Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
1.
Nat Commun ; 13(1): 2027, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35440118

ABSTRACT

Innate lymphoid cells (ILCs) govern immune cell homeostasis in the intestine and protect the host against microbial pathogens. Various cell-intrinsic pathways have been identified that determine ILC development and differentiation. However, the cellular components that regulate ILC sustenance and function in the intestinal lamina propria are less known. Using single-cell transcriptomic analysis of lamina propria fibroblasts, we identify fibroblastic reticular cells (FRCs) that underpin cryptopatches (CPs) and isolated lymphoid follicles (ILFs). Genetic ablation of lymphotoxin-ß receptor expression in Ccl19-expressing FRCs blocks the maturation of CPs into mature ILFs. Interactome analysis shows the major niche factors and processes underlying FRC-ILC crosstalk. In vivo validation confirms that a sustained lymphotoxin-driven feedforward loop of FRC activation including IL-7 generation is critical for the maintenance of functional ILC populations. In sum, our study indicates critical fibroblastic niches within the intestinal lamina propria that control ILC homeostasis and functionality and thereby secure protective gut immunity.


Subject(s)
Immunity, Innate , Lymphocytes , Fibroblasts , Homeostasis , Intestines
2.
Nat Commun ; 12(1): 4734, 2021 08 05.
Article in English | MEDLINE | ID: mdl-34354077

ABSTRACT

The tumor microenvironment (TME) is a complex amalgam of tumor cells, immune cells, endothelial cells and fibroblastic stromal cells (FSC). Cancer-associated fibroblasts are generally seen as tumor-promoting entity. However, it is conceivable that particular FSC populations within the TME contribute to immune-mediated tumor control. Here, we show that intratumoral treatment of mice with a recombinant lymphocytic choriomeningitis virus-based vaccine vector expressing a melanocyte differentiation antigen resulted in T cell-dependent long-term control of melanomas. Using single-cell RNA-seq analysis, we demonstrate that viral vector-mediated transduction reprogrammed and activated a Cxcl13-expressing FSC subset that show a pronounced immunostimulatory signature and increased expression of the inflammatory cytokine IL-33. Ablation of Il33 gene expression in Cxcl13-Cre-positive FSCs reduces the functionality of intratumoral T cells and unleashes tumor growth. Thus, reprogramming of FSCs by a self-antigen-expressing viral vector in the TME is critical for curative melanoma treatment by locally sustaining the activity of tumor-specific T cells.


Subject(s)
Melanoma, Experimental/therapy , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cancer-Associated Fibroblasts/immunology , Cancer-Associated Fibroblasts/pathology , Cellular Reprogramming Techniques/methods , Chemokine CXCL13/genetics , Chemokine CXCL13/immunology , Female , Genetic Vectors , Interleukin-33/deficiency , Interleukin-33/genetics , Interleukin-33/immunology , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/immunology , Lymphocytic choriomeningitis virus/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Stromal Cells/immunology , Stromal Cells/pathology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Tumor Microenvironment/immunology
3.
Nat Immunol ; 22(8): 1042-1051, 2021 08.
Article in English | MEDLINE | ID: mdl-34267375

ABSTRACT

Pathogens and vaccines that produce persisting antigens can generate expanded pools of effector memory CD8+ T cells, described as memory inflation. While properties of inflating memory CD8+ T cells have been characterized, the specific cell types and tissue factors responsible for their maintenance remain elusive. Here, we show that clinically applied adenovirus vectors preferentially target fibroblastic stromal cells in cultured human tissues. Moreover, we used cell-type-specific antigen targeting to define critical cells and molecules that sustain long-term antigen presentation and T cell activity after adenovirus vector immunization in mice. While antigen targeting to myeloid cells was insufficient to activate antigen-specific CD8+ T cells, genetic activation of antigen expression in Ccl19-cre-expressing fibroblastic stromal cells induced inflating CD8+ T cells. Local ablation of vector-targeted cells revealed that lung fibroblasts support the protective function and metabolic fitness of inflating memory CD8+ T cells in an interleukin (IL)-33-dependent manner. Collectively, these data define a critical fibroblastic niche that underpins robust protective immunity operating in a clinically important vaccine platform.


Subject(s)
Adenoviridae/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Interleukin-33/immunology , Lymphocyte Activation/immunology , Stromal Cells/immunology , Adenoviridae/genetics , Animals , Cell Line, Tumor , Chemokine CCL19/metabolism , Chimera/genetics , Epitopes, T-Lymphocyte/immunology , Fibroblasts/cytology , Fibroblasts/metabolism , Genetic Vectors/immunology , Humans , Lung/cytology , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Vaccination
4.
Nat Commun ; 10(1): 1739, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30988302

ABSTRACT

The splenic white pulp is underpinned by poorly characterized stromal cells that demarcate distinct immune cell microenvironments. Here we establish fibroblastic reticular cell (FRC)-specific fate-mapping in mice to define their embryonic origin and differentiation trajectories. Our data show that all reticular cell subsets descend from multipotent progenitors emerging at embryonic day 19.5 from periarterial progenitors. Commitment of FRC progenitors is concluded during the first week of postnatal life through occupation of niches along developing central arterioles. Single cell transcriptomic analysis facilitated deconvolution of FRC differentiation trajectories and indicated that perivascular reticular cells function both as adult lymphoid organizer cells and mural cell progenitors. The lymphotoxin-ß receptor-independent sustenance of postnatal progenitor stemness unveils that systemic immune surveillance in the splenic white pulp is governed through subset specification of reticular cells from a multipotent periarterial progenitor cell. In sum, the finding that discrete signaling events in perivascular niches determine the differentiation trajectories of reticular cell networks explains the development of distinct microenvironmental niches in secondary and tertiary lymphoid tissues that are crucial for the induction and regulation of innate and adaptive immune processes.


Subject(s)
Cell Lineage , Cellular Microenvironment , Fibroblasts/physiology , Animals , Cell Differentiation , Gene Expression Profiling , Immunologic Surveillance , Lymphocytes , Mice , Spleen
5.
Sci Immunol ; 3(26)2018 08 10.
Article in English | MEDLINE | ID: mdl-30097537

ABSTRACT

Immune protection of the body cavities depends on the swift activation of innate and adaptive immune responses in nonclassical secondary lymphoid organs known as fat-associated lymphoid clusters (FALCs). Compared with classical secondary lymphoid organs such as lymph nodes and Peyer's patches, FALCs develop along distinct differentiation trajectories and display a reduced structural complexity. Although it is well established that fibroblastic reticular cells (FRCs) are an integral component of the immune-stimulating infrastructure of classical secondary lymphoid organs, the role of FRCs in FALC-dependent peritoneal immunity remains unclear. Using FRC-specific gene targeting, we found that FRCs play an essential role in FALC-driven immune responses. Specifically, we report that initiation of peritoneal immunity was governed through FRC activation in a myeloid differentiation primary response 88 (MYD88)-dependent manner. FRC-specific ablation of MYD88 blocked recruitment of inflammatory monocytes into FALCs and subsequent CD4+ T cell-dependent B-cell activation and IgG class switching. Moreover, containment of Salmonella infection was compromised in mice lacking MYD88 expression in FRCs, indicating that FRCs in FALCs function as an initial checkpoint in the orchestration of protective immune responses in the peritoneal cavity.


Subject(s)
Fibroblasts/cytology , Fibroblasts/immunology , Intra-Abdominal Fat/immunology , Peritoneal Cavity/physiology , Animals , Chemokine CCL2/immunology , Mice, Inbred C57BL , Mice, Transgenic , Monocytes/immunology , Myeloid Differentiation Factor 88/immunology , Salmonella Infections/immunology , Salmonella typhimurium , Tumor Necrosis Factor-alpha/immunology
6.
Immunity ; 48(1): 120-132.e8, 2018 01 16.
Article in English | MEDLINE | ID: mdl-29343433

ABSTRACT

Group 3 innate lymphoid cells (ILC3s) sense environmental signals and are critical for tissue integrity in the intestine. Yet, which signals are sensed and what receptors control ILC3 function remain poorly understood. Here, we show that ILC3s with a lymphoid-tissue-inducer (LTi) phenotype expressed G-protein-coupled receptor 183 (GPR183) and migrated to its oxysterol ligand 7α,25-hydroxycholesterol (7α,25-OHC). In mice lacking Gpr183 or 7α,25-OHC, ILC3s failed to localize to cryptopatches (CPs) and isolated lymphoid follicles (ILFs). Gpr183 deficiency in ILC3s caused a defect in CP and ILF formation in the colon, but not in the small intestine. Localized oxysterol production by fibroblastic stromal cells provided an essential signal for colonic lymphoid tissue development, and inflammation-induced increased oxysterol production caused colitis through GPR183-mediated cell recruitment. Our findings show that GPR183 promotes lymphoid organ development and indicate that oxysterol-GPR183-dependent positioning within tissues controls ILC3 activity and intestinal homeostasis.


Subject(s)
Colitis/metabolism , Lymphocytes/metabolism , Lymphoid Tissue/metabolism , Oxysterols/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Cell Movement/genetics , Colitis/immunology , Colitis/pathology , Colon/immunology , Colon/pathology , Cytokines/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Ligands , Lymphocytes/pathology , Lymphoid Tissue/pathology , Mice , Real-Time Polymerase Chain Reaction , Signal Transduction
7.
Immunity ; 47(1): 80-92.e4, 2017 07 18.
Article in English | MEDLINE | ID: mdl-28709801

ABSTRACT

Lymph nodes (LNs) are strategically situated throughout the body at junctures of the blood vascular and lymphatic systems to direct immune responses against antigens draining from peripheral tissues. The current paradigm describes LN development as a programmed process that is governed through the interaction between mesenchymal lymphoid tissue organizer (LTo) cells and hematopoietic lymphoid tissue inducer (LTi) cells. Using cell-type-specific ablation of key molecules involved in lymphoid organogenesis, we found that initiation of LN development is dependent on LTi-cell-mediated activation of lymphatic endothelial cells (LECs) and that engagement of mesenchymal stromal cells is a succeeding event. LEC activation was mediated mainly by signaling through receptor activator of NF-κB (RANK) and the non-canonical NF-κB pathway and was steered by sphingosine-1-phosphate-receptor-dependent retention of LTi cells in the LN anlage. Finally, the finding that pharmacologically enforced interaction between LTi cells and LECs promotes ectopic LN formation underscores the central LTo function of LECs.


Subject(s)
Endothelial Cells/physiology , Lymph Nodes/physiology , Mesenchymal Stem Cells/physiology , Organogenesis , Animals , Cell Differentiation , Cells, Cultured , Choristoma , Embryo, Mammalian , Lymphotoxin beta Receptor/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/metabolism , Receptor Activator of Nuclear Factor-kappa B/metabolism , Receptors, Lysosphingolipid/metabolism , Signal Transduction
8.
Nat Immunol ; 17(12): 1388-1396, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27798617

ABSTRACT

Fibroblastic reticular cells (FRCs) of secondary lymphoid organs form distinct niches for interaction with hematopoietic cells. We found here that production of the cytokine IL-15 by FRCs was essential for the maintenance of group 1 innate lymphoid cells (ILCs) in Peyer's patches and mesenteric lymph nodes. Moreover, FRC-specific ablation of the innate immunological sensing adaptor MyD88 unleashed IL-15 production by FRCs during infection with an enteropathogenic virus, which led to hyperactivation of group 1 ILCs and substantially altered the differentiation of helper T cells. Accelerated clearance of virus by group 1 ILCs precipitated severe intestinal inflammatory disease with commensal dysbiosis, loss of intestinal barrier function and diminished resistance to colonization. In sum, FRCs act as an 'on-demand' immunological 'rheostat' by restraining activation of group 1 ILCs and thereby preventing immunopathological damage in the intestine.


Subject(s)
Citrobacter rodentium/immunology , Coronavirus Infections/immunology , Enterobacteriaceae Infections/immunology , Fibroblasts/immunology , Interleukin-15/metabolism , Lymphocytes/immunology , Murine hepatitis virus/immunology , Animals , Cells, Cultured , Immunity, Innate , Lymph Nodes/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Peyer's Patches/pathology , Th1 Cells/immunology , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/metabolism
9.
PLoS Biol ; 14(7): e1002515, 2016 07.
Article in English | MEDLINE | ID: mdl-27415420

ABSTRACT

Fibroblastic reticular cells (FRCs) form the cellular scaffold of lymph nodes (LNs) and establish distinct microenvironmental niches to provide key molecules that drive innate and adaptive immune responses and control immune regulatory processes. Here, we have used a graph theory-based systems biology approach to determine topological properties and robustness of the LN FRC network in mice. We found that the FRC network exhibits an imprinted small-world topology that is fully regenerated within 4 wk after complete FRC ablation. Moreover, in silico perturbation analysis and in vivo validation revealed that LNs can tolerate a loss of approximately 50% of their FRCs without substantial impairment of immune cell recruitment, intranodal T cell migration, and dendritic cell-mediated activation of antiviral CD8+ T cells. Overall, our study reveals the high topological robustness of the FRC network and the critical role of the network integrity for the activation of adaptive immune responses.


Subject(s)
Cell Communication/immunology , Cell Movement/immunology , Fibroblasts/immunology , Lymph Nodes/immunology , T-Lymphocytes/immunology , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Count , Cell Movement/genetics , Chemokine CCL19/genetics , Chemokine CCL19/immunology , Chemokine CCL19/metabolism , Dendritic Cells/cytology , Dendritic Cells/immunology , Fibroblasts/cytology , Fibroblasts/metabolism , Lymph Nodes/cytology , Lymph Nodes/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Models, Immunological , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
10.
Cell Rep ; 15(7): 1527-1541, 2016 05 17.
Article in English | MEDLINE | ID: mdl-27160906

ABSTRACT

Secondary lymphoid tissues provide specialized niches for the initiation of adaptive immune responses and undergo a remarkable expansion in response to inflammatory stimuli. Although the formation of B cell follicles was previously thought to be restricted to the postnatal period, we observed that the draining mesenteric lymph nodes (mLN) of helminth-infected mice form an extensive number of new, centrally located, B cell follicles in response to IL-4Rα-dependent inflammation. IL-4Rα signaling promoted LTα1ß2 (lymphotoxin) expression by B cells, which then interacted with CCL19 positive stromal cells to promote lymphoid enlargement and the formation of germinal center containing B cell follicles. Importantly, de novo follicle formation functioned to promote both total and parasite-specific antibody production. These data reveal a role for type 2 inflammation in promoting stromal cell remodeling and de novo follicle formation by promoting B cell-stromal cell crosstalk.


Subject(s)
Antibody Formation/immunology , B-Lymphocytes/metabolism , Helminthiasis/immunology , Helminthiasis/parasitology , Intestinal Diseases, Parasitic/immunology , Intestinal Diseases, Parasitic/parasitology , Lymphotoxin-alpha/metabolism , Nematospiroides dubius/physiology , Animals , Cell Proliferation , Chemokine CCL19/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Interleukin-4 Receptor alpha Subunit/metabolism , Lymph Nodes/parasitology , Lymph Nodes/pathology , Lymphotoxin beta Receptor/metabolism , Mice, Inbred C57BL , Signal Transduction , Stromal Cells/pathology , T-Lymphocytes/immunology
11.
Elife ; 5: e10561, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26830463

ABSTRACT

Lymph nodes (LNs) are highly confined environments with a cell-dense three-dimensional meshwork, in which lymphocyte migration is regulated by intracellular contractile proteins. However, the molecular cues directing intranodal cell migration remain poorly characterized. Here we demonstrate that lysophosphatidic acid (LPA) produced by LN fibroblastic reticular cells (FRCs) acts locally to LPA2 to induce T-cell motility. In vivo, either specific ablation of LPA-producing ectoenzyme autotaxin in FRCs or LPA2 deficiency in T cells markedly decreased intranodal T cell motility, and FRC-derived LPA critically affected the LPA2-dependent T-cell motility. In vitro, LPA activated the small GTPase RhoA in T cells and limited T-cell adhesion to the underlying substrate via LPA2. The LPA-LPA2 axis also enhanced T-cell migration through narrow pores in a three-dimensional environment, in a ROCK-myosin II-dependent manner. These results strongly suggest that FRC-derived LPA serves as a cell-extrinsic factor that optimizes T-cell movement through the densely packed LN reticular network.


Subject(s)
Cell Movement , Fibroblasts/metabolism , Lysophospholipids/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/physiology , Animals , Mice, Inbred C57BL , Molecular Sequence Data , Sequence Analysis, DNA , rhoA GTP-Binding Protein/metabolism
12.
Eur J Immunol ; 45(8): 2218-31, 2015 08.
Article in English | MEDLINE | ID: mdl-25973789

ABSTRACT

The thymic epithelium forms specialized niches to enable thymocyte differentiation. While the common epithelial progenitor of medullary and cortical thymic epithelial cells (mTECs and cTECs) is well defined, early stages of mTEC lineage specification have remained elusive. Here, we utilized in vivo targeting of mTECs to resolve their differentiation pathways and to determine whether mTEC progenitors participate in thymocyte education. We found that mTECs descend from a lineage committed, podoplanin (PDPN)-expressing progenitor located at the cortico-medullary junction. PDPN(+) junctional TECs (jTECs) represent a distinct TEC population that builds the thymic medulla, but only partially supports negative selection and thymocyte differentiation. Moreover, conditional gene targeting revealed that abrogation of alternative NF-κB pathway signaling in the jTEC stage completely blocked mTEC development. Taken together, this study identifies jTECs as lineage-committed mTEC progenitors and shows that NF-κB-dependent progression of jTECs to mTECs is critical to secure central tolerance.


Subject(s)
Cell Differentiation/immunology , Epithelial Cells/immunology , Membrane Glycoproteins/immunology , NF-kappa B/immunology , Signal Transduction/immunology , Stem Cells/immunology , Thymus Gland/immunology , Animals , Cell Differentiation/genetics , Epithelial Cells/cytology , Membrane Glycoproteins/genetics , Mice , Mice, Transgenic , NF-kappa B/genetics , Signal Transduction/genetics , Stem Cells/cytology , Thymus Gland/cytology
13.
J Immunol ; 192(11): 5192-200, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24778443

ABSTRACT

Abs play a significant role in protection against the intracellular bacterium Salmonella Typhi. In this article, we investigated how long-term protective IgM responses can be elicited by a S. Typhi outer-membrane protein C- and F-based subunit vaccine (porins). We found that repeated Ag exposure promoted a CD4(+) T cell-dependent germinal center reaction that generated mutated IgM-producing B cells and was accompanied by a strong expansion of IFN-γ-secreting T follicular helper cells. Genetic ablation of individual cytokine receptors revealed that both IFN-γ and IL-17 are required for optimal germinal center reactions and production of porin-specific memory IgM(+) B cells. However, more profound reduction of porin-specific IgM B cell responses in the absence of IFN-γR signaling indicated that this cytokine plays a dominant role. Importantly, mutated IgM mAbs against porins exhibited bactericidal capacity and efficiently augmented S. Typhi clearance. In conclusion, repeated vaccination with S. Typhi porins programs type I T follicular helper cell responses that contribute to the diversification of B cell memory and promote the generation of protective IgM Abs.


Subject(s)
B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Germinal Center/immunology , Immunoglobulin M/immunology , Immunologic Memory , Interferon-gamma/immunology , Salmonella typhi/immunology , Animals , CD4-Positive T-Lymphocytes/pathology , Female , Germinal Center/pathology , Humans , Interferon-gamma/genetics , Male , Mice , Mice, Knockout , Salmonella Vaccines/genetics , Salmonella Vaccines/immunology , Typhoid Fever/genetics , Typhoid Fever/immunology , Typhoid Fever/pathology , Typhoid Fever/prevention & control
14.
Eur J Immunol ; 43(12): 3233-43, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23963995

ABSTRACT

Attenuation of T cell-mediated damage of blood endothelial cells (BECs) in transplanted organs is important to prevent transplant vasculopathy (TV) and chronic rejection. Here, we assessed the importance of minor histocompatibility antigen (mHA) distribution and different coinhibitory molecules for T cell-BEC interaction. A transgenic mHA was directed specifically to BECs using the Tie2 promoter and cellular interactions were assessed in graft-versus-host disease-like and heterotopic heart transplantation settings. We found that cognate CD4(+) T-cell help was critical for the activation of BEC-specific CD8(+) T cells. However, systemic mHA expression on BECs efficiently attenuated adoptively transferred, BEC-specific CD4(+) and CD8(+) T cells and hence prevented tissue damage, whereas restriction of mHA expression to heart BECs precipitated the development of TV. Importantly, the lack of the coinhibitory molecules programmed death-1 (PD-1) and B and T lymphocyte attenuator fostered the initial activation of BEC-specific CD4(+) T cells, but did not affect development of TV. In contrast, TV was significantly augmented in the absence of PD-1 on BEC-specific CD8(+) T cells. Taken together, these results indicate that antigen distribution in the vascular bed determines the impact of coinhibition and, as a consequence, critically impinges on T cell-mediated vascular immunopathology.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Endothelial Cells/immunology , Graft Rejection/immunology , Heart Transplantation , Minor Histocompatibility Antigens/immunology , Vascular Diseases/immunology , Allografts , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Graft Rejection/genetics , Graft Rejection/metabolism , Graft Rejection/pathology , Mice , Mice, Knockout , Minor Histocompatibility Antigens/biosynthesis , Minor Histocompatibility Antigens/genetics , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism , Vascular Diseases/genetics , Vascular Diseases/metabolism , Vascular Diseases/pathology
15.
Blood ; 121(20): 4101-9, 2013 May 16.
Article in English | MEDLINE | ID: mdl-23558016

ABSTRACT

It is not known how naive B cells compute divergent chemoattractant signals of the T-cell area and B-cell follicles during in vivo migration. Here, we used two-photon microscopy of peripheral lymph nodes (PLNs) to analyze the prototype G-protein-coupled receptors (GPCRs) CXCR4, CXCR5, and CCR7 during B-cell migration, as well as the integrin LFA-1 for stromal guidance. CXCR4 and CCR7 did not influence parenchymal B-cell motility and distribution, despite their role during B-cell arrest in venules. In contrast, CXCR5 played a nonredundant role in B-cell motility in follicles and in the T-cell area. B-cell migration in the T-cell area followed a random guided walk model, arguing against directed migration in vivo. LFA-1, but not α4 integrins, contributed to B-cell motility in PLNs. However, stromal network guidance was LFA-1 independent, uncoupling integrin-dependent migration from stromal attachment. Finally, we observed that despite a 20-fold reduction of chemokine expression in virus-challenged PLNs, CXCR5 remained essential for B-cell screening of antigen-presenting cells. Our data provide an overview of the contribution of prototype GPCRs and integrins during naive B-cell migration and shed light on the local chemokine availability that these cells compute.


Subject(s)
B-Lymphocytes/physiology , Cell Communication/physiology , Chemokines/physiology , Chemotaxis, Leukocyte/genetics , Lymphocyte Function-Associated Antigen-1/metabolism , Receptors, CCR7/physiology , Receptors, CXCR4/physiology , Receptors, CXCR5/physiology , Stromal Cells/physiology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/physiology , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Cell Communication/drug effects , Chemokines/metabolism , Chemokines/pharmacology , Chemotaxis, Leukocyte/drug effects , Female , Gene Deletion , Lymphocyte Function-Associated Antigen-1/physiology , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, CCR7/genetics , Receptors, CCR7/metabolism , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Receptors, CXCR5/genetics , Receptors, CXCR5/metabolism , Stromal Cells/metabolism
16.
Immunity ; 38(5): 1013-24, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23623380

ABSTRACT

The stromal scaffold of the lymph node (LN) paracortex is built by fibroblastic reticular cells (FRCs). Conditional ablation of lymphotoxin-ß receptor (LTßR) expression in LN FRCs and their mesenchymal progenitors in developing LNs revealed that LTßR-signaling in these cells was not essential for the formation of LNs. Although T cell zone reticular cells had lost podoplanin expression, they still formed a functional conduit system and showed enhanced expression of myofibroblastic markers. However, essential immune functions of FRCs, including homeostatic chemokine and interleukin-7 expression, were impaired. These changes in T cell zone reticular cell function were associated with increased susceptibility to viral infection. Thus, myofibroblasic FRC precursors are able to generate the basic T cell zone infrastructure, whereas LTßR-dependent maturation of FRCs guarantees full immunocompetence and hence optimal LN function during infection.


Subject(s)
Coronavirus Infections/immunology , Lymph Nodes/cytology , Lymph Nodes/metabolism , Myofibroblasts/physiology , T-Lymphocytes/immunology , Animals , Cell Differentiation , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/immunology , Interleukin-7/biosynthesis , Lymph Nodes/immunology , Lymphotoxin beta Receptor/metabolism , Lymphotoxin-beta/biosynthesis , Lymphotoxin-beta/metabolism , Membrane Glycoproteins/biosynthesis , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Murine hepatitis virus/immunology , Myofibroblasts/cytology , Signal Transduction
17.
J Exp Med ; 210(3): 465-73, 2013 Mar 11.
Article in English | MEDLINE | ID: mdl-23420877

ABSTRACT

The development of lymph nodes (LNs) and formation of LN stromal cell microenvironments is dependent on lymphotoxin-ß receptor (LTßR) signaling. In particular, the LTßR-dependent crosstalk between mesenchymal lymphoid tissue organizer and hematopoietic lymphoid tissue inducer cells has been regarded as critical for these processes. Here, we assessed whether endothelial cell (EC)-restricted LTßR signaling impacts on LN development and the vascular LN microenvironment. Using EC-specific ablation of LTßR in mice, we found that conditionally LTßR-deficient animals failed to develop a significant proportion of their peripheral LNs. However, remnant LNs showed impaired formation of high endothelial venules (HEVs). Venules had lost their cuboidal shape, showed reduced segment length and branching points, and reduced adhesion molecule and constitutive chemokine expression. Due to the altered EC-lymphocyte interaction, homing of lymphocytes to peripheral LNs was significantly impaired. Thus, this study identifies ECs as an important LTßR-dependent lymphoid tissue organizer cell population and indicates that continuous triggering of the LTßR on LN ECs is critical for lymphocyte homeostasis.


Subject(s)
Endothelial Cells/physiology , Lymph Nodes/physiology , Lymphotoxin beta Receptor/physiology , Signal Transduction/physiology , Venules/physiology , Animals , Cadherins/physiology , Homeostasis , Lymph Nodes/cytology , Mice , Mice, Inbred C57BL , Transgenes
18.
Blood ; 120(24): 4675-83, 2012 Dec 06.
Article in English | MEDLINE | ID: mdl-22955921

ABSTRACT

Nonhematopoietic stromal cells of secondary lymphoid organs form important scaffold and fluid transport structures, such as lymph node (LN) trabeculae, lymph vessels, and conduits. Furthermore, through the production of chemokines and cytokines, these cells generate a particular microenvironment that determines lymphocyte positioning and supports lymphocyte homeostasis. IL-7 is an important stromal cell-derived cytokine that has been considered to be derived mainly from T-cell zone fibroblastic reticular cells. We show here that lymphatic endothelial cells (LECs) are a prominent source of IL-7 both in human and murine LNs. Using bacterial artificial chromosome transgenic IL-7-Cre mice, we found that fibroblastic reticular cells and LECs strongly up-regulated IL-7 expression during LN remodeling after viral infection and LN reconstruction after avascular transplantation. Furthermore, IL-7-producing stromal cells contributed to de novo formation of LyveI-positive lymphatic structures connecting reconstructed LNs with the surrounding tissue. Importantly, diphtheria toxin-mediated depletion of IL-7-producing stromal cells completely abolished LN reconstruction. Taken together, this study identifies LN LECs as a major source of IL-7 and shows that IL-7-producing stromal cells are critical for reconstruction and remodeling of the distinct LN microenvironment.


Subject(s)
Endothelial Cells/metabolism , Interleukin-7/metabolism , Lymph Nodes/metabolism , Stromal Cells/metabolism , Adult , Animals , Cell Line , Cell Proliferation , Cells, Cultured , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression , Humans , Immunohistochemistry , Interleukin-7/genetics , Kidney/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Lymph Nodes/embryology , Lymph Nodes/transplantation , Lymphatic Vessels/cytology , Lymphatic Vessels/metabolism , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Reverse Transcriptase Polymerase Chain Reaction
19.
J Immunol ; 189(2): 841-9, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22696443

ABSTRACT

Abs confer protection from secondary infection with Legionella pneumophila, the causative agent of a severe form of pneumonia known as Legionnaires' disease. In this study, we demonstrate that Ab-mediated protection is effective across L. pneumophila serogroups, suggesting that Abs specific for conserved protein Ags are sufficient to mediate this protective effect. We used two independent methods to identify immunogenic L. pneumophila protein Ags, namely, the screening of a λ phage library representing the complete L. pneumophila genome and two-dimensional gel electrophoresis combined with Western blot analysis and protein spot identification by mass spectrometry. A total of 30 novel L. pneumophila B cell Ags were identified, the majority of which are located in or associated with the bacterial membrane, where they are accessible for Abs and, therefore, likely to be relevant for Ab-mediated protection against L. pneumophila. Selected B cell Ags were recombinantly expressed and tested in a vaccination protocol. Mice immunized with either single-protein Ags or an Ag combination showed reduced bacterial titers in bronchoalveolar lavage and lung after L. pneumophila challenge. To determine the clinical relevance of these findings, we tested Legionnaires' disease patient sera for reactivity with the identified L. pneumophila Ags. The recognized Ags were indeed conserved across host species, because Abs specific for all three selected Ags could be detected in patient sera, rendering the identified protein Ags potential vaccine candidates.


Subject(s)
Antigens, Bacterial/isolation & purification , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/microbiology , Legionella pneumophila/immunology , Legionnaires' Disease/immunology , Administration, Intranasal , Animals , Antibodies, Bacterial/biosynthesis , Antibodies, Bacterial/blood , Antibodies, Bacterial/therapeutic use , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/immunology , B-Lymphocyte Subsets/metabolism , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/chemical synthesis , Bacterial Vaccines/immunology , Bacteriophage lambda/genetics , Bacteriophage lambda/immunology , Conserved Sequence/immunology , Humans , Immunoglobulin G/biosynthesis , Immunoglobulin G/blood , Legionella pneumophila/pathogenicity , Legionnaires' Disease/blood , Legionnaires' Disease/prevention & control , Mice , Mice, Inbred A , Mice, Inbred C57BL , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Vaccines, Synthetic/therapeutic use
20.
Eur J Immunol ; 42(9): 2311-21, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22730043

ABSTRACT

Myocarditis is a potentially lethal inflammatory heart disease of children and young adults that frequently leads to dilated cardiomyopathy (DCM). Since diagnostic procedures and efficient therapies are lacking, it is important to characterize the critical immune effector pathways underlying the initial cardiac inflammation and the transition from myocarditis to DCM. We describe here a T-cell receptor (TCR) transgenic mouse model with spontaneously developing autoimmune myocarditis that progresses to lethal DCM. Cardiac magnetic resonance imaging revealed early inflammation-associated changes in the ventricle wall including transient thickening of the left ventricle wall. Furthermore, we found that IFN-γ was a major effector cytokine driving the initial inflammatory process and that the cooperation of IFN-γ and IL-17A was essential for the development of the progressive disease. This novel TCR transgenic mouse model permits the identification of the central pathophysiological and immunological processes involved in the transition from autoimmune myocarditis to DCM.


Subject(s)
Autoimmune Diseases/immunology , Cardiomyopathy, Dilated/immunology , Cardiomyopathy, Dilated/pathology , Myocarditis/immunology , Myocarditis/pathology , Th1 Cells/immunology , Th17 Cells/immunology , Animals , Autoantigens/immunology , Autoimmune Diseases/pathology , Disease Models, Animal , Heart Ventricles/immunology , Heart Ventricles/pathology , Inflammation/immunology , Inflammation/pathology , Interferon-gamma/immunology , Interleukin-17/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , Receptors, Antigen, T-Cell/immunology , Ventricular Remodeling/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...