Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Article in English | MEDLINE | ID: mdl-33903153

ABSTRACT

The molecular basis of the persistence of experienced T lymphocytes, also known as "memory T lymphocytes," is still enigmatic. We are beginning to understand their considerable heterogeneity and topographic compartmentalization into memory T cells circulating through the body and those residing in a particular tissue. In some tissues, like murine spleen, subpopulations of memory T cells proliferating in the absence of antigen (homeostatic proliferation) have been described. Other populations are maintained resting in terms of transcription, mobility, and proliferation in dedicated survival niches organized by stromal cells. The survival of these memory T cells is conditional on being in such a niche, where they can persist for a lifetime. Circulating memory T lymphocytes of distinct immune responses slowly decline in numbers over time. The rules governing their entry into and exit from blood, as well as their lifestyle outside of the blood and their relation to resident memory T cells are poorly understood. Homeostasis of circulating, proliferating, and resting memory T cells is obviously controlled by different rheostats: tissue-exit and tissue-entry signals for circulating and proliferation-inducing signals for proliferating memory T cells. For tissue-resident, resting memory T cells, it is the availability of their survival niche. Apparently, this mechanism (i.e., the link between memory T cell and stromal cell) is so robust that it provides efficient T-cell memory over a lifetime in tissues such as the bone marrow.


Subject(s)
Immunologic Memory , Memory T Cells/physiology , Animals , Cell Survival , Homeostasis , Humans
2.
Nat Commun ; 11(1): 2570, 2020 05 22.
Article in English | MEDLINE | ID: mdl-32444631

ABSTRACT

At present, it is not clear how memory B lymphocytes are maintained over time, and whether only as circulating cells or also residing in particular tissues. Here we describe distinct populations of isotype-switched memory B lymphocytes (Bsm) of murine spleen and bone marrow, identified according to individual transcriptional signature and B cell receptor repertoire. A population of marginal zone-like cells is located exclusively in the spleen, while a population of quiescent Bsm is found only in the bone marrow. Three further resident populations, present in spleen and bone marrow, represent transitional and follicular B cells and B1 cells, respectively. A population representing 10-20% of spleen and bone marrow memory B cells is the only one qualifying as circulating. In the bone marrow, all cells individually dock onto VCAM1+ stromal cells and, reminiscent of resident memory T and plasma cells, are void of activation, proliferation and mobility.


Subject(s)
B-Lymphocytes/immunology , Bone Marrow Cells/immunology , Immunoglobulin Class Switching , Immunologic Memory , Spleen/immunology , Adjuvants, Immunologic/pharmacology , Animals , Animals, Wild/immunology , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , Bone Marrow Cells/cytology , Cell Cycle , Cell Proliferation/genetics , Gene Expression Regulation/immunology , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/immunology , Spleen/cytology , Stromal Cells/cytology , Vascular Cell Adhesion Molecule-1/metabolism
3.
EBioMedicine ; 53: 102684, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32114393

ABSTRACT

BACKGROUND: Human immunology research is often limited to peripheral blood. However, there are important differences between blood immune cells and their counterparts residing in secondary lymphoid organs, such as in the case of germinal center (GC) T follicular helper (Tfh) cells and GC B cells. METHODS: We developed a versatile ex vivo lymphoid organ culture platform that is based on human pharyngeal tonsils (adenoids) and allows for drug testing. We systematically phenotyped Tfh and GC B cell subsets in explant- and suspension cultures using multicolor flow cytometry and cytokine multiplex analysis. FINDINGS: Phenotypic changes of certain ex vivo cultured immune cell subsets could be modulated by cytokine addition. Furthermore, we optimized an activation-induced marker assay to evaluate the response to T cell stimulation. We provide proof-of-concept that Tfh and GC B cells could be modulated in these cultures by different anti-inflammatory drugs in unstimulated states and upon activation with vaccine-derived antigens. For example, GC B cells were lost upon CD40L blockade, and clinically approved JAK inhibitors impacted Tfh and GC B cells, including down-regulation of their key transcription factor BCL6. BCL6 regulation was affected by IL-6 signaling in T cells and IL-4 in B cells, respectively. Furthermore, we demonstrated that JAK signaling and TNF signaling contributed to the stimulation-induced activation of tonsil-derived T cells. INTERPRETATION: Our optimized methods, assays, and mechanistic findings can contribute to a better understanding of human GC responses. These insights may be relevant for improving autoimmune disease therapy and vaccination efficacy. FUNDING: This work was supported by a project grant under the joint research cooperation agreement of LMU Munich, LMU University Hospital, and Sanofi-Aventis Deutschland GmbH, as well as by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation) - Emmy Noether Programme BA 5132/1-1 and BA 5132/1-2 (252623821), SFB 1054 Project B12 (210592381), and SFB 914 Project B03 (165054336).


Subject(s)
Adenoids/immunology , Anti-Inflammatory Agents/pharmacology , B-Lymphocytes/immunology , Germinal Center/immunology , T Follicular Helper Cells/immunology , Adenoids/cytology , B-Lymphocytes/drug effects , Cells, Cultured , Child , Child, Preschool , Germinal Center/cytology , Humans , Immunophenotyping/methods , Interleukins/genetics , Interleukins/metabolism , Janus Kinases/metabolism , Proto-Oncogene Proteins c-bcl-6/metabolism , T Follicular Helper Cells/drug effects , Tissue Culture Techniques/methods , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
4.
Eur J Immunol ; 49(9): 1372-1379, 2019 09.
Article in English | MEDLINE | ID: mdl-31149730

ABSTRACT

Bone marrow (BM) stromal cells are important in the development and maintenance of cells of the immune system. Using single cell RNA sequencing, we here explore the functional and phenotypic heterogeneity of individual transcriptomes of 1167 murine BM mesenchymal stromal cells. These cells exhibit a tremendous heterogeneity of gene expression, which precludes the identification of defined subpopulations. However, according to the expression of 108 genes involved in the communication of stromal cells with hematopoietic cells, we have identified 14 non-overlapping subpopulations, with distinct cytokine or chemokine gene expression signatures. With respect to the maintenance of subsets of immune memory cells by stromal cells, we identified distinct subpopulations expressing Il7, Il15 and Tnfsf13b. Together, this study provides a comprehensive dissection of the BM stromal heterogeneity at the single cell transcriptome level and provides a basis to understand their lifestyle and their role as organizers of niches for the long-term maintenance of immune cells.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow/physiology , Stromal Cells/cytology , Transcriptome/genetics , Animals , B-Cell Activating Factor/genetics , Cells, Cultured , Cytokines/genetics , Hematopoietic Stem Cells/cytology , Interleukin-15/genetics , Interleukin-7/genetics , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred C57BL , Sequence Analysis, RNA/methods
5.
Eur J Immunol ; 49(6): 966-968, 2019 06.
Article in English | MEDLINE | ID: mdl-30673129

ABSTRACT

It is a matter of current debate whether the bone marrow is a hub for circulating memory T lymphocytes and/or the home of resident memory T lymphocytes. Here we demonstrate for CD69+ murine CD8+ , and CD69+ murine and human CD4+ memory T lymphocytes of the bone marrow, making up between 30 and 60% of bone marrow memory T lymphocytes, that they express the gene expression signature of tissue-resident memory T lymphocytes. This suggests that a substantial proportion of bone marrow memory T lymphocytes are resident. It adds to previous evidence that bone marrow memory T cells are resting in terms of mobility and proliferation, and maintain exclusive long-term memory to distinct, systemic antigens.


Subject(s)
Bone Marrow Cells/immunology , Immunologic Memory/immunology , Spleen/immunology , T-Lymphocyte Subsets/immunology , Animals , Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , Humans , Lectins, C-Type/immunology , Mice , Spleen/cytology , Transcriptome/immunology
7.
Eur J Immunol ; 46(11): 2566-2573, 2016 11.
Article in English | MEDLINE | ID: mdl-27562840

ABSTRACT

CD40-CD40 ligand (CD40L) signaling plays multiple indispensable roles in cellular and humoral immunity. Impaired memory T-cell responses in the absence of CD40L have been well documented, but the requirement of this interaction for efficient priming of CD8+ T cells especially under inflammatory conditions has been under debate. In contrast to previous publications, we report here that virus-specific CD8+ T-cell responses as well as viral clearance are affected not only in the memory but also in the effector phase in CD40L-/- mice infected with lymphocytic choriomeningitis virus (LCMV) Armstrong strain. Interestingly, a considerable part of the LCMV-specific effector and memory T cells consists of CD40L+ CD8+ T cells. However, deficiency of CD40L in CD8+ T cells did influence neither the quantity nor the quality of primary T-cell responses in LCMV infection. Virus-specific CD8+ T cells in conditional knockout mice, with a selective deletion of the CD40L in CD8+ T cells, were fully functional regarding cytokine production and efficient pathogen clearance. Thus, our results unambiguously demonstrate that while CD40L is critical to generate effective primary CD8+ T-cell responses also under inflammatory conditions, CD40L expression by CD8+ T cells themselves is dispensable in acute LCMV infection.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD40 Ligand/genetics , CD40 Ligand/immunology , CD8-Positive T-Lymphocytes/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Acute Disease , Animals , CD40 Antigens/genetics , CD40 Antigens/immunology , CD40 Ligand/deficiency , CD40 Ligand/metabolism , Cytokines/immunology , Gene Expression Regulation , Immunologic Memory , Lymphocyte Activation , Lymphocytic Choriomeningitis/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes, Cytotoxic/immunology
8.
PLoS One ; 10(8): e0135081, 2015.
Article in English | MEDLINE | ID: mdl-26252021

ABSTRACT

METHODS: NZB/W F1 mice were treated with: 1) anti-CD20, 2) anti-CD20 plus bortezomib, 3) anti-CD20 plus anti-LFA-1/anti-VLA-4 blocking antibodies, 4) anti-CD20 plus bortezomib and anti-LFA-1/anti-VLA4 blocking antibodies. Short- and long-lived plasma cells including autoreactive cells in the bone marrow and spleen were enumerated by flow cytometry and ELISPOT seven days after treatment. Based on these data in another experiment, mice received one cycle of anti-CD20 plus bortezomib followed by four cycles of anti-CD20 therapy every 10 days and were monitored for its effect on plasma cells and disease. RESULTS: Short-lived plasma cells in bone marrow and spleen were efficiently depleted by all regimens targeting plasma cells. Conversely, LLPCs and anti-dsDNA-secreting plasma cells in bone marrow and spleen showed resistance to depletion and were strongly reduced by bortezomib plus anti-CD20. The effective depletion of plasma cells by bortezomib complemented by the continuous depletion of their precursor B cells using anti-CD20 promoted the persistent reduction of IgG anti-dsDNA antibodies, delayed nephritis and prolonged survival in NZB/W F1 mice. CONCLUSIONS: These findings suggest that the effective depletion of LLPCs using bortezomib in combination with a therapy that continuously targeting B cells as their precursors may prevent the regeneration of autoreactive LLPCs and, thus, might represent a promising treatment strategy for SLE and other (auto)antibody-mediated diseases.


Subject(s)
B-Lymphocytes/cytology , Bortezomib/therapeutic use , Lupus Nephritis/blood , Lupus Nephritis/drug therapy , Spleen/immunology , Animals , Antigens, CD20/metabolism , Antineoplastic Agents/therapeutic use , Autoantibodies/immunology , Bone Marrow/pathology , Cell Separation , Disease Models, Animal , Female , Flow Cytometry , Integrin alpha4beta1/metabolism , Lupus Nephritis/immunology , Lymphocyte Function-Associated Antigen-1/metabolism , Mice , Mice, Inbred NZB , Plasma Cells/cytology , Regeneration , Spleen/drug effects , Spleen/pathology
9.
Eur J Immunol ; 45(4): 975-87, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25639669

ABSTRACT

It is believed that memory CD8(+) T cells are maintained in secondary lymphoid tissues, peripheral tissues, and BM by homeostatic proliferation. Their survival has been shown to be dependent on IL-7, but it is unclear where they acquire it. Here we show that in murine BM, memory CD8(+) T cells individually colocalize with IL-7(+) reticular stromal cells. The T cells are resting in terms of global transcription and do not express markers of activation, for example, 4-1BB (CD137), IL-2, or IFN-γ, despite the expression of CD69 on about 30% of the cells. Ninety-five percent of the memory CD8(+) T cells in BM are in G0 phase of cell cycle and do not express Ki-67. Less than 1% is in S/M/G2 of cell cycle, according to propidium iodide staining. While previous publications have estimated the extent of proliferation of CD8(+) memory T cells on the basis of BrdU incorporation, we show here that BrdU itself induces proliferation of CD8(+) memory T cells. Taken together, the present results suggest that CD8(+) memory T cells are maintained as resting cells in the BM in dedicated niches with their survival conditional on IL-7 receptor signaling.


Subject(s)
Bone Marrow Cells/cytology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Resting Phase, Cell Cycle/immunology , Stromal Cells/immunology , Animals , Antigens, CD/biosynthesis , Antigens, Differentiation, T-Lymphocyte/biosynthesis , Bone Marrow Cells/immunology , Cell Proliferation , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Interleukin-7/immunology , Ki-67 Antigen/biosynthesis , Lectins, C-Type/biosynthesis , Mice , Mice, Inbred C57BL , Transcription, Genetic , Tumor Necrosis Factor Receptor Superfamily, Member 9/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...