Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Nat Rev Cancer ; 17(3): 145-159, 2017 03.
Article in English | MEDLINE | ID: mdl-28154375

ABSTRACT

The Notch signalling cascade is an evolutionarily conserved pathway that has a crucial role in regulating development and homeostasis in various tissues. The cellular processes and events that it controls are diverse, and continued investigation over recent decades has revealed how the role of Notch signalling is multifaceted and highly context dependent. Consistent with the far-reaching impact that Notch has on development and homeostasis, aberrant activity of the pathway is also linked to the initiation and progression of several malignancies, and Notch can in fact be either oncogenic or tumour suppressive depending on the tissue and cellular context. The Notch pathway therefore represents an important target for therapeutic agents designed to treat many types of cancer. In this Review, we focus on the latest developments relating specifically to the tumour-suppressor activity of Notch signalling and discuss the potential mechanisms by which Notch can inhibit carcinogenesis in various tissues. Potential therapeutic strategies aimed at restoring or augmenting Notch-mediated tumour suppression will also be highlighted.


Subject(s)
Receptors, Notch/physiology , Tumor Suppressor Proteins/physiology , Animals , Cell Differentiation , Humans , Neoplasms/etiology , Signal Transduction/physiology
2.
J Cell Sci ; 130(6): 1021-1025, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28202689

ABSTRACT

The corneal epithelium acts as a protective barrier on the anterior ocular surface and is essential for maintaining transparency of the cornea and thus visual acuity. During both homeostasis and repair, the corneal epithelium is maintained by self-renewing stem cells, which persist throughout the lifetime of the organism. Importantly, as in other self-renewing tissues, the functional activity of corneal epithelial stem cells (CSCEs) is tightly regulated by the surrounding microenvironment, or niche, which provides a range of cues that maintain the stem cell population. This Cell Science at a Glance article and the accompanying poster will therefore aim to summarise our current understanding of the corneal epithelial stem cell niche and its role in regulating stem cell activity during homeostasis, repair and disease.


Subject(s)
Epithelium, Corneal/cytology , Stem Cell Niche , Stem Cells/cytology , Animals , Humans , Models, Biological
4.
PLoS One ; 11(3): e0151666, 2016.
Article in English | MEDLINE | ID: mdl-26983083

ABSTRACT

Thymus function requires extensive cross-talk between developing T-cells and the thymic epithelium, which consists of cortical and medullary TEC. The transcription factor FOXN1 is the master regulator of TEC differentiation and function, and declining Foxn1 expression with age results in stereotypical thymic involution. Understanding of the dynamics of Foxn1 expression is, however, limited by a lack of single cell resolution data. We have generated a novel reporter of Foxn1 expression, Foxn1G, to monitor changes in Foxn1 expression during embryogenesis and involution. Our data reveal that early differentiation and maturation of cortical and medullary TEC coincides with precise sub-lineage-specific regulation of Foxn1 expression levels. We further show that initiation of thymic involution is associated with reduced cTEC functionality, and proportional expansion of FOXN1-negative TEC in both cortical and medullary sub-lineages. Cortex-specific down-regulation of Foxn1 between 1 and 3 months of age may therefore be a key driver of the early stages of age-related thymic involution.


Subject(s)
Cell Differentiation/physiology , Embryonic Development/physiology , Epithelial Cells/metabolism , Forkhead Transcription Factors/metabolism , Thymus Gland/metabolism , Aging/physiology , Animals , Cell Lineage/physiology , Down-Regulation , Forkhead Transcription Factors/genetics , Mice
5.
Nat Cell Biol ; 18(2): 168-80, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26689676

ABSTRACT

Chronic inflammation is associated with a variety of pathological conditions in epithelial tissues, including cancer, metaplasia and aberrant wound healing. In relation to this, a significant body of evidence suggests that aberration of epithelial stem and progenitor cell function is a contributing factor in inflammation-related disease, although the underlying cellular and molecular mechanisms remain to be fully elucidated. In this study, we have delineated the effect of chronic inflammation on epithelial stem/progenitor cells using the corneal epithelium as a model tissue. Using a combination of mouse genetics, pharmacological approaches and in vitro assays, we demonstrate that chronic inflammation elicits aberrant mechanotransduction in the regenerating corneal epithelium. As a consequence, a YAP-TAZ/ß-catenin cascade is triggered, resulting in the induction of epidermal differentiation on the ocular surface. Collectively, the results of this study demonstrate that chronic inflammation and mechanotransduction are linked and act to elicit pathological responses in regenerating epithelia.


Subject(s)
Cell Differentiation , Corneal Injuries/metabolism , Epithelial Cells/metabolism , Epithelium, Corneal/metabolism , Keratitis/metabolism , Mechanotransduction, Cellular , Regeneration , Stem Cells/metabolism , Acyltransferases , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Administration, Ophthalmic , Animals , Anti-Inflammatory Agents/administration & dosage , Cell Cycle Proteins , Cell Differentiation/drug effects , Chronic Disease , Corneal Injuries/genetics , Corneal Injuries/pathology , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/pathology , Epithelium, Corneal/drug effects , Epithelium, Corneal/injuries , Epithelium, Corneal/pathology , Extracellular Matrix/metabolism , HEK293 Cells , Humans , Inflammation Mediators/metabolism , Keratitis/genetics , Keratitis/pathology , Keratitis/prevention & control , Mechanotransduction, Cellular/drug effects , Mice, Inbred C57BL , Mice, Knockout , Phosphoproteins/genetics , Phosphoproteins/metabolism , Receptor, Notch1/deficiency , Receptor, Notch1/genetics , Regeneration/drug effects , Stem Cells/drug effects , Stem Cells/pathology , Swine , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection , Wnt Signaling Pathway , YAP-Signaling Proteins , beta Catenin/genetics , beta Catenin/metabolism
6.
PLoS One ; 9(12): e114842, 2014.
Article in English | MEDLINE | ID: mdl-25531271

ABSTRACT

Normal thymus function reflects interactions between developing T-cells and several thymic stroma cell types. Within the stroma, key functions reside in the distinct cortical and medullary thymic epithelial cell (TEC) types. It has been demonstrated that, during organogenesis, all TECs can be derived from a common thymic epithelial progenitor cell (TEPC). The properties of this common progenitor are thus of interest. Differentiation of both cTEC and mTEC depends on the epithelial-specific transcription factor FOXN1, although formation of the common TEPC from which the TEC lineage originates does not require FOXN1. Here, we have used a revertible severely hypomorphic allele of Foxn1, Foxn1R, to test the stability of the common TEPC in vivo. By reactivating Foxn1 expression postnatally in Foxn1R/- mice we demonstrate that functional TEPCs can persist in the thymic rudiment until at least 6 months of age, and retain the potential to give rise to both cortical and medullary thymic epithelial cells (cTECs and mTECs). These data demonstrate that the TEPC-state is remarkably stable in vivo under conditions of low Foxn1 expression, suggesting that manipulation of FOXN1 activity may prove a valuable method for long term maintenance of TEPC in vitro.


Subject(s)
Epithelial Cells/cytology , Forkhead Transcription Factors/metabolism , Stem Cells/metabolism , Thymus Gland/cytology , Alleles , Animals , Female , Forkhead Transcription Factors/genetics , Genotype , Immunohistochemistry , Keratins/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Pregnancy Proteins/metabolism , Stem Cells/cytology , Thymus Gland/metabolism , Thymus Gland/pathology
7.
Development ; 141(8): 1627-37, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24715454

ABSTRACT

Thymic involution is central to the decline in immune system function that occurs with age. By regenerating the thymus, it may therefore be possible to improve the ability of the aged immune system to respond to novel antigens. Recently, diminished expression of the thymic epithelial cell (TEC)-specific transcription factor Forkhead box N1 (FOXN1) has been implicated as a component of the mechanism regulating age-related involution. The effects of upregulating FOXN1 function in the aged thymus are, however, unknown. Here, we show that forced, TEC-specific upregulation of FOXN1 in the fully involuted thymus of aged mice results in robust thymus regeneration characterized by increased thymopoiesis and increased naive T cell output. We demonstrate that the regenerated organ closely resembles the juvenile thymus in terms of architecture and gene expression profile, and further show that this FOXN1-mediated regeneration stems from an enlarged TEC compartment, rebuilt from progenitor TECs. Collectively, our data establish that upregulation of a single transcription factor can substantially reverse age-related thymic involution, identifying FOXN1 as a specific target for improving thymus function and, thus, immune competence in patients. More widely, they demonstrate that organ regeneration in an aged mammal can be directed by manipulation of a single transcription factor, providing a provocative paradigm that may be of broad impact for regenerative biology.


Subject(s)
Aging/physiology , Forkhead Transcription Factors/metabolism , Regeneration/physiology , Thymus Gland/physiology , Animals , Cell Proliferation , Cellular Microenvironment , Epithelial Cells/metabolism , Lymphocyte Count , Mice , Mice, Transgenic , Models, Animal , Phenotype , Stem Cells/cytology , Stem Cells/metabolism , T-Lymphocytes/cytology , Thymus Gland/cytology , Up-Regulation
8.
Pigment Cell Melanoma Res ; 27(4): 580-9, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24628886

ABSTRACT

The ciliary body and iris are pigmented epithelial structures in the anterior eye segment that function to maintain correct intra-ocular pressure and regulate exposure of the internal eye structures to light, respectively. The cellular and molecular factors that mediate the development of the ciliary body and iris from the ocular pigmented epithelium remain to be fully elucidated. Here, we have investigated the role of Notch signaling during the development of the anterior pigmented epithelium by using genetic loss- and gain-of-function approaches. Loss of canonical Notch signaling results in normal iris development but absence of the ciliary body. This causes progressive hypotony and over time leads to phthisis bulbi, a condition characterized by shrinkage of the eye and loss of structure/function. Conversely, Notch gain-of-function results in aniridia and profound ciliary body hyperplasia, which causes ocular hypertension and glaucoma-like disease. Collectively, these data indicate that Notch signaling promotes ciliary body development at the expense of iris formation and reveals novel animal models of human ocular pathologies.


Subject(s)
Ciliary Body/embryology , Eye Proteins/metabolism , Iris/embryology , Pigment Epithelium of Eye/embryology , Receptors, Notch/metabolism , Signal Transduction/physiology , Animals , Ciliary Body/cytology , Eye Proteins/genetics , Humans , Iris/cytology , Mice , Mice, Transgenic , Pigment Epithelium of Eye/cytology , Receptors, Notch/genetics
9.
Cancer Cell ; 22(4): 479-93, 2012 Oct 16.
Article in English | MEDLINE | ID: mdl-23079658

ABSTRACT

Inflammation can promote or inhibit cancer progression. In this study we have addressed the role of the proinflammatory cytokine thymic stromal lymphopoietin (TSLP) during skin carcinogenesis. Using conditional loss- and gain-of-function mouse models for Notch and Wnt signaling, respectively, we demonstrate that TSLP-mediated inflammation protects against cutaneous carcinogenesis by acting directly on CD4 and CD8 T cells. Genetic ablation of TSLP receptor (TSLPR) perturbs T-cell-mediated protection and results in the accumulation of CD11b(+)Gr1(+) myeloid cells. These promote tumor growth by secreting Wnt ligands and augmenting ß-catenin signaling in the neighboring epithelium. Epithelial specific ablation of ß-catenin prevents both carcinogenesis and the accumulation of CD11b(+)Gr1(+) myeloid cells, suggesting tumor cells initiate a feed-forward loop that induces protumorigenic inflammation.


Subject(s)
Cytokines/physiology , Skin Neoplasms/etiology , Skin/immunology , Animals , CD11b Antigen/analysis , CD4-Positive T-Lymphocytes/physiology , Hematopoietic System/cytology , Immunoglobulins/physiology , Inflammation/complications , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Myeloid Cells/physiology , Receptors, Cytokine/physiology , Receptors, Notch/physiology , Skin Neoplasms/prevention & control , Wnt Signaling Pathway , beta Catenin/physiology , Thymic Stromal Lymphopoietin
10.
PLoS Genet ; 7(11): e1002348, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22072979

ABSTRACT

The forkhead transcription factor Foxn1 is indispensable for thymus development, but the mechanisms by which it mediates thymic epithelial cell (TEC) development are poorly understood. To examine the cellular and molecular basis of Foxn1 function, we generated a novel and revertible hypomorphic allele of Foxn1. By varying levels of its expression, we identified a number of features of the Foxn1 system. Here we show that Foxn1 is a powerful regulator of TEC differentiation that is required at multiple intermediate stages of TE lineage development in the fetal and adult thymus. We find no evidence for a role for Foxn1 in TEC fate-choice. Rather, we show it is required for stable entry into both the cortical and medullary TEC differentiation programmes and subsequently is needed at increasing dosage for progression through successive differentiation states in both cortical and medullary TEC. We further demonstrate regulation by Foxn1 of a suite of genes with diverse roles in thymus development and/or function, suggesting it acts as a master regulator of the core thymic epithelial programme rather than regulating a particular aspect of TEC biology. Overall, our data establish a genetics-based model of cellular hierarchies in the TE lineage and provide mechanistic insight relating titration of a single transcription factor to control of lineage progression. Our novel revertible hypomorph system may be similarly applied to analyzing other regulators of development.


Subject(s)
Adrenal Medulla/cytology , Cell Differentiation/genetics , Cell Lineage/genetics , Embryonic Development/genetics , Forkhead Transcription Factors/metabolism , Thymus Gland/growth & development , Adrenal Medulla/metabolism , Alleles , Animals , Epithelial Cells/metabolism , Epithelial Cells/physiology , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Developmental , Integrases/chemistry , Integrases/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Tamoxifen/chemistry
11.
Proc Natl Acad Sci U S A ; 105(3): 961-6, 2008 Jan 22.
Article in English | MEDLINE | ID: mdl-18195351

ABSTRACT

The thymus is essential for a functional immune system, because the thymic stroma uniquely supports T lymphocyte development. We have previously identified the epithelial progenitor population from which the thymus arises and demonstrated its ability to generate an organized functional thymus upon transplantation. These thymic epithelial progenitor cells (TEPC) are defined by surface determinants recognized by the mAbs MTS20 and MTS24, which were also recently shown to identify keratinocyte progenitor cells in the skin. However, the biochemical nature of the MTS20 and MTS24 determinants has remained unknown. Here we show, via expression profiling of fetal mouse TEPC and their differentiated progeny and subsequent analyses, that both MTS20 and MTS24 specifically bind an orphan protein of unknown function, Placenta-expressed transcript (Plet)-1. In the postgastrulation embryo, Plet-1 expression is highly restricted to the developing pharyngeal endoderm and mesonephros until day 11.5 of embryogenesis, consistent with the MTS20 and MTS24 staining pattern; both MTS20 and MTS24 specifically bind cell lines transfected with Plet-1; and antibodies to Plet-1 recapitulate MTS20/24 staining. In adult tissues, we demonstrate expression in a number of sites, including mammary and prostate epithelia and in the pancreas, where Plet-1 is specifically expressed by the major duct epithelium, providing a specific cell surface marker for this putative reservoir of pancreatic progenitor/stem cells. Plet-1 will thus provide an invaluable tool for genetic analysis of the lineage relationships and molecular mechanisms operating in the development, homeostasis, and injury in several organ/tissue systems.


Subject(s)
Epithelial Cells/metabolism , Pregnancy Proteins/metabolism , Stem Cells/immunology , Stem Cells/metabolism , Thymus Gland/embryology , Thymus Gland/metabolism , Animals , Antigens, Surface/genetics , Antigens, Surface/immunology , Biomarkers , Cell Line , Embryo, Mammalian/embryology , Embryo, Mammalian/immunology , Embryo, Mammalian/metabolism , Epithelial Cells/immunology , Epithelium/metabolism , Gene Expression Regulation , Gene Expression Regulation, Developmental , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Pancreatic Ducts/metabolism , Pregnancy Proteins/genetics , Pregnancy Proteins/immunology , RNA, Messenger/genetics , Thymus Gland/immunology , Time Factors
12.
Methods Mol Biol ; 380: 125-62, 2007.
Article in English | MEDLINE | ID: mdl-17876091

ABSTRACT

T-cell development occurs principally in the thymus. Here, immature progenitor cells are guided through the differentiation and selection steps required to generate a complex T-cell repertoire that is both self-tolerant and has propensity to bind self major histocompatibility complex. These processes depend on an array of functionally distinct epithelial cell types within the thymic stroma, which have a common developmental origin in the pharyngeal endoderm. Here, we describe the structural and phenotypic attributes of the thymic stroma, and review current cellular and molecular understanding of thymus organogenesis.


Subject(s)
Organogenesis/immunology , Thymus Gland/embryology , Animals , Humans , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Thymus Extracts , Thymus Gland/anatomy & histology , Thymus Gland/cytology , Thymus Gland/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...