Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Bone ; 172: 116760, 2023 07.
Article in English | MEDLINE | ID: mdl-37028583

ABSTRACT

Technologies on the development and differentiation of human induced pluripotent stem cells (hiPSCs) are rapidly improving, and have been applied to create cell types relevant to the bone field. Differentiation protocols to form bona fide bone-forming cells from iPSCs are available, and can be used to probe details of differentiation and function in depth. When applied to iPSCs bearing disease-causing mutations, the pathogenetic mechanisms of diseases of the skeleton can be elucidated, along with the development of novel therapeutics. These cells can also be used for development of cell therapies for cell and tissue replacement.


Subject(s)
Induced Pluripotent Stem Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Cell Differentiation/genetics , Biology
2.
Front Physiol ; 14: 1316635, 2023.
Article in English | MEDLINE | ID: mdl-38274045

ABSTRACT

Mouse and human genetic studies indicate key roles of the Wnt10a ligand in odontogenesis. Previous studies have identified effectors and regulators of the Wnt signaling pathway actively expressed during key stages of tooth morphogenesis. However, limitations in multiplexing and spatial resolution hindered a more comprehensive analysis of these signaling molecules. Here, profiling of transcriptomes using fluorescent multiplex in situ hybridization and single-cell RNA-sequencing (scRNA-seq) provide robust insight into the synchronized expression patterns of Wnt10a, Dkk1, and Sost simultaneously during tooth development. First, we identified Wnt10a transcripts restricted to the epithelium at the stage of tooth bud morphogenesis, contrasting that of Sost and Dkk1 localization to the dental mesenchyme. By embryonic day 15.5 (E15.5), a marked shift of Wnt10a expression from dental epithelium to mesenchyme was noted, while Sost and Dkk1 expression remained enriched in the mesenchyme. By postnatal day 0 (P0), co-localization patterns of Wnt10a, Dkk1, and Sost were observed in both terminally differentiating and secreting odontoblasts of molars and incisors. Interestingly, Wnt10a exhibited robust expression in fully differentiated ameloblasts at the developing cusp tip of both molars and incisors, an observation not previously noted in prior studies. At P7 and 14, after the mineralization of dentin and enamel, Wnt10a expression was limited to odontoblasts. Meanwhile, Wnt modulators showed reduced or absent signals in molars. In contrast, strong signals persisted in ameloblasts (for Wnt10a) and odontoblasts (for Wnt10a, Sost, and Dkk1) towards the proximal end of incisors, near the cervical loop. Our scRNA-seq analysis used CellChat to further contextualize Wnt pathway-mediated communication between cells by examining ligand-receptor interactions among different clusters. The co-localization pattern of Wnt10a, Dkk1, and Sost in both terminally differentiating and secreting odontoblasts of molars and incisors potentially signifies the crucial ligand-modulator interaction along the gradient of cytodifferentiation starting from each cusp tip towards the apical region. These data provide cell type-specific insight into the role of Wnt ligands and mediators during epithelial-mesenchymal interactions in odontogenesis.

3.
J Dev Biol ; 9(4)2021 Sep 22.
Article in English | MEDLINE | ID: mdl-34698187

ABSTRACT

In this case report, we focus on Muenke syndrome (MS), a disease caused by the p.Pro250Arg variant in fibroblast growth factor receptor 3 (FGFR3) and characterized by uni- or bilateral coronal suture synostosis, macrocephaly without craniosynostosis, dysmorphic craniofacial features, and dental malocclusion. The clinical findings of MS are further complicated by variable expression of phenotypic traits and incomplete penetrance. As such, unraveling the mechanisms behind MS will require a comprehensive and systematic way of phenotyping patients to precisely identify the impact of the mutation variant on craniofacial development. To establish this framework, we quantitatively delineated the craniofacial phenotype of an individual with MS and compared this to his unaffected parents using three-dimensional cephalometric analysis of cone beam computed tomography scans and geometric morphometric analysis, in addition to an extensive clinical evaluation. Secondly, given the utility of human induced pluripotent stem cells (hiPSCs) as a patient-specific investigative tool, we also generated the first hiPSCs derived from a family trio, the proband and his unaffected parents as controls, with detailed characterization of all cell lines. This report provides a starting point for evaluating the mechanistic underpinning of the craniofacial development in MS with the goal of linking specific clinical manifestations to molecular insights gained from hiPSC-based disease modeling.

4.
Stem Cell Res ; 46: 101823, 2020 07.
Article in English | MEDLINE | ID: mdl-32505898

ABSTRACT

Muenke syndrome is the leading genetic cause of craniosynostosis and results in a variety of disabling clinical phenotypes. To model the disease and study the pathogenic mechanisms, a human induced pluripotent stem cell (hiPSC) line was generated from a patient diagnosed with Muenke syndrome. Successful reprogramming was validated by morphological features, karyotyping, loss of reprogramming factors, expression of pluripotency markers, mutation analysis and teratoma formation.


Subject(s)
Craniosynostoses , Induced Pluripotent Stem Cells , Craniosynostoses/genetics , Humans , Mutation , Phenotype , Receptor, Fibroblast Growth Factor, Type 3/genetics
5.
Biotechnol J ; 11(1): 58-70, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26663861

ABSTRACT

Although skin grafting is one of the most advanced cell therapy technique, wide application of skin substitutes is hampered by the difficulty in securing sufficient amount of epidermal substitute. Additionally, in understanding the progression of skin aging and disease, and in screening the cosmetic and pharmaceutical products, there is lack of a satisfactory human skin-specific in vitro model. Recently, human embryonic stem cells (hESCs) have been proposed as an unlimited and reliable cell source to obtain almost all cell types present in the human body. This review focuses on the potential off-the-shelf use of hESC-derived keratinocytes for future clinical applications as well as a powerful in vitro skin model to study skin function and integrity, host-pathogen interactions and disease pathogenesis. Furthermore, we discuss the industrial applications of hESC-derived keratinized multi-layer epithelium which provides a human-like test platform for understanding disease pathogenesis, evaluation of new therapeutic modalities and assessment of the safety and efficacy of skin cosmetics and therapeutics. Overall, we conclude that the hESC-derived keratinocytes have great potential for clinical, research and industrial applications.


Subject(s)
Human Embryonic Stem Cells/cytology , Keratinocytes/cytology , Models, Biological , Cell Differentiation , Host-Pathogen Interactions , Humans , Skin/cytology , Tissue Engineering
6.
Stem Cell Reports ; 4(2): 190-8, 2015 Feb 10.
Article in English | MEDLINE | ID: mdl-25680477

ABSTRACT

We generated a RUNX2-yellow fluorescent protein (YFP) reporter system to study osteogenic development from human embryonic stem cells (hESCs). Our studies demonstrate the fidelity of YFP expression with expression of RUNX2 and other osteogenic genes in hESC-derived osteoprogenitor cells, as well as the osteogenic specificity of YFP signal. In vitro studies confirm that the hESC-derived YFP(+) cells have similar osteogenic phenotypes to osteoprogenitor cells generated from bone-marrow mesenchymal stem cells. In vivo studies demonstrate the hESC-derived YFP(+) cells can repair a calvarial defect in immunodeficient mice. Using the engineered hESCs, we monitored the osteogenic development and explored the roles of osteogenic supplements BMP2 and FGF9 in osteogenic differentiation of these hESCs in vitro. Taken together, this reporter system provides a novel system to monitor the osteogenic differentiation of hESCs and becomes useful to identify soluble agents and cell signaling pathways that mediate early stages of human bone development.


Subject(s)
Cell Differentiation/genetics , Core Binding Factor Alpha 1 Subunit/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Osteogenesis/genetics , Animals , Cell Line , Core Binding Factor Alpha 1 Subunit/metabolism , Gene Expression , Genes, Reporter , Humans , Immunophenotyping , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Phenotype
7.
Methods Mol Biol ; 1195: 13-22, 2014.
Article in English | MEDLINE | ID: mdl-24281868

ABSTRACT

For many years, cell therapies have been hampered by limited availability and inter-batch variability of primary cells. Human embryonic stem cell (hESC) can give rise to specialized cells like keratinocytes and recently emerged as a virtually unlimited source of potential therapeutic cells. However, xenogeneic components in differentiation cocktails have been limiting the clinical potential of hESC-derived keratinocytes (hESCs-Kert). Here, we demonstrated efficient differentiation of H9 human embryonic stem cells (H9-hESCs) into keratinocytes (H9-Kert(ACC)) in an autogenic co-culture system. We used activin as the main factor to induce keratinocyte differentiation. H9-Kert(ACC) expressed keratinocyte markers at mRNA and protein levels. Establishment of such animal-free microenvironment for keratinocyte differentiation will accelerate potential clinical application of hESCs.


Subject(s)
Cell Differentiation , Coculture Techniques/methods , Embryonic Stem Cells/cytology , Epidermal Cells , Keratinocytes/cytology , Animals , Embryonic Stem Cells/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Humans , Mice , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Staining and Labeling
8.
J Invest Dermatol ; 133(3): 618-628, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23235526

ABSTRACT

Human embryonic stem cells (hESCs)-derived keratinocytes hold great clinical and research potential. However, the current techniques are hampered by the use of xenogenic components that limits their clinical application. Here we demonstrated an efficient differentiation of H9 hESCs (H9-hESCs) into keratinocytes (H9-Kert) with the minimum use of animal-derived materials. For differentiation, we established two microenvironment systems originated from H9-hESCs (autogenic microenvironment). These autogenic microenvironment systems consist of an autogenic coculture system (ACC) and an autogenic feeder-free system (AFF). In addition, we showed a stage-specific effect of Activin in promoting keratinocyte differentiation from H9-hESCs while repressing the expression of early neural markers in the ACC system. Furthermore, we also explained the effect of Activin in construction of the AFF system made up of extracellular matrix similar to basement membrane extracted from H9-hESC-derived fibroblasts. H9-Kert differentiated in both systems expressed keratinocyte markers at mRNA and protein levels. H9-Kert were also able to undergo terminal differentiation in high Ca(2+) medium. These findings support the transition toward the establishment of an animal-free microenvironment for successful differentiation of hESCs into keratinocytes for potential clinical application.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/cytology , Keratinocytes/cytology , Stem Cell Niche , Activins/pharmacology , Calcium/pharmacology , Cell Differentiation/drug effects , Cell Line , Coculture Techniques , Culture Media, Conditioned/pharmacology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/physiology , Humans , Keratinocytes/drug effects , Keratinocytes/physiology , Tretinoin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...