Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
PLoS Pathog ; 18(8): e1010321, 2022 08.
Article in English | MEDLINE | ID: mdl-35969643

ABSTRACT

Cryptococcosis is a potentially lethal fungal infection of humans caused by organisms within the Cryptococcus neoformans/gattii species complex. Whilst C. neoformans is a relatively common pathogen of immunocompromised individuals, C. gattii is capable of acting as a primary pathogen of immunocompetent individuals. Within the host, both species undergo morphogenesis to form titan cells: exceptionally large cells that are critical for disease establishment. To date, the induction, defining attributes, and underlying mechanism of titanisation have been mainly characterized in C. neoformans. Here, we report the serendipitous discovery of a simple and robust protocol for in vitro induction of titan cells in C. gattii. Using this in vitro approach, we reveal a remarkably high capacity for titanisation within C. gattii, especially in strains associated with the Pacific Northwest Outbreak, and characterise strain-specific differences within the clade. In particular, this approach demonstrates for the first time that cell size changes, DNA amplification, and budding are not always synchronous during titanisation. Interestingly, however, exhibition of these cell cycle phenotypes was correlated with genes associated with cell cycle progression including CDC11, CLN1, BUB2, and MCM6. Finally, our findings reveal exogenous p-Aminobenzoic acid to be a key inducer of titanisation in this organism. Consequently, this approach offers significant opportunities for future exploration of the underlying mechanism of titanisation in this genus.


Subject(s)
Cryptococcus gattii , Cryptococcus neoformans , Fungal Proteins , Humans , Immunocompromised Host , Minichromosome Maintenance Complex Component 6
3.
Blood ; 133(4): 331-343, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30429161

ABSTRACT

Conditional knockout (KO) mouse models are invaluable for elucidating the physiological roles of platelets. The Platelet factor 4-Cre recombinase (Pf4-Cre) transgenic mouse is the current model of choice for generating megakaryocyte/platelet-specific KO mice. Platelets and leukocytes work closely together in a wide range of disease settings, yet the specific contribution of platelets to these processes remains unclear. This is partially a result of the Pf4-Cre transgene being expressed in a variety of leukocyte populations. To overcome this issue, we developed a Gp1ba-Cre transgenic mouse strain in which Cre expression is driven by the endogenous Gp1ba locus. By crossing Gp1ba-Cre and Pf4-Cre mice to the mT/mG dual-fluorescence reporter mouse and performing a head-to-head comparison, we demonstrate more stringent megakaryocyte lineage-specific expression of the Gp1ba-Cre transgene. Broader tissue expression was observed with the Pf4-Cre transgene, leading to recombination in many hematopoietic lineages, including monocytes, macrophages, granulocytes, and dendritic and B and T cells. Direct comparison of phenotypes of Csk, Shp1, or CD148 conditional KO mice generated using either the Gp1ba-Cre or Pf4-Cre strains revealed similar platelet phenotypes. However, additional inflammatory and immunological anomalies were observed in Pf4-Cre-generated KO mice as a result of nonspecific deletion in other hematopoietic lineages. By excluding leukocyte contributions to phenotypes, the Gp1ba-Cre mouse will advance our understanding of the role of platelets in inflammation and other pathophysiological processes in which platelet-leukocyte interactions are involved.


Subject(s)
Blood Platelets/metabolism , Integrases/metabolism , Leukocytes/metabolism , Platelet Glycoprotein GPIb-IX Complex/metabolism , Agglutination , Animals , Bone Marrow Cells/cytology , CSK Tyrosine-Protein Kinase , Cell Lineage , Cell Size , Gene Targeting , Homeostasis , Lymphocyte Count , Megakaryocytes/cytology , Megakaryocytes/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Models, Animal , Phenotype , Platelet Aggregation , Platelet Factor 4/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Recombination, Genetic/genetics , Spleen/cytology , src-Family Kinases/metabolism
4.
Immunity ; 37(4): 721-34, 2012 Oct 19.
Article in English | MEDLINE | ID: mdl-22940098

ABSTRACT

Lymph node development during embryogenesis involves lymphotoxin-ß receptor engagement and subsequent differentiation of a poorly defined population of mesenchymal cells into lymphoid tissue organizer cells. Here, we showed that embryonic mesenchymal cells with characteristics of adipocyte precursors present in the microenvironment of lymph nodes gave rise to lymph node organizer cells. Signaling through the lymphotoxin-ß receptor controlled the fate of adipocyte precursor cells by blocking adipogenesis and instead promoting lymphoid tissue stromal cell differentiation. This effect involved activation of the NF-κB2-RelB signaling pathway and inhibition of the expression of the key adipogenic factors Pparγ and Cebpα. In vivo organogenesis assays show that embryonic and adult adipocyte precursor cells can migrate into newborn lymph nodes and differentiate into a variety of lymph node stromal cells. Thus, we propose that adipose tissues act as a source of lymphoid stroma for lymph nodes and other lymphoid structures associated with fat.


Subject(s)
Adipocytes/immunology , Lymph Nodes/immunology , Signal Transduction , Adipocytes/cytology , Animals , Cell Differentiation , Cell Movement , Cells, Cultured , Lymphotoxin beta Receptor/immunology , Mice , NF-kappa B p52 Subunit/immunology , NF-kappa B p52 Subunit/metabolism , Phenotype , Stromal Cells/immunology , Transcription Factor RelB/immunology , Transcription Factor RelB/metabolism
5.
Immunity ; 36(3): 427-37, 2012 Mar 23.
Article in English | MEDLINE | ID: mdl-22425250

ABSTRACT

The thymic medulla provides a specialized microenvironment for the negative selection of T cells, with the presence of autoimmune regulator (Aire)-expressing medullary thymic epithelial cells (mTECs) during the embryonic-neonatal period being both necessary and sufficient to establish long-lasting tolerance. Here we showed that emergence of the first cohorts of Aire(+) mTECs at this key developmental stage, prior to αß T cell repertoire selection, was jointly directed by Rankl(+) lymphoid tissue inducer cells and invariant Vγ5(+) dendritic epidermal T cell (DETC) progenitors that are the first thymocytes to express the products of gene rearrangement. In turn, generation of Aire(+) mTECs then fostered Skint-1-dependent, but Aire-independent, DETC progenitor maturation and the emergence of an invariant DETC repertoire. Hence, our data attributed a functional importance to the temporal development of Vγ5(+) γδ T cells during thymus medulla formation for αß T cell tolerance induction and demonstrated a Rank-mediated reciprocal link between DETC and Aire(+) mTEC maturation.


Subject(s)
Precursor Cells, T-Lymphoid/cytology , Precursor Cells, T-Lymphoid/immunology , Receptor Activator of Nuclear Factor-kappa B/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Transcription Factors/immunology , Animals , Cell Differentiation/immunology , Cellular Microenvironment , Epithelial Cells/immunology , Female , Fetus/cytology , Fetus/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pregnancy , Signal Transduction/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Transcription Factors/deficiency , Transcription Factors/genetics , AIRE Protein
6.
Eur J Immunol ; 45(9): 2484-93, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26173808

ABSTRACT

Expression of mouse C-type lectin-like receptor 2 (CLEC-2) has been reported on circulating CD11b(high) Gr-1(high) myeloid cells and dendritic cells (DCs) under basal conditions, as well as on a variety of leucocyte subsets following inflammatory stimuli or in vitro cell culture. However, previous studies assessing CLEC-2 expression failed to use CLEC-2-deficient mice as negative controls and instead relied heavily on single antibody clones. Here, we generated CLEC-2-deficient adult mice using two independent approaches and employed two anti-mouse CLEC-2 antibody clones to investigate surface expression on hematopoietic cells from peripheral blood and secondary lymphoid organs. We rule out constitutive CLEC-2 expression on resting DCs and show that CLEC-2 is upregulated in response to LPS-induced systemic inflammation in a small subset of activated DCs isolated from the mesenteric lymph nodes but not the spleen. Moreover, we demonstrate for the first time that peripheral blood B lymphocytes present exogenously derived CLEC-2 and suggest that both circulating B lymphocytes and CD11b(high) Gr-1(high) myeloid cells lose CLEC-2 following entry into secondary lymphoid organs. These results have significant implications for our understanding of CLEC-2 physiological functions.


Subject(s)
B-Lymphocytes/immunology , Dendritic Cells/immunology , Gene Expression Regulation/immunology , Lectins, C-Type/genetics , Myeloid Cells/immunology , Animals , Antibodies, Monoclonal/pharmacology , B-Lymphocytes/pathology , Blood Platelets/immunology , Blood Platelets/pathology , CD11b Antigen/genetics , CD11b Antigen/immunology , Cell Movement/immunology , Dendritic Cells/pathology , Inflammation/chemically induced , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Lectins, C-Type/antagonists & inhibitors , Lectins, C-Type/deficiency , Lipopolysaccharides , Lymph Nodes/immunology , Lymph Nodes/pathology , Mice , Mice, Transgenic , Myeloid Cells/pathology , Organ Specificity , Receptors, Chemokine/genetics , Receptors, Chemokine/immunology , Signal Transduction , Spleen/immunology , Spleen/pathology
7.
Blood ; 123(20): 3200-7, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24532804

ABSTRACT

The importance of CLEC-2, a natural ligand/receptor for Gp38/Podoplanin, in the formation of the lymphatic vasculature has recently been demonstrated. As the development and maintenance of lymph nodes (LNs) is dependent on the formation of the lymphatic vasculature and the differentiation of Gp38/Podoplanin(+) stromal cells, we investigated the role of CLEC-2 in lymphoneogenesis and LN homeostasis. Using constitutive Clec1b(-/-) mice, we showed that while CLEC-2 was not necessary for initiation of the LN anlage, it was required at late stages of development. Constitutive deletion of CLEC-2 induced a profound defect in lymphatic endothelial cell proliferation, resulting in lack of LNs at birth. In contrast, conditional deletion of CLEC-2 in the megakaryocyte/platelet lineage in Clec1b(fl/fl)PF4-Cre mice led to the development of blood-filled LNs and fibrosis, in absence of a proliferative defect of the lymphatic endothelial compartment. This phenotype was also observed in chimeric mice reconstituted with Clec1b(fl/fl)PF4-Cre bone marrow, indicating that CLEC-2 expression in platelets was required for LN integrity. We demonstrated that LNs of Clec1b(fl/fl)PF4-Cre mice are able to sustain primary immune responses but show a defect in immune cell recirculation after repeated immunizations, thus suggesting CLEC-2 as target in chronic immune response.


Subject(s)
Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Lymph Nodes/growth & development , Animals , Blood Platelets/metabolism , Cell Proliferation , Cells, Cultured , Endothelium, Lymphatic/cytology , Endothelium, Lymphatic/metabolism , Gene Deletion , Lymph Nodes/cytology , Lymph Nodes/metabolism , Lymphangiogenesis , Megakaryocytes/metabolism , Mice , Mice, Inbred C57BL
8.
J Immunol ; 189(12): 5519-26, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23152561

ABSTRACT

T cell tolerance in the thymus is a key step in shaping the developing T cell repertoire. Thymic medullary epithelial cells play multiple roles in this process, including negative selection of autoreactive thymocytes, influencing thymic dendritic cell positioning, and the generation of Foxp3(+) regulatory T cells. Previous studies show that medullary thymic epithelial cell (mTEC) development involves hemopoietic cross-talk, and numerous TNFR superfamily members have been implicated in this process. Whereas CD40 and RANK represent key examples, interplay between these receptors, and the individual cell types providing their ligands at both fetal and adult stages of thymus development, remain unclear. In this study, by analysis of the cellular sources of receptor activator for NF-κB ligand (RANKL) and CD40L during fetal and adult cross-talk in the mouse, we show that the innate immune cell system drives initial fetal mTEC development via expression of RANKL, but not CD40L. In contrast, cross-talk involving the adaptive immune system involves both RANKL and CD40L, with analysis of distinct subsets of intrathymic CD4(+) T cells revealing a differential contribution of CD40L by conventional, but not Foxp3(+) regulatory, T cells. We also provide evidence for a stepwise involvement of TNFRs in mTEC development, with CD40 upregulation induced by initial RANK signaling subsequently controlling proliferation within the mTEC compartment. Collectively, our findings show how multiple hemopoietic cell types regulate mTEC development through differential provision of RANKL/CD40L during ontogeny, revealing molecular differences in fetal and adult hemopoietic cross-talk. They also suggest a stepwise process of mTEC development, in which RANK is a master player in controlling the availability of other TNFR family members.


Subject(s)
CD40 Ligand/metabolism , Cellular Senescence/immunology , Gene Expression Regulation, Developmental/immunology , RANK Ligand/biosynthesis , Receptor Cross-Talk/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD40 Ligand/genetics , CD40 Ligand/physiology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cellular Senescence/genetics , Epithelial Cells/cytology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Fetus/immunology , Immunity, Innate/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Organ Culture Techniques , RANK Ligand/genetics , Signal Transduction/genetics , Signal Transduction/immunology , Thymus Gland/metabolism
9.
J Immunol ; 189(5): 2094-8, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22855716

ABSTRACT

Phylogeny shows that CD4 T cell memory and lymph nodes coevolved in placental mammals. In ontogeny, retinoic acid orphan receptor (ROR)γ-dependent lymphoid tissue inducer (LTi) cells program the development of mammalian lymph nodes. In this study, we show that although primary CD4 T cell expansion is normal in RORγ-deficient mice, the persistence of memory CD4 T cells is RORγ-dependent. Furthermore, using bone marrow chimeric mice we demonstrate that LTi cells are the key RORγ-expressing cell type sufficient for memory CD4 T cell survival in the absence of persistent Ag. This effect was specific for CD4 T cells, as memory CD8 T cells survived equally well in the presence or absence of LTi cells. These data demonstrate a novel role for LTi cells, archetypal members of the innate lymphoid cell family, in supporting memory CD4 T cell survival in vivo.


Subject(s)
Immunologic Memory , Lymphoid Tissue/immunology , T-Lymphocytes, Helper-Inducer/immunology , Adaptive Immunity/genetics , Animals , Cell Death/genetics , Cell Death/immunology , Cell Survival/genetics , Cell Survival/immunology , Immunity, Innate/genetics , Immunologic Memory/genetics , Lymphoid Tissue/cytology , Lymphoid Tissue/transplantation , Lymphopenia/genetics , Lymphopenia/immunology , Lymphopenia/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nuclear Receptor Subfamily 1, Group F, Member 3/deficiency , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Radiation Chimera/immunology , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/pathology
10.
J Immunol ; 189(9): 4266-74, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22993205

ABSTRACT

Thymic atrophy is a frequent consequence of infection with bacteria, viruses, and parasites and is considered a common virulence trait between pathogens. Multiple reasons have been proposed to explain this atrophy, including premature egress of immature thymocytes, increased apoptosis, or thymic shutdown to prevent tolerance to the pathogen from developing. The severe loss in thymic cell number can reflect an equally dramatic reduction in thymic output, potentially reducing peripheral T cell numbers. In this study, we examine the relationship between systemic Salmonella infection and thymic function. During infection, naive T cell numbers in peripheral lymphoid organs increase. Nevertheless, this occurs despite a pronounced thymic atrophy caused by viable bacteria, with a peak 50-fold reduction in thymocyte numbers. Thymic atrophy is not dependent upon homeostatic feedback from peripheral T cells or on regulation of endogenous glucocorticoids, as demonstrated by infection of genetically altered mice. Once bacterial numbers fall, thymocyte numbers recover, and this is associated with increases in the proportion and proliferation of early thymic progenitors. During atrophy, thymic T cell maturation is maintained, and single-joint TCR rearrangement excision circle analysis reveals there is only a modest fall in recent CD4(+) thymic emigrants in secondary lymphoid tissues. Thus, thymic atrophy does not necessarily result in a matching dysfunctional T cell output, and thymic homeostasis can constantly adjust to systemic infection to ensure that naive T cell output is maintained.


Subject(s)
Recovery of Function/immunology , Salmonella Infections/immunology , Thymus Gland/immunology , Thymus Gland/pathology , Animals , Atrophy , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/microbiology , Cell Differentiation/immunology , Cell Movement/immunology , Mice , Salmonella Infections/pathology , Salmonella Infections/physiopathology , Salmonella typhimurium/immunology , Thymus Gland/microbiology
11.
J Immunol ; 186(9): 5227-35, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21421850

ABSTRACT

The entry of T cell progenitors to the thymus marks the beginning of a multistage developmental process that culminates in the generation of self-MHC-restricted CD4(+) and CD8(+) T cells. Although multiple factors including the chemokine receptors CCR7 and CCR9 are now defined as important mediators of progenitor recruitment and colonization in both the fetal and adult thymi, the heterogeneity of thymus-colonizing cells that contribute to development of the T cell pool is complex and poorly understood. In this study, in conjunction with lineage potential assays, we perform phenotypic and genetic analyses on thymus-settling progenitors (TSP) isolated from the embryonic mouse thymus anlagen and surrounding perithymic mesenchyme, including simultaneous gene expression analysis of 14 hemopoietic regulators using single-cell multiplex RT-PCR. We show that, despite the known importance of CCL25-CCR9 mediated thymic recruitment of T cell progenitors, embryonic PIR(+)c-Kit(+) TSP can be subdivided into CCR9(+) and CCR9(-) subsets that differ in their requirements for a functional thymic microenvironment for thymus homing. Despite these differences, lineage potential studies of purified CCR9(+) and CCR9(-) TSP reveal a common bias toward T cell-committed progenitors, and clonal gene expression analysis reveals a genetic consensus that is evident between and within single CCR9(+) and CCR9(-) TSP. Collectively, our data suggest that although the earliest T cell progenitors may display heterogeneity with regard to their requirements for thymus colonization, they represent a developmentally homogeneous progenitor pool that ensures the efficient generation of the first cohorts of T cells during thymus development.


Subject(s)
Cell Lineage , Gene Expression Profiling , Lymphoid Progenitor Cells/cytology , Lymphopoiesis , Receptors, CCR/metabolism , T-Lymphocytes/cytology , Thymus Gland/cytology , Animals , Apoptosis/immunology , Cell Differentiation/immunology , Cell Separation , Clone Cells , Embryo, Mammalian , Flow Cytometry , Lymphoid Progenitor Cells/immunology , Lymphoid Progenitor Cells/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microdissection , Receptors, CCR/immunology , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Thymus Gland/embryology
12.
Nat Commun ; 14(1): 4895, 2023 08 14.
Article in English | MEDLINE | ID: mdl-37580395

ABSTRACT

The opportunistic fungal pathogen Cryptococcus neoformans causes lethal infections in immunocompromised patients. Macrophages are central to the host response to cryptococci; however, it is unclear how C. neoformans is recognised and phagocytosed by macrophages. Here we investigate the role of TLR4 in the non-opsonic phagocytosis of C. neoformans. We find that loss of TLR4 function unexpectedly increases phagocytosis of non-opsonised cryptococci by murine and human macrophages. The increased phagocytosis observed in Tlr4-/- cells was dampened by pre-treatment of macrophages with oxidised-LDL, a known ligand of scavenger receptors. The scavenger receptor, macrophage scavenger receptor 1 (MSR1) (also known as SR-A1 or CD204) was upregulated in Tlr4-/- macrophages. Genetic ablation of MSR1 resulted in a 75% decrease in phagocytosis of non-opsonised cryptococci, strongly suggesting that it is a key non-opsonic receptor for this pathogen. We go on to show that MSR1-mediated uptake likely involves the formation of a multimolecular signalling complex involving FcγR leading to SYK, PI3K, p38 and ERK1/2 activation to drive actin remodelling and phagocytosis. Altogether, our data indicate a hitherto unidentified role for TLR4/MSR1 crosstalk in the non-opsonic phagocytosis of C. neoformans.


Subject(s)
Cryptococcosis , Phagocytosis , Scavenger Receptors, Class A , Toll-Like Receptor 4 , Animals , Humans , Mice , Cryptococcus neoformans , Macrophages/microbiology , Toll-Like Receptor 4/genetics , Scavenger Receptors, Class A/metabolism
13.
J Immunol ; 182(1): 130-7, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19109143

ABSTRACT

In the thymus, interactions between immature thymocytes and thymic epithelial cells (TECs) regulate the development and selection of self-tolerant MHC-restricted T cells. Despite the importance of cortical (cTEC) and medullary (mTEC) thymic epithelial cells in fostering T cell production, events in TEC development are still unclear. Although precursor-product relationships during mTEC development have been reported, and some genetic regulators of mTEC development have been identified, stages in cTEC development occurring downstream of recently identified bipotent cTEC/mTEC progenitors remain poorly defined. In this study, we combine analysis of differentiation, proliferation, and gene expression of TECs in the murine thymus, that has enabled us to identify cTEC progenitors, define multiple stages in cTEC development, and identify novel checkpoints in development of the cTEC lineage. We show an essential requirement for FoxN1 in the initial development of cTEC from bipotent progenitors, and demonstrate a stage-specific requirement for CD4(-)8(-) thymocytes in later stages of cTEC development. Collectively, our data establish a program of cTEC development that should provide insight into the formation and function of the thymic cortex for T cell development.


Subject(s)
Cell Differentiation/immunology , Epithelial Cells/cytology , Epithelial Cells/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Animals , Cell Communication/immunology , Cell Lineage/immunology , Cell Proliferation , Epithelial Cells/metabolism , Histocompatibility Antigens Class II/biosynthesis , Histocompatibility Antigens Class II/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Mice, Transgenic , Organ Culture Techniques , Precursor Cells, T-Lymphoid/cytology , Precursor Cells, T-Lymphoid/immunology , Precursor Cells, T-Lymphoid/metabolism , Signal Transduction/immunology , Thymus Gland/embryology , Thymus Gland/metabolism
14.
Eur J Immunol ; 39(9): 2395-402, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19662637

ABSTRACT

Cortical and medullary thymic epithelial cells provide essential signals for a normal programme of T-cell development. Current models of thymus development suggest that thymocyte-derived signals play an important role in establishing thymic microenvironments, a process termed thymus crosstalk. Studies on CD3epsilontg26 mice lacking intrathymic T-cell progenitors provided evidence that normal development of the thymic cortex depends upon thymocyte-derived signals. Importantly, the reported failure to effectively reconstitute adult CD3epsilontg26 mice raised the possibility that such crosstalk must occur within a developmental window, and that closure of this window during the postnatal period renders thymic epithelium refractory to crosstalk signals and unable to effectively impose T-cell selection. We have re-investigated the timing of provision of crosstalk in relation to development of functional thymic microenvironments. We show that transfer of either fetal precursors or adult T-committed precursors into adult CD3epsilontg26 mice initiates key parameters of successful thymic reconstitution including thymocyte development and emigration, restoration of cortical and medullary epithelial architecture, and establishment of thymic tolerance mechanisms including maturation of Foxp3(+) Treg and autoimmune regulator-expressing medullary epithelium. Collectively, our data argue against a temporal window of thymocyte crosstalk, and instead demonstrates continued receptiveness of thymic epithelium for the formation of functionally competent thymic microenvironments.


Subject(s)
Epithelial Cells/immunology , Fetus/immunology , Lymphoid Progenitor Cells/immunology , T-Lymphocytes, Regulatory/immunology , Thymus Gland/immunology , Animals , CD3 Complex/genetics , Epithelial Cells/metabolism , Hematopoiesis, Extramedullary , Immune Tolerance , Lymphoid Progenitor Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , T-Lymphocytes, Regulatory/metabolism , Thymus Gland/cytology
15.
Cell Host Microbe ; 28(6): 774-775, 2020 12 09.
Article in English | MEDLINE | ID: mdl-33301715

ABSTRACT

Phagocytes engulf pathogens into a membrane bound compartment called a phagosome, but what happens when engulfed pathogens start growing? In this issue of Cell Host & Microbe,Westman et al. (2020) show that lysosomes fuse with phagosomes to maintain phagosomal membrane integrity as the fungal pathogen Candida albicans expands.


Subject(s)
Mycoses , Phagosomes , Candida albicans , Humans , Lysosomes , Phagocytes
16.
J Leukoc Biol ; 83(5): 1145-54, 2008 May.
Article in English | MEDLINE | ID: mdl-18270249

ABSTRACT

Hematopoiesis occurs in different tissues during adult and fetal life. Splenic hematopoiesis arises in the fetal period until the first weeks of life. We have analyzed the hematopoietic progenitor content of the fetal spleen (FS) at the embryonic days 14.5-15.5. We first demonstrate that the hematopoietic content of the FS differs largely from its fetal liver (FL) counterpart. The difference mainly concerns the distribution of the different pool of progenitors, as most of the splenic progenitors are comprised in the lineage(-)Sca1(-)cKit(lo) contrary to the FL. We have divided the fetal hematopoietic pool into smaller fractions to enable characterization of the earliest lymphoid progenitors. Among the lymphoid progenitors that already represent a rare population, we were able to separate a population, respectively, enriched in B or T/NK progenitors. Lineage restriction of the different developmental intermediates was tested by clonal assays. We propose a model for fetal splenic hematopoietic progenitors and their distribution.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Lymphocytes/immunology , Spleen/immunology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , CD4-Positive T-Lymphocytes/cytology , Cell Differentiation , Crosses, Genetic , DNA-Binding Proteins/deficiency , Female , Fetus , Flow Cytometry , Hematopoiesis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Pregnancy , Spleen/cytology
17.
J Leukoc Biol ; 84(1): 152-61, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18403646

ABSTRACT

For the formation of lymph nodes and Peyer's patches, lymphoid tissue inducer (LTi) cells are crucial in triggering stromal cells to recruit and retain hematopoietic cells. Although LTi cells have been observed in fetal spleen, not much is known about fetal spleen development and the role of LTi cells in this process. Here, we show that LTi cells collect in a periarteriolar manner in fetal spleen at the periphery of the white pulp anlagen. Expression of the homeostatic chemokines can be detected in stromal and endothelial cells, suggesting that LTi cells are attracted by these chemokines. As lymphotoxin (LT)alpha1beta2 can be detected on B cells but not LTi cells in neonatal spleen, starting at 4 days after birth, the earliest formation of the white pulp in fetal spleen occurs in a LTalpha1beta2-independent manner. The postnatal development of the splenic white pulp, involving the influx of T cells, depends on LTalpha1beta2 expressed by B cells.


Subject(s)
Lymphotoxin alpha1, beta2 Heterotrimer/metabolism , Spleen/cytology , Spleen/embryology , Animals , Animals, Newborn , Humans , Mice , Mice, Inbred C57BL , Stromal Cells/cytology
18.
Lab Anim ; 52(1): 59-68, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28480797

ABSTRACT

Transgenic tumour necrosis factor alpha (TNFα)-driven models of polyarthritis such as the TNFΔARE mouse have proven to be invaluable in delineating aspects of inflammatory disease pathophysiology in humans. Unfortunately, the onset of joint destruction and inflammation in these models represents a significant detriment to breeding management. We examined whether TNFα depleting therapy 'infliximab' might represent a significant refinement in routine breeding. Clinical scores of joint inflammation were assessed in TNFΔARE males receiving either infliximab (10 mg/kg) or saline by twice-weekly intraperitoneal injection. Joint histology and bone morphology were assessed by histological analysis and micro-computed tomography (CT), respectively. Analysis of breeding was examined retrospectively in TNFΔARE males prior to, and following, regular introduction of infliximab. Clinical scores of inflammation were significantly reduced in TNFΔARE males receiving infliximab (control 6.6 arbitrary units [AU] ± 0.88 versus infliximab 4.4 AU ± 1.4; P < 0.05), while measures of pannus invasion and bone erosion by histology and micro-CT were markedly reduced. In the breeding groups, TNFΔARE males receiving infliximab injections sired more litters over their breeding lifespan (control 1.69 ± 0.22 versus infliximab 3.00 ± 0.19; P < 0.005). Furthermore, prior to infliximab, TNFΔARE males had a 26% risk of failing to sire any litters. This was reduced to 7% after the introduction of infliximab. This study is the first to report that regular administration of infliximab is effective at suppressing disease activity and improving animal welfare in TNFΔARE animals. In addition, we have shown that infliximab is highly efficacious in improving breeding behaviour and increasing the number of litters sired by TNFΔARE males.


Subject(s)
Antirheumatic Agents/pharmacology , Arthritis/genetics , Fertility/drug effects , Infliximab/pharmacology , Mice , Rodent Diseases/prevention & control , Tumor Necrosis Factor-alpha/metabolism , Animal Welfare , Animals , Breeding , Disease Models, Animal , Male , Mice, Transgenic
19.
Med Image Anal ; 40: 30-43, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28595091

ABSTRACT

Rheumatoid arthritis (RA) is an autoimmune disease in which chronic inflammation of the synovial joints can lead to destruction of cartilage and bone. Pre-clinical studies attempt to uncover the underlying causes by emulating the disease in genetically different mouse strains and characterising the nature and severity of bone shape changes as indicators of pathology. This paper presents a fully automated method for obtaining quantitative measurements of bone destruction from volumetric micro-CT images of a mouse hind paw. A statistical model of normal bone morphology derived from a training set of healthy examples serves as a template against which a given pathological sample is compared. Abnormalities in bone shapes are identified as deviations from the model statistics, characterised in terms of type (erosion / formation) and quantified in terms of severity (percentage affected bone area). The colour-coded magnitudes of the deviations superimposed on a three-dimensional rendering of the paw show at a glance the severity of malformations for the individual bones and joints. With quantitative data it is possible to derive population statistics characterising differences in bone malformations for different mouse strains and in different anatomical regions. The method was applied to data acquired from three different mouse strains. The derived quantitative indicators of bone destruction have shown agreement both with the subjective visual scores and with the previous biological findings. This suggests that pathological bone shape changes can be usefully and objectively identified as deviations from the model statistics.


Subject(s)
Algorithms , Arthritis, Rheumatoid/diagnostic imaging , Arthritis, Rheumatoid/pathology , Bone and Bones/diagnostic imaging , Bone and Bones/pathology , Imaging, Three-Dimensional/methods , Animals , Disease Models, Animal , Mice
20.
Arthritis Res Ther ; 18(1): 270, 2016 11 18.
Article in English | MEDLINE | ID: mdl-27863512

ABSTRACT

BACKGROUND: We investigated two distinct synovial fibroblast populations that were located preferentially in the lining or sub-lining layers and defined by their expression of either podoplanin (PDPN) or CD248, and explored their ability to undergo self-assembly and transmigration in vivo. METHODS: Synovial fibroblasts (SF) were cultured in vitro and phenotypic changes following stimulation with interleukin (IL)-1ß, tumor necrosis factor (TNF)-α, and transforming growth factor (TGF)-ß1 were examined. To examine the phenotype of SF in vivo, a severe combined immunodeficiency (SCID) human-mouse model of cartilage destruction was utilised. RESULTS: SF in the lining layer in rheumatoid arthritis (RA) expressed high levels of PDPN compared to the normal synovium, whereas CD248 expression was restricted to sub-lining layer cells. TNF-α or IL1 stimulation in vitro resulted in an increased expression of PDPN. In contrast, stimulation with TGF-ß1 induced CD248 expression. In the SCID human-mouse model, rheumatoid SF recapitulated the expression of PDPN and CD248. Fibroblasts adjacent to cartilage expressed PDPN, and attached to, invaded, and degraded cartilage. PDPN+ CD248- SF preceded the appearance of PDPN- CD248+ cells in contralateral implants. CONCLUSIONS: We have identified two distinct SF populations identified by expression of either PDPN or CD248 which are located within different anatomical compartments of the inflamed synovial membrane. These markers discriminate between SF subsets with distinct biological properties. As PDPN-expressing cells are associated with early fibroblast migration and cartilage erosion in vivo, we propose that PDPN-expressing cells may be an attractive therapeutic target in RA.


Subject(s)
Arthritis, Rheumatoid/pathology , Fibroblasts/cytology , Synovial Membrane/cytology , Aged , Animals , Antigens, CD/metabolism , Antigens, Neoplasm/metabolism , Cartilage, Articular/metabolism , Cell Differentiation/physiology , Cytokines/metabolism , Female , Flow Cytometry , Heterografts , Humans , Male , Membrane Glycoproteins/metabolism , Mice , Mice, SCID , Microscopy, Fluorescence , Middle Aged , Polymerase Chain Reaction , Transendothelial and Transepithelial Migration/physiology
SELECTION OF CITATIONS
SEARCH DETAIL