Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Eur J Immunol ; 53(12): e2350546, 2023 12.
Article in English | MEDLINE | ID: mdl-37751619

ABSTRACT

Cryopreservation of mouse thymus depletes donor thymocytes but preserves thymus function when transplanted after thawing into athymic mice. No differences in immune reconstitution were observed between fresh and frozen/thawed transplants suggesting that donor thymocyte depletion does not affect outcome. Thus, cryopreservation of thymus may improve outcomes in thymus transplant patients.


Subject(s)
Immune Reconstitution , Thymocytes , Humans , Animals , Mice , Thymus Gland , Cryopreservation
2.
Development ; 148(15)2021 08 01.
Article in English | MEDLINE | ID: mdl-34323272

ABSTRACT

During positive selection at the transition from CD4+CD8+ double-positive (DP) to single-positive (SP) thymocyte, TCR signalling results in appropriate MHC restriction and signals for survival and progression. We show that the pioneer transcription factors Foxa1 and Foxa2 are required to regulate RNA splicing during positive selection of mouse T cells and that Foxa1 and Foxa2 have overlapping/compensatory roles. Conditional deletion of both Foxa1 and Foxa2 from DP thymocytes reduced positive selection and development of CD4SP, CD8SP and peripheral naïve CD4+ T cells. Foxa1 and Foxa2 regulated the expression of many genes encoding splicing factors and regulators, including Mbnl1, H1f0, Sf3b1, Hnrnpa1, Rnpc3, Prpf4b, Prpf40b and Snrpd3. Within the positively selecting CD69+DP cells, alternative RNA splicing was dysregulated in the double Foxa1/Foxa2 conditional knockout, leading to >850 differentially used exons. Many genes important for this stage of T-cell development (Ikzf1-3, Ptprc, Stat5a, Stat5b, Cd28, Tcf7) and splicing factors (Hnrnpab, Hnrnpa2b1, Hnrnpu, Hnrnpul1, Prpf8) showed multiple differentially used exons. Thus, Foxa1 and Foxa2 are required during positive selection to regulate alternative splicing of genes essential for T-cell development, and, by also regulating splicing of splicing factors, they exert widespread control of alternative splicing.


Subject(s)
Alternative Splicing/genetics , Hepatocyte Nuclear Factor 3-alpha/genetics , Hepatocyte Nuclear Factor 3-beta/genetics , RNA Splicing/genetics , Thymocytes/physiology , Animals , Exons/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA Splicing Factors/genetics , T-Lymphocytes/physiology , Thymus Gland/physiology
3.
Development ; 147(19)2020 10 07.
Article in English | MEDLINE | ID: mdl-32907850

ABSTRACT

Pre-T-cell receptor (TCR) signal transduction is required for developing thymocytes to differentiate from CD4-CD8- double-negative (DN) cell to CD4+CD8+ double-positive (DP) cell. Notch signalling is required for T-cell fate specification and must be maintained throughout ß-selection, but inappropriate Notch activation in DN4 and DP cells is oncogenic. Here, we show that pre-TCR signalling leads to increased expression of the transcriptional repressor Bcl6 and that Bcl6 is required for differentiation to DP. Conditional deletion of Bcl6 from thymocytes reduced pre-TCR-induced differentiation to DP cells, disrupted expansion and enrichment of intracellular TCRß+ cells within the DN population and increased DN4 cell death. Deletion also increased Notch1 activation and Notch-mediated transcription in the DP population. Thus, Bcl6 is required in thymocyte development for efficient differentiation from DN3 to DP and to attenuate Notch1 activation in DP cells. Given the importance of inappropriate NOTCH1 signalling in T-cell acute lymphoblastic leukaemia (T-ALL), and the involvement of BCL6 in other types of leukaemia, this study is important to our understanding of T-ALL.


Subject(s)
Receptor, Notch1/metabolism , Thymocytes/cytology , Thymocytes/metabolism , Animals , Blotting, Western , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/physiology , Flow Cytometry , Genotype , Mice , Receptor, Notch1/genetics , Signal Transduction/genetics , Signal Transduction/physiology
4.
Development ; 145(3)2018 01 31.
Article in English | MEDLINE | ID: mdl-29361554

ABSTRACT

Gli3 is a Hedgehog (Hh)-responsive transcription factor that can function as a transcriptional repressor or activator. We show that Gli3 activity in mouse thymic epithelial cells (TECs) promotes positive selection and differentiation from CD4+ CD8+ to CD4+ CD8- single-positive (SP4) cells in the fetal thymus and that Gli3 represses Shh Constitutive deletion of Gli3, and conditional deletion of Gli3 from TECs, reduced differentiation to SP4, whereas conditional deletion of Gli3 from thymocytes did not. Conditional deletion of Shh from TECs increased differentiation to SP4, and expression of Shh was upregulated in the Gli3-deficient thymus. Use of a transgenic Hh reporter showed that the Hh pathway was active in thymocytes, and increased in the Gli3-deficient fetal thymus. Neutralisation of endogenous Hh proteins in the Gli3-/- thymus restored SP4 differentiation, indicating that Gli3 in TECs promotes SP4 differentiation by repression of Shh Transcriptome analysis showed that Hh-mediated transcription was increased whereas TCR-mediated transcription was decreased in Gli3-/- thymocytes compared with wild type.


Subject(s)
Hedgehog Proteins/metabolism , Nerve Tissue Proteins/metabolism , Thymocytes/cytology , Thymocytes/metabolism , Zinc Finger Protein Gli3/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Epithelial Cells/cytology , Female , Gene Expression Profiling , Hedgehog Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Pregnancy , Repressor Proteins/deficiency , Repressor Proteins/genetics , Repressor Proteins/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Thymocytes/immunology , Thymus Gland/cytology , Thymus Gland/embryology , Thymus Gland/metabolism , Zinc Finger Protein Gli3/deficiency , Zinc Finger Protein Gli3/genetics
5.
Pediatr Transplant ; 25(5): e13930, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33326675

ABSTRACT

Paediatric heart transplantation recipients suffer an increased incidence of infectious, autoimmune and allergic problems. The relative roles of thymus excision and immunosuppressive treatments in contributing to these sequelae are not clear. We compared the immunological phenotypes of 25 heart transplant recipients (Tx), 10 children who underwent thymus excision during non-transplantation cardiac surgery (TE) and 25 age range-matched controls, in two age bands: 1-9 and 10-16 years. Significant differences from controls were seen mainly in the younger age band with Tx showing lower CD3 and CD4 cell counts whilst TE showed lower CD8 cell counts. Naïve T cell and recent thymic emigrant proportions and counts were significantly lower than controls in both groups in the lower age band. T cell recombination excision circle (TREC) levels were lower than controls in both groups in both age bands. There were no differences in regulatory T cells, but in those undergoing thymus excision in infancy, their proportions were higher in TE than Tx, a possible direct effect of immunosuppression. T cell receptor V beta spectratyping showed fewer peaks in both groups than in controls (predominantly in the older age band). Thymus excision in infancy was associated with lower CD8 cell counts and higher proportions of Tregs in TE compared to Tx. These data are consistent with thymus excision, particularly in infancy, being the most important influence on immunological phenotype after heart transplantation.


Subject(s)
Heart Transplantation , Immunophenotyping , T-Lymphocytes, Regulatory/immunology , Thymus Gland/surgery , Adolescent , Antibodies, Monoclonal , Child , Child, Preschool , Female , Humans , Immune Tolerance , Immunosuppression Therapy , Infant , Lymphocyte Count , Male
6.
Eur J Immunol ; 49(1): 66-78, 2019 01.
Article in English | MEDLINE | ID: mdl-30365177

ABSTRACT

The interferon-inducible transmembrane (Ifitm/Fragilis) genes encode homologous proteins that are induced by IFNs. Here, we show that IFITM proteins regulate murine CD4+ Th cell differentiation. Ifitm2 and Ifitm3 are expressed in wild-type (WT) CD4+ T cells. On activation, Ifitm3 was downregulated and Ifitm2 was upregulated. Resting Ifitm-family-deficient CD4+ T cells had higher expression of Th1-associated genes than WT and purified naive Ifitm-family-deficient CD4+ T cells differentiated more efficiently to Th1, whereas Th2 differentiation was inhibited. Ifitm-family-deficient mice, but not Ifitm3-deficient mice, were less susceptible than WT to induction of allergic airways disease, with a weaker Th2 response and less severe disease and lower Il4 but higher Ifng expression and IL-27 secretion. Thus, the Ifitm family is important in adaptive immunity, influencing Th1/Th2 polarization, and Th2 immunopathology.


Subject(s)
Hypersensitivity/immunology , Inflammation/immunology , Membrane Proteins/metabolism , Respiratory System/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Animals , Cell Differentiation/genetics , Cells, Cultured , Interferon-gamma/metabolism , Interleukin-27/metabolism , Interleukin-4/metabolism , Lymphocyte Activation/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Th1-Th2 Balance/genetics
7.
Eur J Immunol ; 48(4): 716-719, 2018 04.
Article in English | MEDLINE | ID: mdl-29318612

ABSTRACT

Nude mouse human thymus transplant model: Fresh or cryopreserved and thawed human thymus slices were transplanted subcutaneously into recipient nude mice. Nude mice subsequently produced mouse CD3+ CD4+ T-cells.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , Thymus Gland/cytology , Thymus Gland/transplantation , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Proliferation , Cryopreservation , Humans , Lymphocyte Count , Mice , Mice, Nude , Transplantation, Heterologous
8.
J Autoimmun ; 93: 131-138, 2018 09.
Article in English | MEDLINE | ID: mdl-30061015

ABSTRACT

The Foxa1 and Foxa2 transcription factors are essential for mouse development. Here we show that they are expressed in thymic epithelial cells (TEC) where they regulate TEC development and function, with important consequences for T-cell development. TEC are essential for T-cell differentiation, lineage decisions and repertoire selection. Conditional deletion of Foxa1 and Foxa2 from murine TEC led to a smaller thymus with a greater proportion of TEC and a greater ratio of medullary to cortical TEC. Cell-surface MHCI expression was increased on cortical TEC in the conditional Foxa1Foxa2 knockout thymus, and MHCII expression was reduced on both cortical and medullary TEC populations. These changes in TEC differentiation and MHC expression led to a significant reduction in thymocyte numbers, reduced positive selection of CD4+CD8+ cells to the CD4 lineage, and increased CD8 cell differentiation. Conditional deletion of Foxa1 and Foxa2 from TEC also caused an increase in the medullary TEC population, and increased expression of Aire, but lower cell surface MHCII expression on Aire-expressing mTEC, and increased production of regulatory T-cells. Thus, Foxa1 and Foxa2 in TEC promote positive selection of CD4SP T-cells and modulate regulatory T-cell production and activity, of importance to autoimmunity.


Subject(s)
Epithelial Cells/immunology , Hepatocyte Nuclear Factor 3-alpha/immunology , Hepatocyte Nuclear Factor 3-beta/immunology , T-Lymphocytes, Regulatory/immunology , Thymocytes/immunology , Thymus Gland/immunology , Animals , Autoimmunity , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Epithelial Cells/cytology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Gene Expression Regulation , Hepatocyte Nuclear Factor 3-alpha/deficiency , Hepatocyte Nuclear Factor 3-alpha/genetics , Hepatocyte Nuclear Factor 3-beta/deficiency , Hepatocyte Nuclear Factor 3-beta/genetics , Lymphocyte Activation , Lymphocyte Count , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Organ Size , Signal Transduction , T-Lymphocytes, Regulatory/cytology , Thymocytes/cytology , Thymus Gland/cytology , Transcription Factors/genetics , Transcription Factors/immunology , AIRE Protein
9.
J Cell Sci ; 128(11): 2085-95, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25908851

ABSTRACT

Different tissues contain diverse and dynamic cellular niches, providing distinct signals to tissue-resident or migratory infiltrating immune cells. Hedgehog (Hh) proteins are secreted inter-cellular signalling molecules, which are essential during development and are important in cancer, post-natal tissue homeostasis and repair. Hh signalling mediated by the Hh-responsive transcription factor Gli2 also has multiple roles in T-lymphocyte development and differentiation. Here, we investigate the function of Gli2 in T-cell signalling and activation. Gene transcription driven by the Gli2 transcriptional activator isoform (Gli2A) attenuated T-cell activation and proliferation following T-cell receptor (TCR) stimulation. Expression of Gli2A in T-cells altered gene expression profiles, impaired the TCR-induced Ca(2+) flux and nuclear expression of NFAT2, suppressed upregulation of molecules essential for activation, and attenuated signalling pathways upstream of the AP-1 and NFκB complexes, leading to reduced activation of these important transcription factors. Inhibition of physiological Hh-dependent transcription increased NFκB activity upon TCR ligation. These data are important for understanding the molecular mechanisms of immunomodulation, particularly in tissues where Hh proteins or other Gli-activating ligands such as TGFß are upregulated, including during inflammation, tissue damage and repair, and in tumour microenvironments.


Subject(s)
Kruppel-Like Transcription Factors/genetics , NF-kappa B/genetics , Receptors, Antigen, T-Cell/genetics , Signal Transduction/genetics , Transcription Factor AP-1/genetics , Transcriptional Activation/genetics , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , Gene Expression Regulation/genetics , Hedgehog Proteins/genetics , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , NFATC Transcription Factors/genetics , T-Lymphocytes/metabolism , Transcriptome/genetics , Transforming Growth Factor beta/genetics , Up-Regulation/genetics , Zinc Finger Protein Gli2
10.
J Autoimmun ; 68: 86-97, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26778835

ABSTRACT

Sonic Hedgehog (Shh) is expressed in the thymus, where it regulates T cell development. Here we investigated the influence of Shh on thymic epithelial cell (TEC) development. Components of the Hedgehog (Hh) signalling pathway were expressed by TEC, and use of a Gli Binding Site-green fluorescence protein (GFP) transgenic reporter mouse demonstrated active Hh-dependent transcription in TEC in the foetal and adult thymus. Analysis of Shh-deficient foetal thymus organ cultures (FTOC) showed that Shh is required for normal TEC differentiation. Shh-deficient foetal thymus contained fewer TEC than wild type (WT), the proportion of medullary TEC was reduced relative to cortical TEC, and cell surface expression of MHC Class II molecules was increased on both cortical and medullary TEC populations. In contrast, the Gli3-deficient thymus, which shows increased Hh-dependent transcription in thymic stroma, had increased numbers of TEC, but decreased cell surface expression of MHC Class II molecules on both cortical and medullary TEC. Neutralisation of endogenous Hh proteins in WT FTOC led to a reduction in TEC numbers, and in the proportion of mature Aire-expressing medullary TEC, but an increase in cell surface expression of MHC Class II molecules on medullary TEC. Likewise, conditional deletion of Shh from TEC in the adult thymus resulted in alterations in TEC differentiation and consequent changes in T cell development. TEC numbers, and the proportion of mature Aire-expressing medullary TEC were reduced, and cell surface expression of MHC Class II molecules on medullary TEC was increased. Differentiation of mature CD4 and CD8 single positive thymocytes was increased, demonstrating the regulatory role of Shh production by TEC on T cell development. Treatment of human thymus explants with recombinant Shh or neutralising anti-Shh antibody indicated that the Hedgehog pathway is also involved in regulation of differentiation from DP to mature SP T cells in the human thymus.


Subject(s)
Cell Differentiation , Epithelial Cells/cytology , Epithelial Cells/metabolism , Hedgehog Proteins/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Hedgehog Proteins/genetics , Humans , Mice , Mice, Knockout , Mice, Transgenic , Signal Transduction , Thymocytes/cytology , Thymocytes/immunology , Thymocytes/metabolism , Thymus Gland/immunology
12.
Blood ; 119(20): 4741-51, 2012 May 17.
Article in English | MEDLINE | ID: mdl-22461491

ABSTRACT

The function of Hedgehog signaling in hematopoiesis is controversial, with different experimental systems giving opposing results. Here we examined the role of Desert Hedgehog (Dhh) in the regulation of murine erythropoiesis. Dhh is one of 3 mammalian Hedgehog family proteins. Dhh is essential for testis development and Schwann cell function. We show, by analysis of Dhh-deficient mice, that Dhh negatively regulates multiple stages of erythrocyte differentiation. In Dhh-deficient bone marrow, the common myeloid progenitor (CMP) population was increased, but differentiation from CMP to granulocyte/macrophage progenitor was decreased, and the mature granulocyte population was decreased, compared with wild-type (WT). In contrast, differentiation from CMP to megakaryocyte/erythrocyte progenitor was increased, and the megakaryocyte/erythrocyte progenitor population was increased. In addition, we found that erythroblast populations were Dhh-responsive in vitro and ex vivo and that Dhh negatively regulated erythroblast differentiation. In Dhh-deficient spleen and bone marrow, BFU-Es and erythroblast populations were increased compared with WT. During recovery of hematopoiesis after irradiation, and under conditions of stress-induced erythropoiesis, erythrocyte differentiation was accelerated in both spleen and bone marrow of Dhh-deficient mice compared with WT.


Subject(s)
Erythropoiesis/genetics , Hedgehog Proteins/physiology , Stress, Physiological/physiology , Age Factors , Animals , Bone Marrow/metabolism , Bone Marrow/radiation effects , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Differentiation/radiation effects , Cells, Cultured , Erythroblasts/metabolism , Erythroblasts/physiology , Erythroblasts/radiation effects , Erythropoiesis/physiology , Erythropoiesis/radiation effects , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Recovery of Function/genetics , Recovery of Function/physiology , Signal Transduction/genetics , Signal Transduction/physiology , Signal Transduction/radiation effects , Spleen/metabolism , Spleen/radiation effects , Stress, Physiological/genetics , Stress, Physiological/radiation effects , Whole-Body Irradiation
13.
Trends Pharmacol Sci ; 44(9): 558-560, 2023 09.
Article in English | MEDLINE | ID: mdl-37296035

ABSTRACT

Allograft vasculopathy (AV) leads to chronic rejection of organ transplants, but its causes are obscure. New research from the Jane-Wit laboratory showed that Sonic Hedgehog (SHH) signalling from damaged graft endothelium drives vasculopathy by promoting proinflammatory cytokine production and NLRP3-inflammasome activation in alloreactive CD4+PTCH1hiPD-1hiT memory cells, offering new diagnostic and therapeutic strategies.


Subject(s)
Hedgehog Proteins , Signal Transduction , Humans , Hedgehog Proteins/physiology , Signal Transduction/physiology , Allografts
14.
FEBS J ; 289(24): 8050-8061, 2022 12.
Article in English | MEDLINE | ID: mdl-34614300

ABSTRACT

Epithelial barrier tissues such as the skin and airway form an essential interface between the mammalian host and its external environment. These physical barriers are crucial to prevent damage and disease from environmental insults and allergens. Failure to maintain barrier function against such risks can lead to severe inflammatory disorders, including atopic dermatitis and asthma. Here, we discuss the role of the morphogen Sonic Hedgehog in postnatal skin and lung and the impact of Shh signalling on repair, inflammation, and atopic disease in these tissues.


Subject(s)
Asthma , Hedgehog Proteins , Animals , Humans , Hedgehog Proteins/genetics , Signal Transduction/physiology , Homeostasis , Inflammation , Mammals
15.
Front Immunol ; 13: 890781, 2022.
Article in English | MEDLINE | ID: mdl-36003391

ABSTRACT

Foxa2, a member of the Forkhead box (Fox) family of transcription factors, plays an important role in the regulation of lung function and lung tissue homeostasis. FOXA2 expression is reduced in the lung and airways epithelium of asthmatic patients and in mice absence of Foxa2 from the lung epithelium contributes to airway inflammation and goblet cell hyperplasia. Here we demonstrate a novel role for Foxa2 in the regulation of T helper differentiation and investigate its impact on lung inflammation. Conditional deletion of Foxa2 from T-cells led to increased Th2 cytokine secretion and differentiation, but decreased Th1 differentiation and IFN-γ expression in vitro. Induction of mouse allergic airway inflammation resulted in more severe disease in the conditional Foxa2 knockout than in control mice, with increased cellular infiltration to the lung, characterized by the recruitment of eosinophils and basophils, increased mucus production and increased production of Th2 cytokines and serum IgE. Thus, these experiments suggest that Foxa2 expression in T-cells is required to protect against the Th2 inflammatory response in allergic airway inflammation and that Foxa2 is important in T-cells to maintain the balance of effector cell differentiation and function in the lung.


Subject(s)
Hepatocyte Nuclear Factor 3-beta , Hypersensitivity , Th2 Cells , Animals , Cell Differentiation , Cytokines/metabolism , Hepatocyte Nuclear Factor 3-beta/genetics , Hepatocyte Nuclear Factor 3-beta/metabolism , Hypersensitivity/metabolism , Inflammation/metabolism , Mice , Th2 Cells/metabolism , Transcription Factors/metabolism
16.
Front Immunol ; 12: 737245, 2021.
Article in English | MEDLINE | ID: mdl-34580585

ABSTRACT

Allergic asthma is a common inflammatory airway disease in which Th2 immune response and inflammation are thought to be triggered by inhalation of environmental allergens. Many studies using mouse models and human tissues and genome-wide association have indicated that Sonic Hedgehog (Shh) and the Hedgehog (Hh) signaling pathway are involved in allergic asthma and that Shh is upregulated in the lung on disease induction. We used a papain-induced mouse model of allergic airway inflammation to investigate the impact of systemic pharmacological inhibition of the Hh signal transduction molecule smoothened on allergic airway disease induction and severity. Smoothened-inhibitor treatment reduced the induction of Shh, IL-4, and IL-13 in the lung and decreased serum IgE, as well as the expression of Smo, Il4, Il13, and the mucin gene Muc5ac in lung tissue. Smoothened inhibitor treatment reduced cellular infiltration of eosinophils, mast cells, basophils, and CD4+ T-cells to the lung, and eosinophils and CD4+ T-cells in the bronchoalveolar lavage. In the mediastinal lymph nodes, smoothened inhibitor treatment reduced the number of CD4+ T-cells, and the cell surface expression of Th2 markers ST2 and IL-4rα and expression of Th2 cytokines. Thus, overall pharmacological smoothened inhibition attenuated T-cell infiltration to the lung and Th2 function and reduced disease severity and inflammation in the airway.


Subject(s)
Anti-Asthmatic Agents/administration & dosage , Anti-Inflammatory Agents/administration & dosage , Asthma/drug therapy , Benzimidazoles/administration & dosage , Chemotaxis, Leukocyte/drug effects , Cytokines/metabolism , Lung/drug effects , Phenylurea Compounds/administration & dosage , Pneumonia/drug therapy , Smoothened Receptor/antagonists & inhibitors , Th2 Cells/drug effects , Animals , Asthma/immunology , Asthma/metabolism , Disease Models, Animal , Female , Injections, Intraperitoneal , Lung/immunology , Lung/metabolism , Male , Mice, Inbred C57BL , Pneumonia/immunology , Pneumonia/metabolism , Signal Transduction , Smoothened Receptor/genetics , Smoothened Receptor/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism
17.
Mol Biochem Parasitol ; 229: 1-5, 2019 04.
Article in English | MEDLINE | ID: mdl-30753856

ABSTRACT

Malaria parasite oocysts generate sporozoites by a process termed sporogony. Essential for successful sporogony of Plasmodium berghei in Anopheles stephensi mosquitoes is a complex of six LCCL lectin domain adhesive-like proteins (LAPs). LAP null mutant oocysts undergo growth and mitosis but fail to form sporozoites. At a cytological level, LAP null mutant oocyst development is indistinguishable from its wildtype counterparts for the first week, supporting the hypothesis that LAP null mutant oocysts develop normally before cytokinesis. We show here that LAP1 null mutant oocysts display highly reduced expression of sporozoite proteins and their transcription factors. Our findings indicate that events leading up to the cytokinesis defect in LAP null mutants occur early in oocyst development.


Subject(s)
Gene Expression Regulation , Malaria/parasitology , Oocysts/metabolism , Plasmodium berghei/genetics , Protozoan Proteins/genetics , Animals , Female , Humans , Mice , Mutation , Oocysts/growth & development , Plasmodium berghei/growth & development , Plasmodium berghei/metabolism , Protozoan Proteins/metabolism , Sporozoites/genetics , Sporozoites/growth & development , Sporozoites/metabolism
18.
Front Immunol ; 10: 1629, 2019.
Article in English | MEDLINE | ID: mdl-31379834

ABSTRACT

Here we investigate the function of Hedgehog (Hh) signaling in thymic γδ T-cell maturation and subset differentiation. Analysis of Hh mutants showed that Hh signaling promotes γδ T-cell development in the thymus and influences γδ T-cell effector subset distribution. Hh-mediated transcription in thymic γδ cells increased γδ T-cell number, and promoted their maturation and increased the γδNKT subset, whereas inhibition of Hh-mediated transcription reduced the thymic γδ T-cell population and increased expression of many genes that are normally down-regulated during γδ T-cell maturation. These changes were also evident in spleen, where increased Hh signaling increased γδNKT cells, but reduced CD27-CD44+ and Vγ2+ populations. Systemic in vivo pharmacological Smoothened-inhibition reduced γδ T-cell and γδNKT cells in the thymus, and also reduced splenic γδ T-cell and γδNKT populations, indicating that Hh signaling also influences homeostasis of peripheral γδ T-cell populations. Taken together our data indicate that Sonic Hedgehog is an important determinant of γδ T-cell effector subset differentiation.


Subject(s)
Hedgehog Proteins/immunology , T-Lymphocyte Subsets/immunology , Thymus Gland/immunology , Animals , Cell Differentiation , Mice, Inbred C57BL , Mice, Transgenic , Smoothened Receptor/immunology , Spleen/immunology
19.
J Clin Invest ; 129(8): 3153-3170, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31264977

ABSTRACT

Hedgehog (Hh) proteins regulate development and tissue homeostasis, but their role in atopic dermatitis (AD) remains unknown. We found that on induction of mouse AD, Sonic Hedgehog (Shh) expression in skin, and Hh pathway action in skin T cells were increased. Shh signaling reduced AD pathology and the levels of Shh expression determined disease severity. Hh-mediated transcription in skin T cells in AD-induced mice increased Treg populations and their suppressive function through increased active transforming growth factor-ß (TGF-ß) in Tregs signaling to skin T effector populations to reduce disease progression and pathology. RNA sequencing of skin CD4+ T cells from AD-induced mice demonstrated that Hh signaling increased expression of immunoregulatory genes and reduced expression of inflammatory and chemokine genes. Addition of recombinant Shh to cultures of naive human CD4+ T cells in iTreg culture conditions increased FOXP3 expression. Our findings establish an important role for Shh upregulation in preventing AD, by increased Gli-driven Treg cell-mediated immune suppression, paving the way for a potential new therapeutic strategy.


Subject(s)
Dermatitis, Atopic/immunology , Hedgehog Proteins/immunology , Signal Transduction/immunology , Skin/immunology , T-Lymphocytes, Regulatory/immunology , Zinc Finger Protein Gli2/immunology , Animals , Dermatitis, Atopic/genetics , Dermatitis, Atopic/pathology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Gene Expression Regulation/immunology , Hedgehog Proteins/genetics , Mice , Mice, Knockout , Signal Transduction/genetics , Skin/pathology , T-Lymphocytes, Regulatory/pathology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/immunology , Zinc Finger Protein Gli2/genetics
20.
J Exp Med ; 214(7): 2041-2058, 2017 Jul 03.
Article in English | MEDLINE | ID: mdl-28533268

ABSTRACT

Before birth, B cells develop in the fetal liver (FL). In this study, we show that Gli3 activity in the FL stroma is required for B cell development. In the Gli3-deficient FL, B cell development was reduced at multiple stages, whereas the Sonic hedgehog (Hh [Shh])-deficient FL showed increased B cell development, and Gli3 functioned to repress Shh transcription. Use of a transgenic Hh-reporter mouse showed that Shh signals directly to developing B cells and that Hh pathway activation was increased in developing B cells from Gli3-deficient FLs. RNA sequencing confirmed that Hh-mediated transcription is increased in B-lineage cells from Gli3-deficient FL and showed that these cells expressed reduced levels of B-lineage transcription factors and B cell receptor (BCR)/pre-BCR-signaling genes. Expression of the master regulators of B cell development Ebf1 and Pax5 was reduced in developing B cells from Gli3-deficient FL but increased in Shh-deficient FL, and in vitro Shh treatment or neutralization reduced or increased their expression, respectively.


Subject(s)
B-Lymphocytes/metabolism , Gene Expression Regulation, Developmental , Hedgehog Proteins/genetics , Kruppel-Like Transcription Factors/genetics , Liver/metabolism , Nerve Tissue Proteins/genetics , Animals , Cell Lineage/genetics , Flow Cytometry , Gene Expression Profiling/methods , Liver/embryology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , PAX5 Transcription Factor/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Trans-Activators/genetics , Zinc Finger Protein Gli3
SELECTION OF CITATIONS
SEARCH DETAIL