Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
Add more filters

Publication year range
1.
PLoS Biol ; 21(10): e3002336, 2023 10.
Article in English | MEDLINE | ID: mdl-37856539

ABSTRACT

The transparent corneal epithelium in the eye is maintained through the homeostasis regulated by limbal stem cells (LSCs), while the nontransparent epidermis relies on epidermal keratinocytes for renewal. Despite their cellular similarities, the precise cell fates of these two types of epithelial stem cells, which give rise to functionally distinct epithelia, remain unknown. We performed a multi-omics analysis of human LSCs from the cornea and keratinocytes from the epidermis and characterized their molecular signatures, highlighting their similarities and differences. Through gene regulatory network analyses, we identified shared and cell type-specific transcription factors (TFs) that define specific cell fates and established their regulatory hierarchy. Single-cell RNA-seq (scRNA-seq) analyses of the cornea and the epidermis confirmed these shared and cell type-specific TFs. Notably, the shared and LSC-specific TFs can cooperatively target genes associated with corneal opacity. Importantly, we discovered that FOSL2, a direct PAX6 target gene, is a novel candidate associated with corneal opacity, and it regulates genes implicated in corneal diseases. By characterizing molecular signatures, our study unveils the regulatory circuitry governing the LSC fate and its association with corneal opacity.


Subject(s)
Corneal Opacity , Epithelium, Corneal , Limbus Corneae , Humans , Limbus Corneae/metabolism , Cornea/metabolism , Epithelium, Corneal/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Cell Differentiation/genetics , Corneal Opacity/metabolism
2.
Stem Cells ; 41(12): 1133-1141, 2023 Dec 14.
Article in English | MEDLINE | ID: mdl-37632794

ABSTRACT

Congenital aniridia is caused by heterozygous mutations on the PAX6 gene leading to reduced amount of PAX6 protein (haploinsufficiency), abnormal eye development, and aniridia-associated keratopathy (AAK). This progressive corneal opacification resembles late-onset limbal stem cell (LSC) deficiency, leading to disrupted corneal epithelial renewal. The factors leading to AAK are not known and defects in native LSC differentiation and/or features leading to ocular surface dysfunction like inflammation and loss of innervation could contribute to development of AAK. Here, we produced induced pluripotent stem cells (hiPSC) from 3 AAK patients and examined whether PAX6 haploinsufficiency affects LSC lineage commitment. During LSC differentiation, characterization of the AAK lines showed lowered PAX6 expression as compared to wild type (WT) controls and expression peak of PAX6 during early phase of differentiation was detected only in the WT hiPSC lines. Whether it reflects developmental regulation remains to be studied further. Nevertheless, the AAK-hiPSCs successfully differentiated toward LSC lineage, in line with the presence of LSCs in young patients before cell loss later in life. In addition, patient-specific LSCs showed similar wound healing capacity as WT cells. However, extensive batch-related variation in the LSC marker expression and wound healing efficacy was detected without clear correlation to AAK. As development and maintenance of corneal epithelium involves an interplay between LSCs and their environment, the AAK-hiPSCs generated here can be further used to study the crosstalk between LSCs and limbal niche including, eg, corneal immune cells, stroma cells, and neurons.


Subject(s)
Aniridia , Corneal Diseases , Epithelium, Corneal , Induced Pluripotent Stem Cells , Limbus Corneae , Humans , Cornea , Epithelium, Corneal/metabolism , Corneal Diseases/genetics , PAX6 Transcription Factor/genetics , PAX6 Transcription Factor/metabolism , Aniridia/genetics
3.
Proc Natl Acad Sci U S A ; 116(35): 17361-17370, 2019 08 27.
Article in English | MEDLINE | ID: mdl-31413199

ABSTRACT

Mutations in transcription factor p63 are associated with developmental disorders that manifest defects in stratified epithelia including the epidermis. The underlying cellular and molecular mechanism is however not yet understood. We established an epidermal commitment model using human induced pluripotent stem cells (iPSCs) and characterized differentiation defects of iPSCs derived from ectrodactyly, ectodermal dysplasia, and cleft lip/palate (EEC) syndrome patients carrying p63 mutations. Transcriptome analyses revealed stepwise cell fate transitions during epidermal commitment: Specification from multipotent simple epithelium to basal stratified epithelia and ultimately to the mature epidermal fate. Differentiation defects of EEC iPSCs caused by p63 mutations occurred during the specification switch from the simple epithelium to the basal-stratified epithelial fate. Single-cell transcriptome and pseudotime analyses of cell states identified mesodermal activation that was associated with the deviated commitment route of EEC iPSCs. Integrated analyses of differentially regulated genes and p63-dependent dynamic genomic enhancers during epidermal commitment suggest that p63 directly controls epidermal gene activation at the specification switch and has an indirect effect on mesodermal gene repression. Importantly, inhibitors of mesodermal induction enhanced epidermal commitment of EEC iPSCs. Our findings demonstrate that p63 is required for specification of stratified epithelia, and that epidermal commitment defects caused by p63 mutations can be reversed by repressing mesodermal induction. This study provides insights into disease mechanisms underlying stratified epithelial defects caused by p63 mutations and suggests potential therapeutic strategies for the disease.


Subject(s)
Cleft Lip/genetics , Cleft Palate/genetics , Ectodermal Dysplasia/genetics , Epithelium/metabolism , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Epidermis/embryology , Epidermis/metabolism , Epithelium/embryology , Gene Expression , Gene Expression Profiling , Humans , Immunohistochemistry , Induced Pluripotent Stem Cells/metabolism , Keratinocytes/metabolism , Mutation , Sequence Analysis, RNA , Single-Cell Analysis , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism
4.
Biochem Biophys Res Commun ; 582: 100-104, 2021 12 10.
Article in English | MEDLINE | ID: mdl-34700241

ABSTRACT

Aniridia is a panocular inherited rare eye disease linked to heterozygous mutations on the PAX6 gene, which fail to properly produce sufficient protein essential for normal eye development and function. Most of the patients suffer from aniridia-related keratopathy, a progressive opacification of the cornea. There is no effective treatment for this blinding disease. Here we screen for small compounds and identified Ritanserin, a serotonin 2A receptor antagonist, that can rescue PAX6 haploinsufficiency of mutant limbal cells, defective cell migration and PAX6-target gene expression. We further demonstrated that Ritanserin activates PAX6 production through the selective inactivation of the MEK/ERK signaling pathway. Our data strongly suggest that repurposing this therapeutic molecule could be effective in preventing or treating existing blindness by restoring corneal transparency.


Subject(s)
Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Ophthalmic Solutions/pharmacology , PAX6 Transcription Factor/genetics , Ritanserin/pharmacology , Serotonin Antagonists/pharmacology , Stem Cells/drug effects , Aniridia/drug therapy , Aniridia/genetics , Aniridia/metabolism , Aniridia/pathology , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Repositioning/methods , Epithelium, Corneal/drug effects , Epithelium, Corneal/metabolism , Epithelium, Corneal/pathology , Gene Expression Regulation , HEK293 Cells , Haploinsufficiency , Humans , Limbus Corneae/drug effects , Limbus Corneae/metabolism , Limbus Corneae/pathology , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Models, Biological , PAX6 Transcription Factor/agonists , PAX6 Transcription Factor/metabolism , Receptor, Serotonin, 5-HT2A/genetics , Receptor, Serotonin, 5-HT2A/metabolism , Signal Transduction/drug effects , Stem Cells/metabolism , Stem Cells/pathology
5.
Stem Cells ; 37(9): 1166-1175, 2019 09.
Article in English | MEDLINE | ID: mdl-31237401

ABSTRACT

Dermal papilla cells (DPCs) play a pivotal role in the regulation of hair follicle (HF) growth, formation, and cycling, mainly through paracrine mechanisms. In the last decade, extracellular vesicles (EVs) have been recognized as a new paracrine mechanism that can modify the physiological state of recipient cells by transferring biological material. Herein, we investigated the effect of EVs isolated from stimulated human dermal fibroblasts (DFs) on DPC activation and HF growth. We found that these EVs (st-EVs) enhanced HF growth ex vivo. Comparative transcriptomic analysis on DPCs identified specific activation of the NDP gene, encoding the non-Wnt ligand Norrin. We found that Norrin was secreted by st-EVs-stimulated DPCs activating in a noncell autonomous manner ß-catenin pathway in follicular keratinocytes (human HF keratinocyte [HHFK]) and hair growth ex vivo. Although Norrin-specific receptor Frizzled4 was barely detected in HHFK, we found its presence in DF-EVs. Accordingly, DF-EVs provided Frizzled4 to potentiate Norrin effects ex vivo. Our study identifies DF-EVs as efficient activators of DPCs and Norrin as a novel modulatory player in HF physiopathology. Stem Cells 2019;37:1166-1175.


Subject(s)
Cell Proliferation/genetics , Dermis/metabolism , Extracellular Vesicles/metabolism , Eye Proteins/genetics , Fibroblasts/metabolism , Hair Follicle/metabolism , Nerve Tissue Proteins/genetics , Cell Line , Cells, Cultured , Dermis/cytology , Eye Proteins/metabolism , Fibroblasts/cytology , Gene Expression Profiling/methods , Gene Expression Regulation , Hair Follicle/cytology , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Nerve Tissue Proteins/metabolism
6.
Stem Cells ; 37(3): 417-429, 2019 03.
Article in English | MEDLINE | ID: mdl-30548157

ABSTRACT

Mutations in key transcription factors SOX2 and P63 were linked with developmental defects and postnatal abnormalities such as corneal opacification, neovascularization, and blindness. The latter phenotypes suggest that SOX2 and P63 may be involved in corneal epithelial regeneration. Although P63 has been shown to be a key regulator of limbal stem cells, the expression pattern and function of SOX2 in the adult cornea remained unclear. Here, we show that SOX2 regulates P63 to control corneal epithelial stem/progenitor cell function. SOX2 and P63 were co-expressed in the stem/progenitor cell compartments of the murine cornea in vivo and in undifferentiated human limbal epithelial stem/progenitor cells in vitro. In line, a new consensus site that allows SOX2-mediated regulation of P63 enhancer was identified while repression of SOX2 reduced P63 expression, suggesting that SOX2 is upstream to P63. Importantly, knockdown of SOX2 significantly attenuated cell proliferation, long-term colony-forming potential of stem/progenitor cells, and induced robust cell differentiation. However, this effect was reverted by forced expression of P63, suggesting that SOX2 acts, at least in part, through P63. Finally, miR-450b was identified as a direct repressor of SOX2 that was required for SOX2/P63 downregulation and cell differentiation. Altogether, we propose that SOX2/P63 pathway is an essential regulator of corneal stem/progenitor cells while mutations in SOX2 or P63 may disrupt epithelial regeneration, leading to loss of corneal transparency and blindness. Stem Cells 2019;37:417-429.


Subject(s)
Cell Differentiation , Cell Proliferation , Epithelium, Corneal/metabolism , SOXB1 Transcription Factors/metabolism , Signal Transduction , Stem Cells/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Alkaloids , Animals , Mice , NIH 3T3 Cells , Piperidines , SOXB1 Transcription Factors/genetics , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics
7.
Mol Cell ; 46(5): 662-73, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22681888

ABSTRACT

Embryonic stem cells (ESCs) maintain high genomic plasticity, which is essential for their capacity to enter diverse differentiation pathways. Posttranscriptional modifications of chromatin histones play a pivotal role in maintaining this plasticity. We now report that one such modification, monoubiquitylation of histone H2B on lysine 120 (H2Bub1), catalyzed by the E3 ligase RNF20, increases during ESC differentiation and is required for efficient execution of this process. This increase is particularly important for the transcriptional induction of relatively long genes during ESC differentiation. Furthermore, we identify the deubiquitinase USP44 as a negative regulator of H2B ubiquitylation, whose downregulation during ESC differentiation contributes to the increase in H2Bub1. Our findings suggest that optimal ESC differentiation requires dynamic changes in H2B ubiquitylation patterns, which must occur in a timely and well-coordinated manner.


Subject(s)
Cell Differentiation/genetics , Embryonic Stem Cells/cytology , Endopeptidases/physiology , Histones/metabolism , Ubiquitin-Protein Ligases/physiology , Animals , Chromatin Assembly and Disassembly , Down-Regulation , Embryonic Stem Cells/metabolism , Endopeptidases/metabolism , Epigenesis, Genetic , Humans , Mice , Models, Genetic , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Specific Proteases , Ubiquitination
8.
Stem Cells ; 36(9): 1421-1429, 2018 09.
Article in English | MEDLINE | ID: mdl-29808941

ABSTRACT

Heterozygous PAX6 gene mutations leading to haploinsufficiency are the main cause of congenital aniridia, a rare and progressive panocular disease characterized by reduced visual acuity. Up to 90% of patients suffer from aniridia-related keratopathy (ARK), caused by a combination of factors including limbal epithelial stem cell (LSC) deficiency, impaired healing response and abnormal differentiation of the corneal epithelium. It usually begins in the first decade of life, resulting in recurrent corneal erosions, sub-epithelial fibrosis, and corneal opacification. Unfortunately, there are currently no efficient treatments available for these patients and no in vitro model for this pathology. We used CRISPR/Cas9 technology to introduce into the PAX6 gene of LSCs a heterozygous nonsense mutation found in ARK patients. Nine clones carrying a p.E109X mutation on one allele were obtained with no off-target mutations. Compared with the parental LSCs, heterozygous mutant LSCs displayed reduced expression of PAX6 and marked slow-down of cell proliferation, migration and detachment. Moreover, addition to the culture medium of recombinant PAX6 protein fused to a cell penetrating peptide was able to activate the endogenous PAX6 gene and to rescue phenotypic defects of mutant LSCs, suggesting that administration of such recombinant PAX6 protein could be a promising therapeutic approach for aniridia-related keratopathy. More generally, our results demonstrate that introduction of disease mutations into LSCs by CRISPR/Cas9 genome editing allows the creation of relevant cellular models of ocular disease that should greatly facilitate screening of novel therapeutic approaches. Stem Cells 2018;36:1421-1429.


Subject(s)
Aniridia/genetics , CRISPR-Cas Systems/physiology , Epithelium, Corneal/metabolism , Gene Editing/methods , PAX6 Transcription Factor/genetics , Aniridia/pathology , Humans
9.
Nucleic Acids Res ; 43(2): 862-74, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25567987

ABSTRACT

p63 is a crucial regulator of epidermal development, but its transcriptional control has remained elusive. Here, we report the identification of a long-range enhancer (p63LRE) that is composed of two evolutionary conserved modules (C38 and C40), acting in concert to control tissue- and layer-specific expression of the p63 gene. Both modules are in an open and active chromatin state in human and mouse keratinocytes and in embryonic epidermis, and are strongly bound by p63. p63LRE activity is dependent on p63 expression in embryonic skin, and also in the commitment of human induced pluripotent stem cells toward an epithelial cell fate. A search for other transcription factors involved in p63LRE regulation revealed that the CAAT enhancer binding proteins Cebpa and Cebpb and the POU domain-containing protein Pou3f1 repress p63 expression during keratinocyte differentiation by binding the p63LRE enhancer. Collectively, our data indicate that p63LRE is composed of additive and partly redundant enhancer modules that act to direct robust p63 expression selectively in the basal layer of the epidermis.


Subject(s)
Enhancer Elements, Genetic , Epidermis/embryology , Epidermis/metabolism , Gene Expression Regulation, Developmental , Keratinocytes/metabolism , Phosphoproteins/genetics , Trans-Activators/genetics , Animals , Cell Differentiation/genetics , Cells, Cultured , Humans , Keratinocytes/cytology , Mice, Inbred C57BL , Morphogenesis/genetics , Transcription Factors/metabolism , Transcriptional Activation
10.
Stem Cells ; 33(8): 2374-80, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25873344

ABSTRACT

Recent studies suggest that the metabolic network is an important part of the molecular circuitry that underlies pluripotency. Of the metabolic pathways that were implicated in the pluripotency balance, "energy" metabolism is particularly notable. Its mechanism of action on pluripotency-regulating genes has been partially elucidated when three metabolites, namely acetate, S-adenosylmethionine, and O-linked ß-N-acetylglucosamine were recently shown to link cytosolic signals to pluripotent gene expression. The cytosolic levels of these metabolites are the result of environmental perturbations, making them sensitive messengers, which are assumed to diffuse through the nuclear pores, being small molecules. Recent work also suggests that the modulation of the levels of these metabolites in pluripotent cells controls the balance between pluripotency and early commitment via epigenetic modifications. Here, we review recent studies that link metabolism and pluripotency via epigenetic modifications that occur through these three metabolites.


Subject(s)
Energy Metabolism/physiology , Epigenesis, Genetic/physiology , Pluripotent Stem Cells/metabolism , Acetates/metabolism , Acetylglucosamine/metabolism , Animals , Humans , Pluripotent Stem Cells/cytology , S-Adenosylmethionine/metabolism
11.
Proc Natl Acad Sci U S A ; 110(6): 2152-6, 2013 Feb 05.
Article in English | MEDLINE | ID: mdl-23355677

ABSTRACT

Ectodermal dysplasia is a group of congenital syndromes affecting a variety of ectodermal derivatives. Among them, ectrodactyly, ectodermal dysplasia, and cleft lip/palate (EEC) syndrome is caused by single point mutations in the p63 gene, which controls epidermal development and homeostasis. Phenotypic defects of the EEC syndrome include skin defects and limbal stem-cell deficiency. In this study, we designed a unique cellular model that recapitulated major embryonic defects related to EEC. Fibroblasts from healthy donors and EEC patients carrying two different point mutations in the DNA binding domain of p63 were reprogrammed into induced pluripotent stem cell (iPSC) lines. EEC-iPSC from both patients showed early ectodermal commitment into K18(+) cells but failed to further differentiate into K14(+) cells (epidermis/limbus) or K3/K12(+) cells (corneal epithelium). APR-246 (PRIMA-1(MET)), a small compound that restores functionality of mutant p53 in human tumor cells, could revert corneal epithelial lineage commitment and reinstate a normal p63-related signaling pathway. This study illustrates the relevance of iPSC for p63 related disorders and paves the way for future therapy of EEC.


Subject(s)
Cleft Lip/drug therapy , Cleft Lip/pathology , Cleft Palate/drug therapy , Cleft Palate/pathology , Ectodermal Dysplasia/drug therapy , Ectodermal Dysplasia/pathology , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/pathology , Quinuclidines/pharmacology , Binding Sites/genetics , Cell Differentiation/drug effects , Cell Line , Cleft Lip/genetics , Cleft Lip/metabolism , Cleft Palate/genetics , Cleft Palate/metabolism , Ectodermal Dysplasia/genetics , Ectodermal Dysplasia/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelium, Corneal/pathology , Humans , Induced Pluripotent Stem Cells/metabolism , Models, Biological , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Point Mutation , Signal Transduction/drug effects , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
12.
Stem Cells ; 32(12): 3137-49, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25186014

ABSTRACT

Human embryonic stem cells (hESCs) represent a potential source of transplantable cells for regenerative medicine, but development of teratoma even in syngenic recipients represents a critical obstacle to safe stem cell-based therapies. We hypothesized that hESCs escape the immune surveillance by regulating the environmental immune system. Using cocultures of hESCs with allogenic peripheral blood mononuclear cells, we demonstrated that hESCs prevent proliferation and activation of human CD4+ T lymphocytes, an effect dependent upon monocytes. Altered expression of key signaling molecules responsible for the crosstalk of monocytes with T cells was detected in the presence of hESCs. Analyzing the mechanism of action, we demonstrated that hESCs were able to downregulate intracellular glutathione levels in both monocytes and CD4+ cells by suppressing glutamate cysteine ligase expression and to alter MHCII and CD80 expression in monocytes. These effects were achieved at least partially via TGF-beta signaling, and both monocyte phenotype and GCLC expression were affected by Caspase-3 proteolytic activity. Altogether, our results demonstrate a novel immune-suppressive mechanism used by hESCs.


Subject(s)
Cell Differentiation/physiology , Dendritic Cells/cytology , Human Embryonic Stem Cells/metabolism , Lymphocyte Activation/immunology , Signal Transduction , T-Lymphocytes/metabolism , Transforming Growth Factor beta/metabolism , Antigens, CD/immunology , Cell Differentiation/immunology , Coculture Techniques/methods , Humans , Monocytes/cytology , Signal Transduction/physiology , T-Lymphocytes/immunology , Transforming Growth Factor beta/immunology
13.
Exp Dermatol ; 24(8): 618-22, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25939713

ABSTRACT

Alopecia-neurological defects-endocrinopathy (ANE) syndrome is a rare inherited hair disorder, which was shown to result from decreased expression of the RNA-binding motif protein 28 (RBM28). In this study, we attempted to delineate the role of RBM28 in hair biology. First, we sought to obtain evidence for the direct involvement of RBM28 in hair growth. When RBM28 was downregulated in human hair follicle (HF) organ cultures, we observed catagen induction and HF growth arrest, indicating that RBM28 is necessary for normal hair growth. We also aimed at identifying molecular targets of RBM28. Given that an RBM28 homologue was recently found to regulate miRNA biogenesis in C. elegans and given the known pivotal importance of miRNAs for proper hair follicle development, we studied global miRNA expression profile in cells knocked down for RBM28. This analysis revealed that RBM28 controls the expression of miR-203. miR-203 was found to regulate in turn TP63, encoding the transcription factor p63, which is critical for hair morphogenesis. In conclusion, RBM28 contributes to HF growth regulation through modulation of miR-203 and p63 activity.


Subject(s)
Alopecia/metabolism , Endocrine System Diseases/metabolism , Gene Expression Regulation , Hair Follicle/metabolism , Intellectual Disability/metabolism , MicroRNAs/physiology , RNA-Binding Proteins/physiology , Transcription Factors/physiology , Tumor Suppressor Proteins/physiology , Alopecia/physiopathology , Cells, Cultured , Endocrine System Diseases/physiopathology , Genes, Reporter , Hair/growth & development , Hair Follicle/growth & development , Humans , Intellectual Disability/physiopathology , Keratinocytes/metabolism , Morphogenesis , Organ Culture Techniques , RNA Interference , RNA, Small Interfering/genetics , RNA-Binding Proteins/genetics , Transfection , Up-Regulation
14.
J Cell Sci ; 125(Pt 19): 4640-50, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22767507

ABSTRACT

The mechanisms underlying the immunomodulatory effects of mesenchymal stem cells (MSCs) have been investigated under extreme conditions of strong T cell activation, which induces the rapid death of activated lymphocytes. The objective of this study was to investigate these mechanisms in the absence of additional polyclonal activation. In co-cultures of peripheral mononuclear blood cells with human MSCs (hereafter referred to as hMSCs), we observed a striking decrease in the level of CD8 expression on CD8+ cells, together with decreased expression of CD28 and CD44, and impaired production of IFN-gamma and Granzyme B. This effect was specific to hMSCs, because it was not observed with several other cell lines. Downregulation of CD8 expression required CD14+ monocytes to be in direct contact with the CD8+ cells, whereas the effects of hMSCs on the CD14+ cells were essentially mediated by soluble factors. The CD14+ monocytes exhibited a tolerogenic pattern when co-cultured with hMSCs, with a clear decrease in CD80 and CD86 co-stimulatory molecules, and an increase in the inhibitory receptors ILT-3 and ILT-4. CD8+ cells that were preconditioned by MSCs had similar effects on monocytes and were able to inhibit lymphocyte proliferation. Injection of hMSCs in humanized NSG mice showed similar trends, in particular decreased levels of CD44 and CD28 in human immune cells. Our study demonstrates a new immunomodulation mechanism of action of hMSCs through the modulation of CD8+ cells towards a non-cytotoxic and/or suppressive phenotype. This mechanism of action has to be taken into account in clinical trials, where it should be beneficial in grafts and autoimmune diseases, but potentially detrimental in malignant diseases.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Immune Tolerance/immunology , Mesenchymal Stem Cells/cytology , Monocytes/cytology , Monocytes/immunology , Adult , Animals , Biomarkers/metabolism , CD8 Antigens/metabolism , Cell Adhesion Molecules/metabolism , Down-Regulation , Female , Humans , Immunization , Lipopolysaccharide Receptors/metabolism , Lymphocyte Activation/genetics , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Phenotype , Transcription, Genetic
15.
Proc Natl Acad Sci U S A ; 108(52): 21099-104, 2011 Dec 27.
Article in English | MEDLINE | ID: mdl-22160706

ABSTRACT

The p53 family member TAp73 is a transcription factor that plays a key role in many biological processes, including neuronal development. In particular, we have shown that p73 drives the expression of miR-34a, but not miR-34b and c, in mouse cortical neurons. miR-34a in turn modulates the expression of synaptic targets including synaptotagmin-1 and syntaxin-1A. Here we show that this axis is retained in mouse ES cells committed to differentiate toward a neurological phenotype. Moreover, overexpression of miR-34a alters hippocampal spinal morphology, and results in electrophysiological changes consistent with a reduction in spinal function. Therefore, the TAp73/miR-34a axis has functional relevance in primary neurons. These data reinforce a role for miR-34a in neuronal development.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/cytology , MicroRNAs/metabolism , Neurites/physiology , Nuclear Proteins/metabolism , Spine/cytology , Animals , Blotting, Western , Cell Differentiation/genetics , Electrophysiology , Embryonic Stem Cells/metabolism , Mice , Mice, Knockout , Nuclear Proteins/genetics , Real-Time Polymerase Chain Reaction , Spine/physiology , Synaptotagmin I/metabolism , Syntaxin 1/metabolism
16.
Ocul Surf ; 34: 225-234, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39127390

ABSTRACT

BACKGROUND-AIM: PAX6 is a key regulator of eye development and epithelial homeostasis in the cornea. When deficient, chronic corneal inflammation, neovascularization and limbal stem cell deficiency can occur. Here we investigated the potential of duloxetine, a generic serotonin reuptake inhibitor that can upregulate PAX6 in vitro, for its in vivo activity in the context of corneal inflammation. METHODS: Duloxetine tolerance was tested in a human limbal stem cell line and isogenic CRISPR-knockout PAX6+/- cells. C57BL/6-Wildtype mice were administered duloxetine eye drops at concentrations of 1 µM - 2 mM and tested for toxicity and corneal PAX6 expression. In LPS-induced corneal inflammation in mice, duloxetine's effect on PAX6 expression, corneal opacification and inflammatory responses were evaluated by in vivo corneal imaging, immunostaining, and whole-transcriptome microarray analysis. RESULTS: No toxicity was observed in vitro for duloxetine concentrations up to 10µΜ. In vivo, duloxetine drops were well-tolerated up to 50 µM. Duloxetine drops at 10µΜ significantly upregulated PAX6 protein levels in the cornea by 30 % within 2 days. In the LPS model, duloxetine resulted in a sustained 33 % PAX6 protein upregulation in the cornea at 7 days, and in reduced opacity within 2 days, accompanied by a significant dampening of IL-17A signaling, neutrophil degranulation, microglial activation, macrophage markers, and MMP expression, despite non-significant changes in total inflammatory cell infiltration. CONCLUSION: Short-term administration of a repurposed generic drug, duloxetine, upregulates PAX6 protein levels in the cornea of mice and exerts an anti-inflammatory activity by dampening innate immune responses.

17.
Cell Death Dis ; 15(2): 174, 2024 Feb 26.
Article in English | MEDLINE | ID: mdl-38409173

ABSTRACT

miR-184-knockout mice display perturbed epidermal stem cell differentiation. However, the potential role of miR-184 in skin pathology is unclear. Here, we report that miR-184 controls epidermal stem cell dynamics and that miR-184 ablation enhances skin carcinogenesis in mice. In agreement, repression of miR-184 in human squamous cell carcinoma (SCC) enhances neoplastic hallmarks of human SCC cells in vitro and tumor development in vivo. Characterization of miR-184-regulatory network, suggests that miR-184 inhibits pro-oncogenic pathways, cell proliferation, and epithelial to mesenchymal transformation. Of note, depletion of miR-184 enhances the levels of ß-catenin under homeostasis and following experimental skin carcinogenesis. Finally, the repression of ß-catenin by miR-184, inhibits the neoplastic phenotype of SCC cells. Taken together, miR-184 behaves as an epidermal tumor suppressor, and may provide a potentially useful target for skin SCC therapy.


Subject(s)
Carcinoma, Squamous Cell , MicroRNAs , Skin Neoplasms , Animals , Humans , Mice , beta Catenin/genetics , beta Catenin/metabolism , Carcinogenesis/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/pathology
18.
Stem Cells ; 30(5): 898-909, 2012 May.
Article in English | MEDLINE | ID: mdl-22367714

ABSTRACT

Approximately 6 million people worldwide are suffering from severe visual impairments or blindness due to corneal diseases. Corneal allogeneic transplantation is often required to restore vision; however, shortage in corneal grafts and immunorejections remain major challenges. The molecular basis of corneal diseases is poorly understood largely due to lack of appropriate cellular models. Here, we described a robust differentiation of human-induced pluripotent stem cells (hiPSCs) derived from hair follicles or skin fibroblasts into corneal epithelial-like cells. We found that BMP4, coupled with corneal fibroblast-derived conditioned medium and collagen IV allowed efficient corneal epithelial commitment of hiPSCs in a manner that recapitulated corneal epithelial lineage development with high purity. Organotypic reconstitution assays suggested the ability of these cells to stratify into a corneal-like epithelium. This model allowed us identifying miR-450b-5p as a molecular switch of Pax6, a major regulator of eye development. miR-450b-5p and Pax6 were reciprocally distributed at the presumptive epidermis and ocular surface, respectively. miR-450b-5p inhibited Pax6 expression and corneal epithelial fate in vitro, altogether, suggesting that by repressing Pax6, miR-450b-5p triggers epidermal specification of the ectoderm, while its absence allows ocular epithelial development. Additionally, miR-184 was detectable in early eye development and corneal epithelial differentiation of hiPSCs. The knockdown of miR-184 resulted in a decrease in Pax6 and K3, in line with recent findings showing that a point mutation in miR-184 leads to corneal dystrophy. Altogether, these data indicate that hiPSCs are valuable for modeling corneal development and may pave the way for future cell-based therapy.


Subject(s)
Cell Lineage/physiology , Cornea/embryology , Gene Expression Regulation, Developmental/physiology , MicroRNAs/biosynthesis , Models, Biological , Pluripotent Stem Cells/metabolism , Animals , Cell Differentiation/physiology , Cornea/cytology , Eye Proteins/biosynthesis , Eye Proteins/genetics , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Humans , Mice , MicroRNAs/genetics , PAX6 Transcription Factor , Paired Box Transcription Factors/biosynthesis , Paired Box Transcription Factors/genetics , Pluripotent Stem Cells/cytology , Repressor Proteins/biosynthesis , Repressor Proteins/genetics
19.
Trends Biochem Sci ; 33(12): 583-91, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18848452

ABSTRACT

The epidermis and its appendages provide organisms with protection from the environment, keeping pathogens out and preventing the loss of essential body fluids. To perform both functions, the skin has elaborated a complex differentiation process known as cornification. The renewal capacity of the skin, which is responsible for maintaining tissue homeostasis, regenerating hair and repairing the epidermis after injury, resides in the basal proliferating compartment in which epidermal stem cells are located. These cells possess the remarkable capacity to both self-perpetuate and give rise to the differentiating cells that form mature tissues. Recent findings indicate that microRNAs have an essential role in orchestrating the formation of epidermis and skin appendages, in particular, at the interface between stemness and differentiation.


Subject(s)
Cell Differentiation/genetics , MicroRNAs/genetics , Skin/cytology , Stem Cells/cytology , Animals , DEAD-box RNA Helicases/deficiency , Dermatitis, Atopic/physiopathology , Down-Regulation , Endoribonucleases/deficiency , Epidermal Cells , Gene Expression Regulation , Homeostasis , Humans , MicroRNAs/physiology , Psoriasis/physiopathology , Ribonuclease III , Skin/embryology , Skin/growth & development , Skin Neoplasms/physiopathology
20.
Cells ; 12(4)2023 02 14.
Article in English | MEDLINE | ID: mdl-36831285

ABSTRACT

Mitogen-activated protein kinase (MAPK) pathways represent ubiquitous cellular signal transduction pathways that regulate all aspects of life and are frequently altered in disease. Once activated through phosphorylation, these MAPKs in turn phosphorylate and activate transcription factors present either in the cytoplasm or in the nucleus, leading to the expression of target genes and, as a consequence, they elicit various biological responses. The aim of this work is to provide a comprehensive review focusing on the roles of MAPK signaling pathways in ocular pathophysiology and the potential to influence these for the treatment of eye diseases. We summarize the current knowledge of identified MAPK-targeting compounds in the context of ocular diseases such as macular degeneration, cataract, glaucoma and keratopathy, but also in rare ocular diseases where the cell differentiation, proliferation or migration are defective. Potential therapeutic interventions are also discussed. Additionally, we discuss challenges in overcoming the reported eye toxicity of some MAPK inhibitors.


Subject(s)
Mitogen-Activated Protein Kinases , p38 Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Signal Transduction , Phosphorylation , MAP Kinase Signaling System/physiology
SELECTION OF CITATIONS
SEARCH DETAIL