Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 7.511
Filtrer
1.
Stem Cell Res Ther ; 15(1): 193, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38956724

RÉSUMÉ

BACKGROUND: The human induced pluripotent stem cells (hiPSCs) can generate all the cells composing the human body, theoretically. Therefore, hiPSCs are thought to be a candidate source of stem cells for regenerative medicine. The major challenge of allogeneic hiPSC-derived cell products is their immunogenicity. The hypoimmunogenic cell strategy is allogenic cell therapy without using immune suppressants. Advances in gene engineering technology now permit the generation of hypoimmunogenic cells to avoid allogeneic immune rejection. In this study, we generated a hypoimmunogenic hiPSC (HyPSC) clone that had diminished expression of human leukocyte antigen (HLA) class Ia and class II and expressed immune checkpoint molecules and a safety switch. METHODS: First, we generated HLA class Ia and class II double knockout (HLA class Ia/II DKO) hiPSCs. Then, a HyPSC clone was generated by introducing exogenous ß-2-microglobulin (B2M), HLA-G, PD-L1, and PD-L2 genes, and the Rapamycin-activated Caspase 9 (RapaCasp9)-based suicide gene as a safety switch into the HLA class Ia/II DKO hiPSCs. The characteristics and immunogenicity of the HyPSCs and their derivatives were analyzed. RESULTS: We found that the expression of HLA-G on the cell surface can be enhanced by introducing the exogenous HLA-G gene along with B2M gene into HLA class Ia/II DKO hiPSCs. The HyPSCs retained a normal karyotype and had the characteristics of pluripotent stem cells. Moreover, the HyPSCs could differentiate into cells of all three germ layer lineages including CD45+ hematopoietic progenitor cells (HPCs), functional endothelial cells, and hepatocytes. The HyPSCs-derived HPCs exhibited the ability to evade innate and adaptive immunity. Further, we demonstrated that RapaCasp9 could be used as a safety switch in vitro and in vivo. CONCLUSION: The HLA class Ia/II DKO hiPSCs armed with HLA-G, PD-L1, PD-L2, and RapaCasp9 molecules are a potential source of stem cells for allogeneic transplantation.


Sujet(s)
Immunité acquise , Antigène CD274 , Antigènes HLA-G , Immunité innée , Cellules souches pluripotentes induites , Ligand-2 de la protéine-1 de mort cellulaire programmée , Humains , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/cytologie , Cellules souches pluripotentes induites/immunologie , Antigène CD274/métabolisme , Antigène CD274/génétique , Antigène CD274/immunologie , Antigènes HLA-G/génétique , Antigènes HLA-G/métabolisme , Antigènes HLA-G/immunologie , Ligand-2 de la protéine-1 de mort cellulaire programmée/métabolisme , Ligand-2 de la protéine-1 de mort cellulaire programmée/génétique , Animaux , Souris
2.
Cell Stem Cell ; 31(7): 941-943, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38971143

RÉSUMÉ

Induced pluripotent stem cell-derived cardiomyocyte (iPSC-CM) therapy has emerged as a highly promising field of heart repair. Lin et al.1 presented compelling evidence on the long-term engraftment and maturation of autologous iPSC-CMs in two rhesus macaques, demonstrating unprecedented cardiac autografting data in large animal models without the need of immunosuppressants.


Sujet(s)
Cellules souches pluripotentes induites , Macaca mulatta , Myocytes cardiaques , Animaux , Cellules souches pluripotentes induites/cytologie , Myocytes cardiaques/cytologie , Autogreffes , Humains , Survie cellulaire , Différenciation cellulaire
3.
Cells ; 13(13)2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38994988

RÉSUMÉ

Bioelectric signals possess the ability to robustly control and manipulate patterning during embryogenesis and tissue-level regeneration. Endogenous local and global electric fields function as a spatial 'pre-pattern', controlling cell fates and tissue-scale anatomical boundaries; however, the mechanisms facilitating these robust multiscale outcomes are poorly characterized. Computational modeling addresses the need to predict in vitro patterning behavior and further elucidate the roles of cellular bioelectric signaling components in patterning outcomes. Here, we modified a previously designed image pattern recognition algorithm to distinguish unique spatial features of simulated non-excitable bioelectric patterns under distinct cell culture conditions. This algorithm was applied to comparisons between simulated patterns and experimental microscopy images of membrane potential (Vmem) across cultured human iPSC colonies. Furthermore, we extended the prediction to a novel co-culture condition in which cell sub-populations possessing different ionic fluxes were simulated; the defining spatial features were recapitulated in vitro with genetically modified colonies. These results collectively inform strategies for modeling multiscale spatial characteristics that emerge in multicellular systems, characterizing the molecular contributions to heterogeneity of membrane potential in non-excitable cells, and enabling downstream engineered bioelectrical tissue design.


Sujet(s)
Cellules souches pluripotentes induites , Potentiels de membrane , Humains , Cellules souches pluripotentes induites/cytologie , Cellules souches pluripotentes induites/métabolisme , Potentiels de membrane/physiologie , Algorithmes , Simulation numérique , Modèles biologiques , Techniques de coculture
4.
Cells ; 13(13)2024 Jul 06.
Article de Anglais | MEDLINE | ID: mdl-38995007

RÉSUMÉ

Primary cilia are finger-like sensory organelles that extend from the bodies of most cell types and have a distinct lipid and protein composition from the plasma membrane. This partitioning is maintained by a diffusion barrier that restricts the entry of non-ciliary proteins, and allows the selective entry of proteins harboring a ciliary targeting sequence (CTS). However, CTSs are not stereotyped and previously reported sequences are insufficient to drive efficient ciliary localisation across diverse cell types. Here, we describe a short peptide sequence that efficiently targets transmembrane proteins to primary cilia in all tested cell types, including human neurons. We generate human-induced pluripotent stem cell (hiPSC) lines stably expressing a transmembrane construct bearing an extracellular HaloTag and intracellular fluorescent protein, which enables the bright, specific labeling of primary cilia in neurons and other cell types to facilitate studies of cilia in health and disease. We demonstrate the utility of this resource by developing an image analysis pipeline for the automated measurement of primary cilia to detect changes in their length associated with altered signaling or disease state.


Sujet(s)
Cils vibratiles , Cellules souches pluripotentes induites , Protéines membranaires , Cils vibratiles/métabolisme , Humains , Protéines membranaires/métabolisme , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/cytologie , Animaux , Neurones/métabolisme , Séquence d'acides aminés , Lignée cellulaire , Transport des protéines
5.
Sci Rep ; 14(1): 15760, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38977828

RÉSUMÉ

Manufacturing regenerative medicine requires continuous monitoring of pluripotent cell culture and quality assessment while eliminating cell destruction and contaminants. In this study, we employed a novel method to monitor the pluripotency of stem cells through image analysis, avoiding the traditionally used invasive procedures. This approach employs machine learning algorithms to analyze stem cell images to predict the expression of pluripotency markers, such as OCT4 and NANOG, without physically interacting with or harming cells. We cultured induced pluripotent stem cells under various conditions to induce different pluripotent states and imaged the cells using bright-field microscopy. Pluripotency states of induced pluripotent stem cells were assessed using invasive methods, including qPCR, immunostaining, flow cytometry, and RNA sequencing. Unsupervised and semi-supervised learning models were applied to evaluate the results and accurately predict the pluripotency of the cells using only image analysis. Our approach directly links images to invasive assessment results, making the analysis of cell labeling and annotation of cells in images by experts dispensable. This core achievement not only contributes for safer and more reliable stem cell research but also opens new avenues for real-time monitoring and quality control in regenerative medicine manufacturing. Our research fills an important gap in the field by providing a viable, noninvasive alternative to traditional invasive methods for assessing pluripotency. This innovation is expected to make a significant contribution to improving regenerative medicine manufacturing because it will enable a more detailed and feasible understanding of cellular status during the manufacturing process.


Sujet(s)
Marqueurs biologiques , Cellules souches pluripotentes induites , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/cytologie , Marqueurs biologiques/métabolisme , Humains , Facteur de transcription Oct-3/métabolisme , Facteur de transcription Oct-3/génétique , Protéine homéotique Nanog/métabolisme , Protéine homéotique Nanog/génétique , Traitement d'image par ordinateur/méthodes , Apprentissage machine , Médecine régénérative/méthodes , Cytométrie en flux/méthodes , Animaux , Différenciation cellulaire , Cellules cultivées
6.
ACS Biomater Sci Eng ; 10(7): 4525-4540, 2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38973308

RÉSUMÉ

Lumenogenesis within the epiblast represents a critical step in early human development, priming the embryo for future specification and patterning events. However, little is known about the specific mechanisms that drive this process due to the inability to study the early embryo in vivo. While human pluripotent stem cell (hPSC)-based models recapitulate many aspects of the human epiblast, most approaches for generating these 3D structures rely on ill-defined, reconstituted basement membrane matrices. Here, we designed synthetic, nonadhesive polyethylene glycol (PEG) hydrogel matrices to better understand the role of matrix mechanical cues in iPSC morphogenesis, specifically elastic modulus. First, we identified a narrow range of hydrogel moduli that were conducive to the hPSC viability, pluripotency, and differentiation. We then used this platform to investigate the effects of the hydrogel modulus on lumenogenesis, finding that matrices of intermediate stiffness yielded the most epiblast-like aggregates. Conversely, stiffer matrices impeded lumen formation and apico-basal polarization, while the softest matrices yielded polarized but aberrant structures. Our approach offers a simple, modular platform for modeling the human epiblast and investigating the role of matrix cues in its morphogenesis.


Sujet(s)
Différenciation cellulaire , Hydrogels , Morphogenèse , Polyéthylène glycols , Humains , Hydrogels/composition chimique , Hydrogels/pharmacologie , Polyéthylène glycols/composition chimique , Polyéthylène glycols/pharmacologie , Différenciation cellulaire/effets des médicaments et des substances chimiques , Cellules souches pluripotentes/cytologie , Cellules souches pluripotentes/effets des médicaments et des substances chimiques , Feuillets embryonnaires/cytologie , Module d'élasticité , Cellules souches pluripotentes induites/cytologie , Cellules souches pluripotentes induites/effets des médicaments et des substances chimiques
7.
Cell Commun Signal ; 22(1): 356, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38982464

RÉSUMÉ

BACKGROUND: Stem cell-derived extracellular vesicles (EVs) are an emerging class of therapeutics with excellent biocompatibility, bioactivity and pro-regenerative capacity. One of the potential targets for EV-based medicines are cardiovascular diseases (CVD). In this work we used EVs derived from human induced pluripotent stem cells (hiPSCs; hiPS-EVs) cultured under different oxygen concentrations (21, 5 and 3% O2) to dissect the molecular mechanisms responsible for cardioprotection. METHODS: EVs were isolated by ultrafiltration combined with size exclusion chromatography (UF + SEC), followed by characterization by nanoparticle tracking analysis, atomic force microscopy (AFM) and Western blot methods. Liquid chromatography and tandem mass spectrometry coupled with bioinformatic analyses were used to identify differentially enriched proteins in various oxygen conditions. We directly compared the cardioprotective effects of these EVs in an oxygen-glucose deprivation/reoxygenation (OGD/R) model of cardiomyocyte (CM) injury. Using advanced molecular biology, fluorescence microscopy, atomic force spectroscopy and bioinformatics techniques, we investigated intracellular signaling pathways involved in the regulation of cell survival, apoptosis and antioxidant response. The direct effect of EVs on NRF2-regulated signaling was evaluated in CMs following NRF2 inhibition with ML385. RESULTS: We demonstrate that hiPS-EVs derived from physiological hypoxia at 5% O2 (EV-H5) exert enhanced cytoprotective function towards damaged CMs compared to EVs derived from other tested oxygen conditions (normoxia; EV-N and hypoxia 3% O2; EV-H3). This resulted from higher phosphorylation rates of Akt kinase in the recipient cells after transfer, modulation of AMPK activity and reduced apoptosis. Furthermore, we provide direct evidence for improved calcium signaling and sustained contractility in CMs treated with EV-H5 using AFM measurements. Mechanistically, our mass spectrometry and bioinformatics analyses revealed differentially enriched proteins in EV-H5 associated with the antioxidant pathway regulated by NRF2. In this regard, EV-H5 increased the nuclear translocation of NRF2 protein and enhanced its transcription in CMs upon OGD/R. In contrast, inhibition of NRF2 with ML385 abolished the protective effect of EVs on CMs. CONCLUSIONS: In this work, we demonstrate a superior cardioprotective function of EV-H5 compared to EV-N and EV-H3. Such EVs were most effective in restoring redox balance in stressed CMs, preserving their contractile function and preventing cell death. Our data support the potential use of hiPS-EVs derived from physiological hypoxia, as cell-free therapeutics with regenerative properties for the treatment of cardiac diseases.


Sujet(s)
Antioxydants , Vésicules extracellulaires , Cellules souches pluripotentes induites , Myocytes cardiaques , Facteur-2 apparenté à NF-E2 , Protéines proto-oncogènes c-akt , Transduction du signal , Myocytes cardiaques/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Vésicules extracellulaires/métabolisme , Facteur-2 apparenté à NF-E2/métabolisme , Humains , Protéines proto-oncogènes c-akt/métabolisme , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/cytologie , Transduction du signal/effets des médicaments et des substances chimiques , Antioxydants/pharmacologie , Stress oxydatif/effets des médicaments et des substances chimiques , Hypoxie cellulaire/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Extracellular Signal-Regulated MAP Kinases/métabolisme , Animaux
8.
Curr Protoc ; 4(7): e1101, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38980221

RÉSUMÉ

Cardiovascular diseases have emerged as one of the leading causes of human mortality, but the discovery of new drugs has been hindered by the absence of suitable in vitro platforms. In recent decades, continuously refined protocols for differentiating human induced pluripotent stem cells (hiPSCs) into hiPSC-derived cardiomyocytes (hiPSC-CMs) have significantly advanced disease modeling and drug screening; however, this has led to an increasing need to monitor the function of hiPSC-CMs. The precise regulation of action potentials (APs) and intracellular calcium (Ca2+) transients is critical for proper excitation-contraction coupling and cardiomyocyte function. These important parameters are usually adversely affected in cardiovascular diseases or under cardiotoxic conditions and can be measured using optical imaging-based techniques. However, this procedure is complex and technologically challenging. We have adapted the IonOptix system to simultaneously measure APs and Ca2+ transients in hiPSC-CMs loaded with the fluorescent dyes FluoVolt and Rhod 2, respectively. This system serves as a powerful high-throughput platform to facilitate the discovery of new compounds to treat cardiovascular diseases with the cellular phenotypes of abnormal APs and Ca2+ handling. Here, we present a comprehensive protocol for hiPSC-CM preparation, device setup, optical imaging, and data analysis. © 2024 Wiley Periodicals LLC. Basic Protocol 1: Maintenance and seeding of hiPSC-CMs Basic Protocol 2: Simultaneous detection of action potentials and Ca2+ transients in hiPSC-CMs.


Sujet(s)
Potentiels d'action , Calcium , Cellules souches pluripotentes induites , Myocytes cardiaques , Imagerie optique , Humains , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/cytologie , Cellules souches pluripotentes induites/effets des médicaments et des substances chimiques , Myocytes cardiaques/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Potentiels d'action/effets des médicaments et des substances chimiques , Calcium/métabolisme , Imagerie optique/méthodes , Différenciation cellulaire/effets des médicaments et des substances chimiques
9.
Nat Commun ; 15(1): 5929, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-39009604

RÉSUMÉ

Human iPSC-derived cardiomyocytes (hiPSC-CMs) have proven invaluable for cardiac disease modeling and regeneration. Challenges with quality, inter-batch consistency, cryopreservation and scale remain, reducing experimental reproducibility and clinical translation. Here, we report a robust stirred suspension cardiac differentiation protocol, and we perform extensive morphological and functional characterization of the resulting bioreactor-differentiated iPSC-CMs (bCMs). Across multiple different iPSC lines, the protocol produces 1.2E6/mL bCMs with ~94% purity. bCMs have high viability after cryo-recovery (>90%) and predominantly ventricular identity. Compared to standard monolayer-differentiated CMs, bCMs are more reproducible across batches and have more mature functional properties. The protocol also works with magnetically stirred spinner flasks, which are more economical and scalable than bioreactors. Minor protocol modifications generate cardiac organoids fully in suspension culture. These reproducible, scalable, and resource-efficient approaches to generate iPSC-CMs and organoids will expand their applications, and our benchmark data will enable comparison to cells produced by other cardiac differentiation protocols.


Sujet(s)
Bioréacteurs , Techniques de culture cellulaire , Différenciation cellulaire , Cellules souches pluripotentes induites , Myocytes cardiaques , Organoïdes , Humains , Cellules souches pluripotentes induites/cytologie , Myocytes cardiaques/cytologie , Myocytes cardiaques/physiologie , Organoïdes/cytologie , Techniques de culture cellulaire/méthodes , Reproductibilité des résultats , Cellules cultivées , Cryoconservation/méthodes
10.
Nat Commun ; 15(1): 5834, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38992003

RÉSUMÉ

We present Dystrophic Epidermolysis Bullosa Cell Therapy (DEBCT), a scalable platform producing autologous organotypic iPS cell-derived induced skin composite (iSC) grafts for definitive treatment. Clinical-grade manufacturing integrates CRISPR-mediated genetic correction with reprogramming into one step, accelerating derivation of COL7A1-edited iPS cells from patients. Differentiation into epidermal, dermal and melanocyte progenitors is followed by CD49f-enrichment, minimizing maturation heterogeneity. Mouse xenografting of iSCs from four patients with different mutations demonstrates disease modifying activity at 1 month. Next-generation sequencing, biodistribution and tumorigenicity assays establish a favorable safety profile at 1-9 months. Single cell transcriptomics reveals that iSCs are composed of the major skin cell lineages and include prominent holoclone stem cell-like signatures of keratinocytes, and the recently described Gibbin-dependent signature of fibroblasts. The latter correlates with enhanced graftability of iSCs. In conclusion, DEBCT overcomes manufacturing and safety roadblocks and establishes a reproducible, safe, and cGMP-compatible therapeutic approach to heal lesions of DEB patients.


Sujet(s)
Thérapie cellulaire et tissulaire , Collagène de type VII , Épidermolyse bulleuse dystrophique , Cellules souches pluripotentes induites , Humains , Épidermolyse bulleuse dystrophique/thérapie , Épidermolyse bulleuse dystrophique/génétique , Animaux , Cellules souches pluripotentes induites/transplantation , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/cytologie , Souris , Collagène de type VII/génétique , Collagène de type VII/métabolisme , Thérapie cellulaire et tissulaire/méthodes , Fibroblastes/métabolisme , Différenciation cellulaire , Kératinocytes/métabolisme , Kératinocytes/transplantation , Peau/métabolisme , Transplantation autologue , Mâle , Mutation , Femelle , Transplantation de peau/méthodes , Édition de gène/méthodes , Systèmes CRISPR-Cas
11.
Cell Syst ; 15(7): 649-661.e9, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-38981488

RÉSUMÉ

Organoids derived from human stem cells are a promising approach for disease modeling, regenerative medicine, and fundamental research. However, organoid variability and limited control over morphological outcomes remain as challenges. One open question is the extent to which engineering control over culture conditions can guide organoids to specific compositions. Here, we extend a DNA "velcro" cell patterning approach, precisely controlling the number and ratio of human induced pluripotent stem cell-derived progenitors contributing to nephron progenitor (NP) organoids and mosaic NP/ureteric bud (UB) tip cell organoids within arrays of microwells. We demonstrate long-term control over organoid size and morphology, decoupled from geometric constraints. We then show emergent trends in organoid tissue proportions that depend on initial progenitor cell composition. These include higher nephron and stromal cell representation in mosaic NP/UB organoids vs. NP-only organoids and a "goldilocks" initial cell ratio in mosaic organoids that optimizes the formation of proximal tubule structures.


Sujet(s)
Organoïdes , Organoïdes/cytologie , Organoïdes/métabolisme , Humains , Cellules souches pluripotentes induites/cytologie , Cellules souches pluripotentes induites/métabolisme , Néphrons/cytologie , Différenciation cellulaire/physiologie , Cellules souches/cytologie
12.
Sci Rep ; 14(1): 16129, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38997397

RÉSUMÉ

The choroid, a vascularized tissue situated between the retina and the sclera, plays a crucial role in maintaining ocular homeostasis. Despite its significance, research on choroidal abnormalities and the establishment of effective in vitro models have been limited. In this study, we developed an in vitro choroid model through the co-culture of human induced pluripotent stem cells (hiPSC)-derived endothelial cells (ECs) and mouse choroidal fibroblasts (msCFs) with hiPSC-derived retinal pigment epithelial (RPE) cells via a permeable membrane. This model, inclusive of ECs, CFs, and RPE cells, exhibited similarities with in vivo choroidal vessels, as confirmed through immunohistochemistry of extracellular matrix markers and vascular-related markers, as well as choroid angiogenesis sprouting assay analysis. The effectiveness of our in vitro model was demonstrated in assessing vascular changes induced by drugs targeting vasoregulation. Our model offers a valuable tool for gaining insights into the pathological mechanisms underlying choroid development and the progression of choroidal vascular diseases.


Sujet(s)
Choroïde , Techniques de coculture , Cellules endothéliales , Cellules souches pluripotentes induites , Épithélium pigmentaire de la rétine , Choroïde/vascularisation , Choroïde/métabolisme , Animaux , Humains , Souris , Cellules souches pluripotentes induites/cytologie , Cellules souches pluripotentes induites/métabolisme , Cellules endothéliales/métabolisme , Épithélium pigmentaire de la rétine/métabolisme , Épithélium pigmentaire de la rétine/cytologie , Fibroblastes/métabolisme , Néovascularisation choroïdienne/métabolisme , Néovascularisation choroïdienne/anatomopathologie , Cellules cultivées
13.
Molecules ; 29(13)2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38999126

RÉSUMÉ

Given the pivotal role of neuronal populations in various biological processes, assessing their collective output is crucial for understanding the nervous system's complex functions. Building on our prior development of a spiral scanning mechanism for the rapid acquisition of Raman spectra from single cells and incorporating machine learning for label-free evaluation of cell states, we investigated whether the Paint Raman Express Spectroscopy System (PRESS) can assess neuronal activities. We tested this hypothesis by examining the chemical responses of glutamatergic neurons as individual neurons and autonomic neuron ganglia as neuronal populations derived from human-induced pluripotent stem cells. The PRESS successfully acquired Raman spectra from both individual neurons and ganglia within a few seconds, achieving a signal-to-noise ratio sufficient for detailed analysis. To evaluate the ligand responsiveness of the induced neurons and ganglia, the Raman spectra were subjected to principal component and partial least squares discriminant analyses. The PRESS detected neuronal activity in response to glutamate and nicotine, which were absent in the absence of calcium. Additionally, the PRESS induced dose-dependent neuronal activity changes. These findings underscore the capability of the PRESS to assess individual neuronal activity and elucidate neuronal population dynamics and pharmacological responses, heralding new opportunities for drug discovery and regenerative medicine advancement.


Sujet(s)
Acide glutamique , Cellules souches pluripotentes induites , Neurones , Analyse spectrale Raman , Analyse spectrale Raman/méthodes , Neurones/métabolisme , Neurones/physiologie , Humains , Cellules souches pluripotentes induites/cytologie , Cellules souches pluripotentes induites/métabolisme , Nicotine/pharmacologie , Analyse en composantes principales
14.
Methods Mol Biol ; 2805: 89-100, 2024.
Article de Anglais | MEDLINE | ID: mdl-39008175

RÉSUMÉ

Engineered heart tissues (EHTs) have been shown to be a valuable platform for disease investigation and therapeutic testing by increasing human induced pluripotent stem cell-derived cardiomyocyte (hiPSC-CM) maturity and better recreating the native cardiac environment. The protocol detailed in this chapter describes the generation of miniaturized EHTs (mEHTs) incorporating hiPSC-CMs and human stromal cells in a fibrin hydrogel. This platform utilizes an array of silicone posts designed to fit in a standard 96-well tissue culture plate. Stromal cells and hiPSC-CMs are cast in a fibrin matrix suspended between two silicone posts, forming an mEHT that produces synchronous muscle contractions. The platform presented here has the potential to be used for high throughput characterization and screening of disease phenotypes and novel therapeutics through measurements of the myocardial function, including contractile force and calcium handling, and its compatibility with immunostaining.


Sujet(s)
Cellules souches pluripotentes induites , Myocytes cardiaques , Ingénierie tissulaire , Humains , Ingénierie tissulaire/méthodes , Myocytes cardiaques/cytologie , Myocytes cardiaques/métabolisme , Cellules souches pluripotentes induites/cytologie , Cellules souches pluripotentes induites/métabolisme , Hydrogels/composition chimique , Différenciation cellulaire , Fibrine/métabolisme , Cellules cultivées , Techniques de culture cellulaire/méthodes , Cellules stromales/cytologie , Techniques de culture de tissus/méthodes , Techniques de culture de tissus/instrumentation
15.
ACS Appl Mater Interfaces ; 16(28): 36030-36046, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-38951110

RÉSUMÉ

Differentiation of induced pluripotent stem cells (iPSCs) is an extremely complex process that has proven difficult to study. In this research, we utilized nanotopography to elucidate details regarding iPSC differentiation by developing a nanodot platform consisting of nanodot arrays of increasing diameter. Subjecting iPSCs cultured on the nanodot platform to a cardiomyocyte (CM) differentiation protocol revealed several significant gene expression profiles that were associated with poor differentiation. The observed expression trends were used to select existing small-molecule drugs capable of modulating differentiation efficiency. BRD K98 was repurposed to inhibit CM differentiation, while iPSCs treated with NSC-663284, carmofur, and KPT-330 all exhibited significant increases in not only CM marker expression but also spontaneous beating, suggesting improved CM differentiation. In addition, quantitative polymerase chain reaction was performed to determine the gene regulation responsible for modulating differentiation efficiency. Multiple genes involved in extracellular matrix remodeling were correlated with a CM differentiation efficiency, while genes involved in the cell cycle exhibited contrasting expression trends that warrant further studies. The results suggest that expression profiles determined via short time-series expression miner analysis of nanodot-cultured iPSC differentiation can not only reveal drugs capable of enhancing differentiation efficiency but also highlight crucial sets of genes related to processes such as extracellular matrix remodeling and the cell cycle that can be targeted for further investigation. Our findings confirm that the nanodot platform can be used to reveal complex mechanisms behind iPSC differentiation and could be an indispensable tool for optimizing iPSC technology for clinical applications.


Sujet(s)
Différenciation cellulaire , Cellules souches pluripotentes induites , Myocytes cardiaques , Cellules souches pluripotentes induites/cytologie , Cellules souches pluripotentes induites/effets des médicaments et des substances chimiques , Cellules souches pluripotentes induites/métabolisme , Différenciation cellulaire/effets des médicaments et des substances chimiques , Myocytes cardiaques/cytologie , Myocytes cardiaques/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Humains , Nanoparticules/composition chimique , Cellules cultivées , Nanostructures/composition chimique
16.
Curr Protoc ; 4(7): e1096, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38984433

RÉSUMÉ

With recent advances in the reprogramming of somatic cells into induced Pluripotent Stem Cells (iPSCs), gene editing technologies, and protocols for the directed differentiation of stem cells into heterogeneous tissues, iPSC-derived kidney organoids have emerged as a useful means to study processes of renal development and disease. Considerable advances guided by knowledge of fundamental renal developmental signaling pathways have been made with the use of exogenous morphogens to generate more robust kidney-like tissues in vitro. However, both biochemical and biophysical microenvironmental cues are major influences on tissue development and self-organization. In the context of engineering the biophysical aspects of the microenvironment, the use of hydrogel extracellular scaffolds for organoid studies has been gaining interest. Two families of hydrogels have recently been the subject of significant attention: self-assembling peptide hydrogels (SAPHs), which are fully synthetic and chemically defined, and gelatin methacryloyl (GelMA) hydrogels, which are semi-synthetic. Both can be used as support matrices for growing kidney organoids. Based on our recently published work, we highlight methods describing the generation of human iPSC (hiPSC)-derived kidney organoids and their maturation within SAPHs and GelMA hydrogels. We also detail protocols required for the characterization of such organoids using immunofluorescence imaging. Together, these protocols should enable the user to grow hiPSC-derived kidney organoids within hydrogels of this kind and evaluate the effects that the biophysical microenvironment provided by the hydrogels has on kidney organoid maturation. © 2024 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Directed differentiation of human induced pluripotent stem cells (hiPSCs) into kidney organoids and maturation within mechanically tunable self-assembling peptide hydrogels (SAPHs) Alternate Protocol: Encapsulation of day 9 nephron progenitor aggregates in gelatin methacryloyl (GelMA) hydrogels. Support Protocol 1: Human induced pluripotent stem cell (hiPSC) culture. Support Protocol 2: Organoid fixation with paraformaldehyde (PFA) Basic Protocol 2: Whole-mount immunofluorescence imaging of kidney organoids. Basic Protocol 3: Immunofluorescence of organoid cryosections.


Sujet(s)
Hydrogels , Cellules souches pluripotentes induites , Rein , Organoïdes , Cellules souches pluripotentes induites/cytologie , Organoïdes/cytologie , Hydrogels/composition chimique , Humains , Rein/cytologie , Techniques de culture cellulaire/méthodes , Différenciation cellulaire
17.
Stem Cell Res Ther ; 15(1): 191, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38956608

RÉSUMÉ

BACKGROUND: Stem cell-derived therapies hold the potential for treatment of regenerative clinical indications. Static culture has a limited ability to scale up thus restricting its use. Suspension culturing can be used to produce target cells in large quantities, but also presents challenges related to stress and aggregation stability. METHODS: Utilizing a design of experiments (DoE) approach in vertical wheel bioreactors, we evaluated media additives that have versatile properties. The additives evaluated are Heparin sodium salt (HS), polyethylene glycol (PEG), poly (vinyl alcohol) (PVA), Pluronic F68 and dextran sulfate (DS). Multiple response variables were chosen to assess cell growth, pluripotency maintenance and aggregate stability in response to the additive inputs, and mathematical models were generated and tuned for maximal predictive power. RESULTS: Expansion of iPSCs using 100 ml vertical wheel bioreactor assay for 4 days on 19 different media combinations resulted in models that can optimize pluripotency, stability, and expansion. The expansion optimization resulted in the combination of PA, PVA and PEG with E8. This mixture resulted in an expansion doubling time that was 40% shorter than that of E8 alone. Pluripotency optimizer highlighted the importance of adding 1% PEG to the E8 medium. Aggregate stability optimization that minimizes aggregate fusion in 3D culture indicated that the interaction of both Heparin and PEG can limit aggregation as well as increase the maintenance capacity and expansion of hiPSCs, suggesting that controlling fusion is a critical parameter for expansion and maintenance. Validation of optimized solution on two cell lines in bioreactors with decreased speed of 40 RPM, showed consistency and prolonged control over aggregates that have high frequency of pluripotency markers of OCT4 and SOX2 (> 90%). A doubling time of around 1-1.4 days was maintained after passaging as clumps in the optimized medium. Controlling aggregate fusion allowed for a decrease in bioreactor speed and therefore shear stress exerted on the cells in a large-scale expansion. CONCLUSION: This study resulted in a control of aggregate size within suspension cultures, while informing about concomitant state control of the iPSC state. Wider application of this approach can address media optimization complexity and bioreactor scale-up challenges.


Sujet(s)
Bioréacteurs , Cellules souches pluripotentes induites , Humains , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/cytologie , Techniques de culture cellulaire/méthodes , Prolifération cellulaire , Agrégation cellulaire/effets des médicaments et des substances chimiques , Polyéthylène glycols/composition chimique , Polyéthylène glycols/pharmacologie , Différenciation cellulaire
18.
Curr Protoc ; 4(7): e1097, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39036931

RÉSUMÉ

In the heart in vivo, vasculature forms a semi-permeable endothelial barrier for selective nutrient and (immune) cell delivery to the myocardium and removal of waste products. Crosstalk between the vasculature and the heart cells regulates homeostasis in health and disease. To model heart development and disease in vitro it is important that essential features of this crosstalk are captured. Cardiac organoid and microtissue models often integrate endothelial cells (ECs) to form microvascular networks inside the 3D structure. However, in static culture without perfusion, these networks may fail to show essential functionality. Here, we describe a protocol to generate an in vitro model of human induced pluripotent stem cell (hiPSC)-derived vascularized cardiac microtissues on a microfluidic organ-on-chip platform (VMToC) in which the blood vessels are perfusable. First, prevascularized cardiac microtissues (MT) are formed by combining hiPSC-derived cardiomyocytes, ECs, and cardiac fibroblasts in a pre-defined ratio. Next, these prevascularized MTs are integrated in the chips in a fibrin hydrogel containing additional vascular cells, which self-organize into tubular structures. The MTs become vascularized through anastomosis between the pre-existing microvasculature in the MT and the external vascular network. The VMToCs are then ready for downstream structural and functional assays and basic characterization. Using this protocol, cardiac MTs can be efficiently and robustly vascularized and perfused within 7 days. In vitro vascularized organoid and MT models have the potential to transition current 3D cardiac models to more physiologically relevant organ models that allow the role of the endothelial barrier in drug and inflammatory response to be investigated. © 2024 The Author(s). Current Protocols published by Wiley Periodicals LLC. Basic Protocol: Generation of VMToC Support Protocol 1: Functional Characterization of VMToC Support Protocol 2: Structural Characterization of VMToC.


Sujet(s)
Cellules souches pluripotentes induites , Laboratoires sur puces , Myocytes cardiaques , Humains , Cellules souches pluripotentes induites/cytologie , Cellules souches pluripotentes induites/métabolisme , Myocytes cardiaques/cytologie , Cellules endothéliales/cytologie , Myocarde/cytologie , Myocarde/métabolisme , Différenciation cellulaire
19.
Cells ; 13(12)2024 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-38920642

RÉSUMÉ

The advent of induced pluripotent stem cell (iPSC) technology has brought about transformative advancements in regenerative medicine, offering novel avenues for disease modeling, drug testing, and cell-based therapies. Patient-specific iPSC-based treatments hold the promise of mitigating immune rejection risks. However, the intricacies and costs of producing autologous therapies present commercial challenges. The hair follicle is a multi-germ layered versatile cell source that can be harvested at any age. It is a rich source of keratinocytes, fibroblasts, multipotent stromal cells, and the newly defined Hair Follicle-Associated Pluripotent Stem Cells (HAP). It can also be obtained non-invasively and transported via regular mail channels, making it the ideal starting material for an autologous biobank. In this study, cryopreserved hair follicle-derived iPSC lines (HF-iPS) were established through integration-free vectors, encompassing a diverse cohort. These genetically stable lines exhibited robust expression of pluripotency markers, and showcased tri-lineage differentiation potential. The HF-iPSCs effectively differentiated into double-positive cKIT+/CXCR4+ definitive endoderm cells and NKX6.1+/PDX1+ pancreatic progenitor cells, affirming their pluripotent attributes. We anticipate that the use of plucked hair follicles as an accessible, non-invasive cell source to obtain patient cells, in conjunction with the use of episomal vectors for reprogramming, will improve the future generation of clinically applicable pancreatic progenitor cells for the treatment of Type I Diabetes.


Sujet(s)
Différenciation cellulaire , Follicule pileux , Cellules souches pluripotentes induites , Cellules souches pluripotentes induites/cytologie , Cellules souches pluripotentes induites/métabolisme , Humains , Follicule pileux/cytologie , Follicule pileux/métabolisme , Pancréas/cytologie , Pancréas/métabolisme , Femelle
20.
Cells ; 13(12)2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38920646

RÉSUMÉ

Dopaminergic neurons are the predominant brain cells affected in Parkinson's disease. With the limited availability of live human brain dopaminergic neurons to study pathological mechanisms of Parkinson's disease, dopaminergic neurons have been generated from human-skin-cell-derived induced pluripotent stem cells. Originally, induced pluripotent stem-cell-derived dopaminergic neurons were generated using small molecules. These neurons took more than two months to mature. However, the transcription-factor-mediated differentiation of induced pluripotent stem cells has revealed quicker and cheaper methods to generate dopaminergic neurons. In this study, we compared and contrasted three protocols to generate induced pluripotent stem-cell-derived dopaminergic neurons using transcription-factor-mediated directed differentiation. We deviated from the established protocols using lentivirus transduction to stably integrate different transcription factors into the AAVS1 safe harbour locus of induced pluripotent stem cells. We used different media compositions to generate more than 90% of neurons in the culture, out of which more than 85% of the neurons were dopaminergic neurons within three weeks. Therefore, from our comparative study, we reveal that a combination of transcription factors along with small molecule treatment may be required to generate a pure population of human dopaminergic neurons.


Sujet(s)
Différenciation cellulaire , Neurones dopaminergiques , Cellules souches pluripotentes induites , Facteurs de transcription , Humains , Neurones dopaminergiques/métabolisme , Neurones dopaminergiques/cytologie , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/cytologie , Facteurs de transcription/métabolisme , Lentivirus/génétique , Lentivirus/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE