Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
2.
Commun Biol ; 5(1): 1037, 2022 09 29.
Article in English | MEDLINE | ID: mdl-36175547

ABSTRACT

Few human tumours present with a recurrent pathognomonic mutation in a transcription factor. Thymomas are an exception, with the majority of some subtypes exhibiting a distinct somatically acquired missense mutation in the general transcription factor GTF2I. Co-dominant expression of wild-type and mutated forms of Gtf2i in the mouse thymic epithelium is associated with aberrant thymic architecture and reduced thymopoietic activity. Phenotypic and molecular characterization of the mutant epithelium indicates that medullary differentiation is particularly affected as a result of impaired differentiation of bi-potent epithelial progenitors. The resulting gene expression signature is dominated by that of immature cortex-like thymic epithelial cells. TCR repertoire analysis of the cytopenic T cell compartment indicates efficient intrathymic selection; hence, despite marked homeostatic proliferation of T cell clones, autoimmunity is not observed. Thus, our transgenic mouse model recapitulates some aspects of the pathophysiology of a genetically defined type of human thymoma.


Subject(s)
Thymoma , Thymus Neoplasms , Transcription Factors, General , Transcription Factors, TFIII , Transcription Factors, TFII , Animals , Humans , Mice , Mice, Transgenic , Mutation , Receptors, Antigen, T-Cell , Thymoma/genetics , Thymus Neoplasms/genetics , Transcription Factors , Transcription Factors, TFII/genetics
3.
Nature ; 606(7912): 165-171, 2022 06.
Article in English | MEDLINE | ID: mdl-35614226

ABSTRACT

T cell development in the thymus is essential for cellular immunity and depends on the organotypic thymic epithelial microenvironment. In comparison with other organs, the size and cellular composition of the thymus are unusually dynamic, as exemplified by rapid growth and high T cell output during early stages of development, followed by a gradual loss of functional thymic epithelial cells and diminished naive T cell production with age1-10. Single-cell RNA sequencing (scRNA-seq) has uncovered an unexpected heterogeneity of cell types in the thymic epithelium of young and aged adult mice11-18; however, the identities and developmental dynamics of putative pre- and postnatal epithelial progenitors have remained unresolved1,12,16,17,19-27. Here we combine scRNA-seq and a new CRISPR-Cas9-based cellular barcoding system in mice to determine qualitative and quantitative changes in the thymic epithelium over time. This dual approach enabled us to identify two principal progenitor populations: an early bipotent progenitor type biased towards cortical epithelium and a postnatal bipotent progenitor population biased towards medullary epithelium. We further demonstrate that continuous autocrine provision of Fgf7 leads to sustained expansion of thymic microenvironments without exhausting the epithelial progenitor pools, suggesting a strategy to modulate the extent of thymopoietic activity.


Subject(s)
Epithelial Cells , Stem Cells , T-Lymphocytes , Thymus Gland , Aging , Animals , Autocrine Communication , CRISPR-Cas Systems , Cellular Microenvironment , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelium , Fibroblast Growth Factor 7 , Mice , RNA-Seq , Single-Cell Analysis , Stem Cells/cytology , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Thymus Gland/cytology
4.
Open Biol ; 11(2): 200383, 2021 02.
Article in English | MEDLINE | ID: mdl-33622100

ABSTRACT

In vertebrates, the development of lymphocytes from undifferentiated haematopoietic precursors takes place in so-called primary lymphoid organs, such as the thymus. Therein, lymphocytes undergo a complex differentiation and selection process that culminates in the generation of a pool of mature T cells that collectively express a self-tolerant repertoire of somatically diversified antigen receptors. Throughout this entire process, the microenvironment of the thymus in large parts dictates the sequence and outcome of the lymphopoietic activity. In vertebrates, direct genetic evidence in some species and circumstantial evidence in others suggest that the formation of a functional thymic microenvironment is controlled by members of the Foxn1/4 family of transcription factors. In teleost fishes, both Foxn1 and Foxn4 contribute to thymopoietic activity, whereas Foxn1 is both necessary and sufficient in the mammalian thymus. The evolutionary history of Foxn1/4 genes suggests that an ancient Foxn4 gene lineage gave rise to the Foxn1 genes in early vertebrates, raising the question of the thymopoietic capacity of the ancestor common to all vertebrates. Recent attempts to reconstruct the early events in the evolution of thymopoietic tissues by replacement of the mouse Foxn1 gene by Foxn1-like genes isolated from various chordate species suggest a plausible scenario. It appears that the primordial thymus was a bi-potent lymphoid organ, supporting both B cell and T cell development; however, during the course of vertebrate, evolution B cell development was gradually diminished converting the thymus into a site specialized in T cell development.


Subject(s)
Lymphopoiesis , Stem Cell Niche , Thymus Gland/cytology , Animals , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Humans , Receptors, Antigen/genetics , Receptors, Antigen/metabolism , Thymus Gland/immunology
5.
Sci Adv ; 6(48)2020 11.
Article in English | MEDLINE | ID: mdl-33246964

ABSTRACT

The onset of lymphocyte development in the vertebrate primordial thymus, about 500 million years ago, represents one of the foundational events of the emerging adaptive immune system. Here, we retrace the evolutionary trajectory of thymopoiesis, from early vertebrates to mammals, guided by members of the Foxn1/4 transcription factor gene family, which direct the differentiation of the thymic microenvironment. Molecular engineering in transgenic mice recapitulated a gene duplication event, exon replacements, and altered expression patterns. These changes predictably modified the lymphopoietic characteristics of the thymus, identifying molecular features contributing to conversion of a primordial bipotent lymphoid organ to a tissue specializing in T cell development. The phylogenetic reconstruction associates increasing efficiency of T cell generation with diminishing B cell-generating capacity of the thymus during jawed vertebrate evolution.


Subject(s)
Lymphopoiesis , T-Lymphocytes , Animals , B-Lymphocytes , Lymphopoiesis/genetics , Mammals , Mice , Mice, Transgenic , Phylogeny , Vertebrates
6.
Sci Rep ; 8(1): 11095, 2018 07 23.
Article in English | MEDLINE | ID: mdl-30038304

ABSTRACT

The numbers of thymic epithelial cells (TECs) and thymocytes steadily increase during embryogenesis. To examine this dynamic, we generated several TEC-specific transgenic mouse lines, which express fluorescent proteins in the nucleus, the cytosol and in the membranes under the control of the Foxn1 promoter. These tools enabled us to determine TEC numbers in tissue sections by confocal fluorescent microscopy, and in the intact organ by light-sheet microscopy. Compared to histological procedures, flow cytometric analysis of thymic cellularity is shown to underestimate the numbers of TECs by one order of magnitude; using enzymatic digestion of thymic tissue, the loss of cortical TECs (cTECs) is several fold greater than that of medullary TECs (mTECs), although different cTEC subsets appear to be still present in the final preparation. Novel reporter lines driven by Psmb11 and Prss16 promoters revealed the trajectory of differentiation of cTEC-like cells, and, owing to the additional facility of conditional cell ablation, allowed us to follow the recovery of such cells after their depletion during embryogenesis. Multiparametric histological analyses indicate that the new transgenic reporter lines not only reveal the unique morphologies of different TEC subsets, but are also conducive to the analysis of the complex cellular interactions in the thymus.


Subject(s)
Epithelium/embryology , Thymus Gland/embryology , Animals , Cell Communication , Cellular Microenvironment , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelium/metabolism , Gene Expression , Genes, Reporter , Mice, Transgenic , Stromal Cells/cytology , Stromal Cells/metabolism , Thymus Gland/metabolism
7.
Eur J Immunol ; 45(3): 932-42, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25408420

ABSTRACT

T-cell lymphopenia following BM transplantation or diseases such as AIDS result in immunodeficiency. Novel approaches to ameliorate this situation are urgently required. Herein, we describe a novel stromal cell free culture system in which Lineage(-) Sca1(+)c-kit(+) BM hematopoietic progenitors very efficiently differentiate into pro-T cells. This culture system consists of plate-bound Delta-like 4 Notch ligand and the cytokines SCF and IL-7. The pro-T cells developing in these cultures express CD25, CD117, and partially CD44; express cytoplasmic CD3ε; and have their TCRß locus partially D-J rearranged. They could be expanded for over 3 months and used to reconstitute the T-cell compartments of sublethally irradiated T-cell-deficient CD3ε(-/-) mice or lethally irradiated WT mice. Pro-T cells generated in this system could partially correct the T-cell lymphopenia of pre-Tα(-/-) mice. However, reconstituted CD3ε(-/-) mice suffered from a wasting disease that was prevented by co-injection of purified CD4(+) CD25(high) WT Treg cells. In a T-cell-sufficient or T-lymphopenic setting, the development of disease was not observed. Thus, this in vitro culture system represents a powerful tool to generate large numbers of pro-T cells for transplantation and possibly with clinical applications.


Subject(s)
Cell Culture Techniques/methods , Gene Rearrangement, alpha-Chain T-Cell Antigen Receptor/immunology , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor/immunology , Precursor Cells, T-Lymphoid/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocytes, Regulatory/immunology , Adaptor Proteins, Signal Transducing , Animals , Antigens, CD/genetics , Antigens, CD/immunology , CD3 Complex/genetics , CD3 Complex/immunology , Calcium-Binding Proteins , Cells, Cultured , Female , Gene Rearrangement, alpha-Chain T-Cell Antigen Receptor/genetics , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor/genetics , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Precursor Cells, T-Lymphoid/cytology , Receptors, Antigen, T-Cell, alpha-beta/genetics , Stromal Cells , T-Lymphocytes, Regulatory/cytology
8.
Eur J Immunol ; 44(10): 2893-902, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25044476

ABSTRACT

Some signs of potential autoimmunity, such as the appearance of antinuclear antibodies (ANAs) become prevalent with age. In most cases, elderly people with ANAs remain healthy. Here, we investigated whether the same holds true for inbred strains of mice. Indeed, we show that most mice of the C57BL/6 (B6) strain spontaneously produced IgG ANA at 8-12 months of age, showed IgM deposition in kidneys and lymphocyte infiltrates in submandibular salivary glands. Despite all of this, the mice remained healthy. ANA production is likely CD4(+) T-cell dependent, since old (40-50 weeks of age) B6 mice deficient for MHC class II do not produce IgG ANAs. BM chimeras showed that ANA production was not determined by age-related changes in radiosensitive, hematopoietic progenitor cells, and that the CD4(+) T cells that promote ANA production were radioresistant. Thymectomy of B6 mice at 5 weeks of age led to premature alterations in T-cell homeostasis and ANA production, by 15 weeks of age, similar to that in old mice. Our findings suggest that a disturbed T-cell homeostasis may drive the onset of some autoimmune features.


Subject(s)
Aging/immunology , Autoimmunity/immunology , CD4-Positive T-Lymphocytes/immunology , Homeostasis/immunology , Animals , Antibodies, Antinuclear/blood , Antibodies, Antinuclear/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorescent Antibody Technique, Indirect , Immunohistochemistry , Mice , Mice, Inbred C57BL
9.
Bioconjug Chem ; 25(5): 879-87, 2014 May 21.
Article in English | MEDLINE | ID: mdl-24766622

ABSTRACT

The TNF-related apoptosis-inducing ligand (TRAIL) is a powerful inducer of apoptosis in tumor cells; however, clinical studies with recombinant soluble TRAIL were rather disappointing. Here, we developed TRAIL-functionalized liposomes (LipoTRAIL, LT) to mimic membrane-displayed TRAIL for efficient activation of death receptors DR4 and DR5 and enhanced induction of apoptosis, which were combined with an anti-EGFR single-chain Fv fragment (scFv) for targeted delivery to EGFR-positive tumor cells. These immuno-LipoTRAILs (ILTs) bound specifically to EGFR-expressing cells (Colo205) and exhibited increased cytotoxicity compared with that of nontargeted LTs. Compared to that of the soluble TRAIL, the plasma half-life of the functionalized liposomes was strongly extended, and increased antitumor activity of LT and ILT was demonstrated in a xenograft tumor model. Thus, we established a multifunctional liposomal TRAIL formulation (ILT) with improved pharmacokinetic and pharmacodynamic behavior, characterized by targeted delivery and increased induction of apoptosis due to multivalent TRAIL presentation.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Delivery Systems , Liposomes/immunology , Nanoparticles/chemistry , Neoplasms, Experimental/drug therapy , TNF-Related Apoptosis-Inducing Ligand/immunology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/immunology , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Liposomes/chemistry , Mice , Mice, Inbred Strains , Mice, Nude , Models, Molecular , Neoplasms, Experimental/pathology , Particle Size , Structure-Activity Relationship , Surface Properties , TNF-Related Apoptosis-Inducing Ligand/chemistry
10.
Eur J Immunol ; 44(5): 1299-312, 2014 May.
Article in English | MEDLINE | ID: mdl-24497074

ABSTRACT

Autoimmune diseases develop when self-specific T cells that escaped negative selection initiate a harmful immune response against self. However, factors, which influence the initiation and progression of an autoimmune response remain incompletely understood. By establishing a double-transgenic BALB/c mouse system in which different amounts of a cell-surface neo-self-antigen are expressed under the CD11c promoter, we demonstrate that antigen dose dramatically influences T-cell tolerance mechanisms. Moderate antigen expression in both hematopoietic and nonhematopoietic cells favors the development of antigen-specific Treg cells and the establishment of a tolerogenic environment. In marked contrast, a high dose of antigen expression results in very stringent negative selection, in poor development of antigen-specific Treg cells and in the early onset of anemia and splenomegaly and the late development of arthritis and high titers of IgG auto Abs. Disease is initiated by autoreactive T cells, which escape negative selection by expressing a second TCR with a different specificity or an altered affinity. Transfer of Ag-specific Treg cells ameliorates the early onset signs of disease but does not prevent the development of long-term chronic pathologies. Altogether, our results suggest that Ag dose directly affects Treg-cell generation and thus, the set-up of T-cell tolerance.


Subject(s)
Autoantigens/immunology , Autoimmune Diseases/immunology , Immunoglobulin G/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Autoantibodies/blood , Autoantibodies/immunology , Autoantigens/blood , Autoantigens/genetics , Autoimmune Diseases/blood , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , CD11c Antigen/blood , CD11c Antigen/genetics , CD11c Antigen/immunology , Dose-Response Relationship, Immunologic , Immune Tolerance , Immunoglobulin G/blood , Immunoglobulin G/genetics , Mice , Mice, Inbred BALB C , Mice, Transgenic , Receptors, Antigen, T-Cell , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology
11.
Haematologica ; 99(4): 638-46, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24463214

ABSTRACT

Cytokines are essential regulators of hematopoiesis, acting in an instructive or permissive way. Fms-like tyrosine kinase 3 ligand (FLT3L) is an important cytokine for the development of several hematopoietic populations. Its receptor (FLT3) is expressed on both myeloid and lymphoid progenitors and deletion of either the receptor or its ligand leads to defective developmental potential of hematopoietic progenitors. In vivo administration of FLT3L promotes expansion of progenitors with combined myeloid and lymphoid potential. To investigate further the role of this cytokine in hematopoietic development, we generated transgenic mice expressing high levels of human FLT3L. These transgenic mice displayed a dramatic expansion of dendritic and myeloid cells, leading to splenomegaly and blood leukocytosis. Bone marrow myeloid and lymphoid progenitors were significantly increased in numbers but retained their developmental potential. Furthermore, the transgenic mice developed anemia together with a reduction in platelet numbers. FLT3L was shown to rapidly reduce the earliest erythroid progenitors when injected into wild-type mice, indicating a direct negative role of the cytokine on erythropoiesis. We conclude that FLT3L acts on multipotent progenitors in an instructive way, inducing their development into myeloid/lymphoid lineages while suppressing their megakaryocyte/erythrocyte potential.


Subject(s)
Hematopoiesis/physiology , Membrane Proteins/genetics , Anemia/genetics , Anemia/metabolism , Animals , Bone Marrow/immunology , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Differentiation , Dendritic Cells/immunology , Dendritic Cells/metabolism , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Gene Expression , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Leukocytosis/genetics , Leukocytosis/metabolism , Lymphatic Diseases/genetics , Lymphatic Diseases/pathology , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/metabolism , Megakaryocyte Progenitor Cells/cytology , Megakaryocyte Progenitor Cells/metabolism , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Myeloid Cells/immunology , Myeloid Cells/metabolism , Spleen/cytology , Spleen/immunology , Spleen/metabolism , Splenomegaly/genetics , Splenomegaly/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...