCR7 and CCR9 expression was important for γδ T-cell localization within thymic medulla or cortex, respectively. However, γδ T-cell motility was unaltered in CCR7- or CCR9-deficient thymi. Together, our results suggest that proper intrathymic localization is important for normal γδ T-cell development.


Subject(s)
Cell Movement/physiology , Receptors, Antigen, T-Cell, gamma-delta/immunology , Receptors, CCR7/immunology , T-Lymphocytes/immunology , Thymus Gland/immunology , Animals , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Mice , Mice, Knockout , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, CCR/biosynthesis , Receptors, CCR/genetics , Receptors, CCR/immunology , Receptors, CCR7/biosynthesis , Receptors, CCR7/genetics , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism
9.
J Immunol ; 186(5): 2918-25, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21270406

ABSTRACT

Mouse CMV (MCMV) infection rapidly induces the proliferation of NK cells, which correlates with immunological protection. Whether NK cells primed during acute response against MCMV are maintained for the long term is not known. In this study, we used TcrdH2BeGFP mice in which maturing NK cells are genetically labeled with a pulse of very stable histone-2B-eGFP. In this system, we found that the reporter protein was diluted out upon NK cell division during acute MCMV infection. At the same time, mature NK cells in uninfected mice showed only very limited turnover in vivo. Three months after primary infection when MCMV latency was established, the majority of peripheral NK cells still displayed a higher record of proliferation than NK cells in mock-infected controls. This observation included both Ly49H(+) and Ly49H(-) NK cells. Conversely, naive NK cells did not show more proliferation after transfer into latently MCMV-infected mice than that after transfer into mock-infected control mice. This indicated that the observed alterations of the NK cell compartment in MCMV latency were "legacy" (i.e., resulting from prior events during the initial immune response). Together, these results suggest that antiviral immune responses induce sustained alterations of innate lymphocyte populations that extend far beyond the first days of acute infection.


Subject(s)
Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , Immunity, Innate/genetics , Killer Cells, Natural/immunology , Killer Cells, Natural/virology , Virus Latency/immunology , Acute Disease , Animals , Cytomegalovirus Infections/pathology , Green Fluorescent Proteins/genetics , Histones/genetics , Killer Cells, Natural/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muromegalovirus/genetics , Muromegalovirus/immunology , NK Cell Lectin-Like Receptor Subfamily A/biosynthesis , NK Cell Lectin-Like Receptor Subfamily A/deficiency , NK Cell Lectin-Like Receptor Subfamily A/physiology , Virus Latency/genetics
10.
J Exp Med ; 206(12): 2593-601, 2009 Nov 23.
Article in English | MEDLINE | ID: mdl-19917776

ABSTRACT

Mucosal vaccination via the respiratory tract can elicit protective immunity in animal infection models, but the underlying mechanisms are still poorly understood. We show that a single intranasal application of the replication-deficient modified vaccinia virus Ankara, which is widely used as a recombinant vaccination vector, results in prominent induction of bronchus-associated lymphoid tissue (BALT). Although initial peribronchiolar infiltrations, characterized by the presence of dendritic cells (DCs) and few lymphocytes, can be found 4 d after virus application, organized lymphoid structures with segregated B and T cell zones are first observed at day 8. After intratracheal application, in vitro-differentiated, antigen-loaded DCs rapidly migrate into preformed BALT and efficiently activate antigen-specific T cells, as revealed by two-photon microscopy. Furthermore, the lung-specific depletion of DCs in mice that express the diphtheria toxin receptor under the control of the CD11c promoter interferes with BALT maintenance. Collectively, these data identify BALT as tertiary lymphoid structures supporting the efficient priming of T cell responses directed against unrelated airborne antigens while crucially requiring DCs for its sustained presence.


Subject(s)
Bronchi/immunology , Dendritic Cells/immunology , Lymphoid Tissue/immunology , T-Lymphocytes/immunology , Vaccination , Vaccinia virus/immunology , Animals , Antigens, Viral/immunology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Bronchi/cytology , CD11c Antigen/immunology , Cell Movement/immunology , Dendritic Cells/cytology , Lymphoid Tissue/cytology , Mice , Mice, Knockout , T-Lymphocytes/cytology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...