Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 126
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell Mol Life Sci ; 81(1): 197, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38664263

RESUMEN

Congenital heart defects are associated with significant health challenges, demanding a deep understanding of the underlying biological mechanisms and, thus, better devices or platforms that can recapitulate human cardiac development. The discovery of human pluripotent stem cells has substantially reduced the dependence on animal models. Recent advances in stem cell biology, genetic editing, omics, microfluidics, and sensor technologies have further enabled remarkable progress in the development of in vitro platforms with increased fidelity and efficiency. In this review, we provide an overview of advancements in in vitro cardiac development platforms, with a particular focus on technological innovation. We categorize these platforms into four areas: two-dimensional solid substrate cultures, engineered substrate architectures that enhance cellular functions, cardiac organoids, and embryos/explants-on-chip models. We conclude by addressing current limitations and presenting future perspectives.


Asunto(s)
Evaluación Preclínica de Medicamentos , Corazón , Ingeniería de Tejidos , Humanos , Animales , Evaluación Preclínica de Medicamentos/métodos , Ingeniería de Tejidos/métodos , Organoides/metabolismo , Organoides/citología , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Cardiopatías Congénitas/genética , Dispositivos Laboratorio en un Chip
2.
Arch Toxicol ; 96(2): 389-402, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34973109

RESUMEN

The absence of in vitro platforms for human pulmonary toxicology studies is becoming an increasingly serious concern. The respiratory system has a dynamic mechanical structure that extends from the airways to the alveolar region. In addition, the epithelial, endothelial, stromal, and immune cells are highly organized in each region and interact with each other to function synergistically. These cells of varied lineage, particularly epithelial cells, have been difficult to use for long-term culture in vitro, thus limiting the development of useful experimental tools. This limitation has set a large distance between the bench and the bedside for analyzing the pathogenic mechanisms, the efficacy of candidate therapeutic agents, and the toxicity of compounds. Several researchers have proposed solutions to these problems by reporting on methods for generating human lung epithelial cells derived from pluripotent stem cells (PSCs). Moreover, the use of organoid culture, organ-on-a-chip, and material-based techniques have enabled the maintenance of functional PSC-derived lung epithelial cells as well as primary cells. The aforementioned technological advances have facilitated the in vitro recapitulation of genetic lung diseases and the detection of ameliorating or worsening effects of genetic and chemical interventions, thus indicating the future possibility of more sophisticated preclinical compound assessments in vitro. In this review, we will update the recent advances in lung cell culture methods, principally focusing on human PSC-derived lung epithelial organoid culture systems with the hope of their future application in toxicology studies.


Asunto(s)
Pulmón/citología , Células Madre Pluripotentes/citología , Toxicología/métodos , Animales , Técnicas de Cultivo de Célula , Evaluación Preclínica de Medicamentos/métodos , Evaluación Preclínica de Medicamentos/tendencias , Humanos , Dispositivos Laboratorio en un Chip , Organoides/citología , Toxicología/tendencias
3.
Int J Mol Sci ; 22(14)2021 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-34299287

RESUMEN

Organoids represent one of the most important advancements in the field of stem cells during the past decade. They are three-dimensional in vitro culturing models that originate from self-organizing stem cells and can mimic the in vivo structural and functional specificities of body organs. Organoids have been established from multiple adult tissues as well as pluripotent stem cells and have recently become a powerful tool for studying development and diseases in vitro, drug screening, and host-microbe interaction. The use of stem cells-that have self-renewal capacity to proliferate and differentiate into specialized cell types-for organoids culturing represents a major advancement in biomedical research. Indeed, this new technology has a great potential to be used in a multitude of fields, including cancer research, hereditary and infectious diseases. Nevertheless, organoid culturing is still rife with many challenges, not limited to being costly and time consuming, having variable rates of efficiency in generation and maintenance, genetic stability, and clinical applications. In this review, we aim to provide a synopsis of pluripotent stem cell-derived organoids and their use for disease modeling and other clinical applications.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Técnicas de Cultivo de Órganos/métodos , Organoides/citología , Células Madre Pluripotentes/citología , Animales , Humanos , Modelos Biológicos , Organoides/efectos de los fármacos , Organoides/metabolismo , Células Madre Pluripotentes/efectos de los fármacos , Células Madre Pluripotentes/metabolismo
4.
Nat Commun ; 12(1): 753, 2021 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-33531489

RESUMEN

Cellular models are needed to study human development and disease in vitro, and to screen drugs for toxicity and efficacy. Current approaches are limited in the engineering of functional tissue models with requisite cell densities and heterogeneity to appropriately model cell and tissue behaviors. Here, we develop a bioprinting approach to transfer spheroids into self-healing support hydrogels at high resolution, which enables their patterning and fusion into high-cell density microtissues of prescribed spatial organization. As an example application, we bioprint induced pluripotent stem cell-derived cardiac microtissue models with spatially controlled cardiomyocyte and fibroblast cell ratios to replicate the structural and functional features of scarred cardiac tissue that arise following myocardial infarction, including reduced contractility and irregular electrical activity. The bioprinted in vitro model is combined with functional readouts to probe how various pro-regenerative microRNA treatment regimes influence tissue regeneration and recovery of function as a result of cardiomyocyte proliferation. This method is useful for a range of biomedical applications, including the development of precision models to mimic diseases and the screening of drugs, particularly where high cell densities and heterogeneity are important.


Asunto(s)
Bioimpresión/métodos , Hidrogeles/química , Ingeniería de Tejidos/métodos , Ingeniería Biomédica/métodos , Enfermedades Cardiovasculares , Sistemas de Liberación de Medicamentos/métodos , Evaluación Preclínica de Medicamentos/métodos , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Esferoides Celulares/citología
5.
Gastroenterology ; 160(3): 831-846.e10, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33039464

RESUMEN

BACKGROUND & AIMS: Preclinical identification of compounds at risk of causing drug induced liver injury (DILI) remains a significant challenge in drug development, highlighting a need for a predictive human system to study complicated DILI mechanism and susceptibility to individual drug. Here, we established a human liver organoid (HLO)-based screening model for analyzing DILI pathology at organoid resolution. METHODS: We first developed a reproducible method to generate HLO from storable foregut progenitors from pluripotent stem cell (PSC) lines with reproducible bile transport function. The qRT-PCR and single cell RNA-seq determined hepatocyte transcriptomic state in cells of HLO relative to primary hepatocytes. Histological and ultrastructural analyses were performed to evaluate micro-anatomical architecture. HLO based drug-induced liver injury assays were transformed into a 384 well based high-speed live imaging platform. RESULTS: HLO, generated from 10 different pluripotent stem cell lines, contain polarized immature hepatocytes with bile canaliculi-like architecture, establishing the unidirectional bile acid transport pathway. Single cell RNA-seq profiling identified diverse and zonal hepatocytic populations that in part emulate primary adult hepatocytes. The accumulation of fluorescent bile acid into organoid was impaired by CRISPR-Cas9-based gene editing and transporter inhibitor treatment with BSEP. Furthermore, we successfully developed an organoid based assay with multiplexed readouts measuring viability, cholestatic and/or mitochondrial toxicity with high predictive values for 238 marketed drugs at 4 different concentrations (Sensitivity: 88.7%, Specificity: 88.9%). LoT positively predicts genomic predisposition (CYP2C9∗2) for Bosentan-induced cholestasis. CONCLUSIONS: Liver organoid-based Toxicity screen (LoT) is a potential assay system for liver toxicology studies, facilitating compound optimization, mechanistic study, and precision medicine as well as drug screening applications.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Hepatocitos/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento/métodos , Hígado/efectos de los fármacos , Organoides/efectos de los fármacos , Línea Celular , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Evaluación Preclínica de Medicamentos/métodos , Hepatocitos/patología , Humanos , Hígado/citología , Hígado/patología , Organoides/patología , Células Madre Pluripotentes/citología , Pruebas de Toxicidad Aguda/métodos
6.
J Mol Histol ; 52(1): 87-99, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33179120

RESUMEN

Human pluripotent stem cell (hPSC)-derived cardiomyocytes (CMs) hold great promise for the repair of the injured heart, but optimal cell production in a fully chemically defined and cost-effective system is essential for the efficacy and safety of cell transplantation therapies. In this study, we provided a simple and efficient strategy for cardiac differentiation from hPSCs and performed functional evaluation in a rat model of myocardial infarction. Using a chemically defined medium including four components, recombinant human albumin, ascorbic acid, human transferrin, and RPMI 1640, we developed a manageable and cost-effective protocol for robust generation of CMs from hPSCs. Interestingly, the addition of transferrin helped hPSCs to transit from TeSR-E8 medium to the simple cardiac differentiation medium and successfully initiated mesoderm differentiation without significant cell death. The CM generation efficiency was up to 85% based on cTnT expression. We performed transcriptome profiling from differentiation day 0 to 35, and characterized interesting dynamic change of cardiac genes. CMs derived from transferrin-supplemented simple medium have similar transcriptome and the maturation level compared to those generated in B27 minus insulin medium as well as their in vivo counterparts. Importantly, after transplantation, hPSC-derived CMs survived in the infarcted rat heart, significantly improved the physiological function and reduced fibrosis. Our study offers an easy-to-use and cost-effective method for cardiac differentiation and facilitates the translational application of hPSC-derived CMs for heart repair.


Asunto(s)
Infarto del Miocardio/patología , Infarto del Miocardio/terapia , Miocitos Cardíacos/citología , Células Madre Pluripotentes/citología , Regeneración , Transferrina/farmacología , Animales , Muerte Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/trasplante , Inhibidores de Proteínas Quinasas/farmacología , Ratas , Transcriptoma/genética
7.
Cell Death Differ ; 28(1): 24-34, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33318601

RESUMEN

While inner ear disorders are common, our ability to intervene and recover their sensory function is limited. In vitro models of the inner ear, like the organoid system, could aid in identifying new regenerative drugs and gene therapies. Here, we provide a perspective on the status of in vitro inner ear models and guidance on how to improve their applicability in translational research. We highlight the generation of inner ear cell types from pluripotent stem cells as a particularly promising focus of research. Several exciting recent studies have shown how the developmental signaling cues of embryonic and fetal development can be mimicked to differentiate stem cells into "inner ear organoids" containing otic progenitor cells, hair cells, and neurons. However, current differentiation protocols and our knowledge of embryonic and fetal inner ear development in general, have a bias toward the sensory epithelia of the inner ear. We propose that a more holistic view is needed to better model the inner ear in vitro. Moving forward, attention should be made to the broader diversity of neuroglial and mesenchymal cell types of the inner ear, and how they interact in space or time during development. With improved control of epithelial, neuroglial, and mesenchymal cell fate specification, inner ear organoids would have the ability to truly recapitulate neurosensory function and dysfunction. We conclude by discussing how single-cell atlases of the developing inner ear and technical innovations will be critical tools to advance inner ear organoid platforms for future pre-clinical applications.


Asunto(s)
Diferenciación Celular/fisiología , Oído Interno/citología , Modelos Biológicos , Organoides/citología , Animales , Técnicas de Cultivo de Célula , Células Cultivadas , Oído Interno/crecimiento & desarrollo , Epitelio/fisiología , Células Ciliadas Auditivas Internas/citología , Humanos , Organoides/crecimiento & desarrollo , Células Madre Pluripotentes/citología
8.
Cell Chem Biol ; 27(12): 1561-1572.e7, 2020 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-33125912

RESUMEN

The proliferation of human pancreatic progenitor cells (PPCs) is critical for developing cell therapies for diabetes. Here, using transcriptome analysis combined with small interfering RNA (siRNA) screening, we revealed that WNT7B is a downstream growth factor of AT7867, a compound known to promote the proliferation of PPCs generated from human pluripotent stem cells. Feeder cell lines stably expressing mouse Wnt7a or Wnt7b, but not other Wnts, enhanced PPC proliferation in the absence of AT7867. Importantly, Wnt7a/b ligands did not activate the canonical Wnt pathway, and PPC proliferation depended on the non-canonical Wnt/PKC pathway. A comparison of the phosphoproteome in response to AT7867 or a newly synthesized AT7867 derivative uncovered the function of YY1 as a transcriptional regulator of WNT7B. Overall, our data highlight unknown roles of non-canonical WNT7B/PKC signaling and YY1 in human PPC proliferation and will contribute to the stable supply of a cell source for pancreatic disease modeling and therapeutic applications.


Asunto(s)
Páncreas/citología , Células Madre Pluripotentes/citología , Transducción de Señal , Proteínas Wnt/metabolismo , Factor de Transcripción YY1/metabolismo , Animales , Línea Celular , Proliferación Celular , Células Nutrientes/citología , Humanos , Ratones , Proteína Quinasa C/metabolismo
9.
Biomed Pharmacother ; 131: 110730, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32920519

RESUMEN

Natural products remain a rich source of new drugs, and the search for bioactive molecules from nature continues to play an important role in the development of new medicines. Also, there is increasing use of herbal medicines for the treatment of a plethora of diseases, and demands for more scientific evidence for their efficacy and safety remains a huge challenge. The propensity of stem cells to differentiate into almost every cell type not only holds promise for the delivery of cell-based therapies for currently incurable diseases or a useful tool in studying cell physiology and pathophysiology. Increasingly, stem cells are becoming an important tool in preclinical drug screening and toxicity testing. In this review, we examine the scientific advances made towards the use of pluripotent stem cells as a model for the screening of plant-based medicines. The combination of well-established in vitro electrophysiological and a plethora of toxicogenomic technologies, together with the optimisation of culture methods of herbal plants and pluripotent stem cells can be explored to establish the basis for efficacy, and tissue/organ-based toxicities of many currently used medicinal plants whose efficacies and toxicities remain unknown.


Asunto(s)
Productos Biológicos/farmacología , Descubrimiento de Drogas/métodos , Evaluación Preclínica de Medicamentos/métodos , Plantas Medicinales , Células Madre Pluripotentes/efectos de los fármacos , Productos Biológicos/toxicidad , Plantas Medicinales/toxicidad , Células Madre Pluripotentes/citología
10.
Adv Drug Deliv Rev ; 161-162: 90-109, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32835746

RESUMEN

Chronic lung diseases remain major healthcare burdens, for which the only curative treatment is lung transplantation. In vitro human models are promising platforms for identifying and testing novel compounds to potentially decrease this burden. Directed differentiation of pluripotent stem cells is an important strategy to generate lung cells to create such models. Current lung directed differentiation protocols are limited as they do not 1) recapitulate the diversity of respiratory epithelium, 2) generate consistent or sufficient cell numbers for drug discovery platforms, and 3) establish the histologic tissue-level organization critical for modeling lung function. In this review, we describe how lung development has formed the basis for directed differentiation protocols, and discuss the utility of available protocols for lung epithelial cell generation and drug development. We further highlight tissue engineering strategies for manipulating biophysical signals during directed differentiation such that future protocols can recapitulate both chemical and physical cues present during lung development.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Pulmón/fisiología , Ingeniería de Tejidos/métodos , Animales , Embriología , Humanos , Pulmón/crecimiento & desarrollo , Ratones , Células Madre Pluripotentes/citología , Transducción de Señal/fisiología
11.
Stem Cell Reports ; 15(3): 587-596, 2020 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-32763158

RESUMEN

Current platforms for studying the mechanical properties of human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) as single cells do not measure forces directly, require numerous assumptions, and cannot study cell mechanics at different loading conditions. We present a method for directly measuring the active and passive forces generated by single-cell hPSC-CMs at different stretch levels. Utilizing this technique, single hPSC-CMs exhibited positive length-tension relationship and appropriate inotropic, klinotropic, and lusitropic changes in response to pharmacological treatments (isoproterenol and verapamil). The unique potential of the approach for drug testing and disease modeling was exemplified by doxorubicin and omecamtiv mecarbil drug studies revealing their known actions to suppress (doxorubicin) or augment (omecamtiv mecarbil at low dose) cardiomyocyte contractility, respectively. Finally, mechanistic insights were gained regarding the cellular effects of these drugs as doxorubicin treatment led to cellular mechanical alternans and high doses of omecamtiv mecarbil suppressed contractility and worsened the cellular diastolic properties.


Asunto(s)
Evaluación Preclínica de Medicamentos , Miocitos Cardíacos/citología , Células Madre Pluripotentes/citología , Análisis de la Célula Individual , Fenómenos Biomecánicos/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular , Forma de la Célula/efectos de los fármacos , Doxorrubicina/farmacología , Humanos , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Células Madre Pluripotentes/efectos de los fármacos , Células Madre Pluripotentes/metabolismo , Urea/análogos & derivados , Urea/farmacología
12.
Proc Natl Acad Sci U S A ; 117(33): 19854-19865, 2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32759214

RESUMEN

The blood-retina barrier and blood-brain barrier (BRB/BBB) are selective and semipermeable and are critical for supporting and protecting central nervous system (CNS)-resident cells. Endothelial cells (ECs) within the BRB/BBB are tightly coupled, express high levels of Claudin-5 (CLDN5), a junctional protein that stabilizes ECs, and are important for proper neuronal function. To identify novel CLDN5 regulators (and ultimately EC stabilizers), we generated a CLDN5-P2A-GFP stable cell line from human pluripotent stem cells (hPSCs), directed their differentiation to ECs (CLDN5-GFP hPSC-ECs), and performed flow cytometry-based chemogenomic library screening to measure GFP expression as a surrogate reporter of barrier integrity. Using this approach, we identified 62 unique compounds that activated CLDN5-GFP. Among them were TGF-ß pathway inhibitors, including RepSox. When applied to hPSC-ECs, primary brain ECs, and retinal ECs, RepSox strongly elevated barrier resistance (transendothelial electrical resistance), reduced paracellular permeability (fluorescein isothiocyanate-dextran), and prevented vascular endothelial growth factor A (VEGFA)-induced barrier breakdown in vitro. RepSox also altered vascular patterning in the mouse retina during development when delivered exogenously. To determine the mechanism of action of RepSox, we performed kinome-, transcriptome-, and proteome-profiling and discovered that RepSox inhibited TGF-ß, VEGFA, and inflammatory gene networks. In addition, RepSox not only activated vascular-stabilizing and barrier-establishing Notch and Wnt pathways, but also induced expression of important tight junctions and transporters. Taken together, our data suggest that inhibiting multiple pathways by selected individual small molecules, such as RepSox, may be an effective strategy for the development of better BRB/BBB models and novel EC barrier-inducing therapeutics.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Células Madre Pluripotentes/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Barrera Hematorretinal/efectos de los fármacos , Barrera Hematorretinal/metabolismo , Diferenciación Celular , Línea Celular , Proliferación Celular/efectos de los fármacos , Claudina-5/genética , Claudina-5/metabolismo , Evaluación Preclínica de Medicamentos , Células Endoteliales/citología , Células Endoteliales/metabolismo , Edición Génica , Genoma , Humanos , Ratones , Ratones Noqueados , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Pirazoles/farmacología , Piridinas/farmacología , Uniones Estrechas/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
13.
STAR Protoc ; 1(1)2020 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-32728670

RESUMEN

Human pluripotent stem cells (PSCs) can be differentiated into retinal organoids with proper neural layer organization, yet the protocols are technically challenging and time consuming. We have modified a widely used differentiation protocol by switching all-trans retinoic acid with 9-cis retinal to accelerate photoreceptor differentiation and improve morphogenesis. In this report, we provide a detailed and improved protocol to generate retinal organoids from human pluripotent stem cells. For complete details on the use and execution of this protocol, please refer to Kaya et al. (2019).


Asunto(s)
Técnicas Citológicas , Organoides/citología , Células Madre Pluripotentes/citología , Células Fotorreceptoras Retinianas Bastones/citología , Diterpenos , Humanos , Retinaldehído
14.
Pediatr Surg Int ; 36(1): 1-10, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31555860

RESUMEN

Recent advances in culturing of intestinal stem cells and pluripotent stem cells have led to the development of intestinal organoids. These are self-organizing 3D structures, which recapitulate the characteristics and physiological features of in vivo intestinal epithelium. Intestinal organoids have allowed the development of novel in vitro models to study various gastrointestinal diseases expanding our understanding of the pathophysiology of diseases and leading to the development of innovative therapies. This article aims to summarize the current usage of intestinal organoids as a model of gastrointestinal diseases and the potential applications of intestinal organoids in infants and children. Intestinal organoids allow the study of intestinal epithelium responses to stress factors. Mimicking intestinal injury such as necrotizing enterocolitis, intestinal organoids increases the expression of pro-inflammatory cytokine genes and shows disruption of tight junctions after they are injured by lipopolysaccharide and hypoxia. In cystic fibrosis, intestinal organoids derived from rectal biopsies have provided benefits in genetic studies and development of novel therapeutic gene modulation. Transplantation of intestinal organoids via enema has been shown to rescue damaged colonic epithelium in mice. In addition, tissue-engineered small intestine derived from intestinal organoids have been successfully established providing a potential novel treatment and a new hope for children with short bowel syndrome.


Asunto(s)
Intestinos/citología , Organoides/citología , Atresia Biliar/patología , Atresia Biliar/terapia , Diferenciación Celular , Proliferación Celular , Niño , Fibrosis Quística/terapia , Desarrollo de Medicamentos , Enterocolitis Necrotizante/patología , Terapia Genética , Enfermedad de Hirschsprung/patología , Enfermedad de Hirschsprung/terapia , Humanos , Lactante , Mucosa Intestinal/citología , Hígado/citología , Células Madre Mesenquimatosas/citología , Modelos Biológicos , Células Madre Pluripotentes/citología , Síndrome del Intestino Corto/terapia , Ingeniería de Tejidos
15.
Med Sci (Paris) ; 35(6-7): 549-555, 2019.
Artículo en Francés | MEDLINE | ID: mdl-31274085

RESUMEN

The study of gut diseases is often limited by the access to human biological tissues and animal models that do not faithfully mimic the human pathologies. In this context, the development of intestinal organoids from human pluripotent stem cells is paving the way of gastrointestinal physiology and digestive disease study. In this review, we recall the embryonic development of the digestive tract and its translation to human pluripotent stem cell differentiation. We also present the different types of intestinal organoids that can be generated, as well as their applications in research.


TITLE: Façonner l'intestin à partir des cellules souches pluripotentes humaines. ABSTRACT: L'étude des maladies digestives est parfois limitée par l'accès aux tissus de patients et les modèles précliniques ne sont pas toujours fidèles aux pathologies observées chez l'homme. Dans ce contexte, le développement d'organoïdes intestinaux à partir de cellules souches pluripotentes humaines représente une avancée importante dans l'étude des processus physiologiques et des pathologies digestives. Dans cette revue, nous rappelons les étapes majeures du développement du tractus digestif chez l'homme et décrivons le rationnel de la différenciation dirigée des cellules souches pluripotentes humaines. Nous faisons également un état des lieux sur les différents types d'organoïdes intestinaux existants et leurs applications en recherche fondamentale et préclinique. Enfin, nous discutons des opportunités offertes par les organoïdes intestinaux humains dans un contexte de médecine de précision et de médecine réparatrice.


Asunto(s)
Intestinos/citología , Organoides/citología , Células Madre Pluripotentes/citología , Animales , Diferenciación Celular/fisiología , Células Cultivadas , Evaluación Preclínica de Medicamentos/métodos , Enfermedades Gastrointestinales/patología , Enfermedades Gastrointestinales/terapia , Tracto Gastrointestinal/citología , Tracto Gastrointestinal/crecimiento & desarrollo , Tracto Gastrointestinal/fisiología , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/fisiología , Intestinos/fisiología , Organoides/fisiología , Células Madre Pluripotentes/fisiología , Regeneración/fisiología , Técnicas de Cultivo de Tejidos
16.
Biomaterials ; 216: 119244, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31207406

RESUMEN

During early post-implantation human embryogenesis, the epiblast (EPI) within the blastocyst polarizes to generate a cyst with a central lumen. Cells at the uterine pole of the EPI cyst then undergo differentiation to form the amniotic ectoderm (AM), a tissue essential for further embryonic development. While the causes of early pregnancy failure are complex, improper lumenogenesis or amniogenesis of the EPI represent possible contributing factors. Here we report a novel AM microtissue array platform that allows quantitative phenotyping of lumenogenesis and amniogenesis of the EPI and demonstrate its potential application for embryonic toxicity profiling. Specifically, a human pluripotent stem cell (hPSC)-based amniogenic differentiation protocol was developed using a two-step micropatterning technique to generate a regular AM microtissue array with defined tissue sizes. A computer-assisted analysis pipeline was developed to automatically process imaging data and quantify morphological and biological features of AM microtissues. Analysis of the effects of cell density, cyst size and culture conditions revealed a clear connection between cyst size and amniogenesis of hPSC. Using this platform, we demonstrated that pharmacological inhibition of ROCK signaling, an essential mechanotransductive pathway, suppressed lumenogenesis but did not perturb amniogenic differentiation of hPSC, suggesting uncoupled regulatory mechanisms for AM morphogenesis vs. cytodifferentiation. The AM microtissue array was further applied to screen a panel of clinically relevant drugs, which successfully detected their differential teratogenecity. This work provides a technological platform for toxicological screening of clinically relevant drugs for their effects on lumenogenesis and amniogenesis during early human peri-implantation development, processes that have been previously inaccessible to study.


Asunto(s)
Amnios/citología , Evaluación Preclínica de Medicamentos , Ectodermo/citología , Células Madre Pluripotentes/citología , Análisis de Matrices Tisulares , Amnios/efectos de los fármacos , Amnios/metabolismo , Línea Celular , Evaluación Preclínica de Medicamentos/métodos , Ectodermo/efectos de los fármacos , Ectodermo/metabolismo , Humanos , Células Madre Pluripotentes/efectos de los fármacos , Células Madre Pluripotentes/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Análisis de Matrices Tisulares/métodos , Ingeniería de Tejidos/métodos , Quinasas Asociadas a rho/antagonistas & inhibidores
17.
Future Med Chem ; 11(11): 1305-1322, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31161803

RESUMEN

Most neurodegenerative diseases are characterized by a complex and mostly still unresolved pathology. This fact, together with the lack of reliable disease models, has precluded the development of effective therapies counteracting the disease progression. The advent of human pluripotent stem cells has revolutionized the field allowing the generation of disease-relevant neural cell types that can be used for disease modeling, drug screening and, possibly, cell transplantation purposes. In this Review, we discuss the applications of human pluripotent stem cells, the development of efficient protocols for the derivation of the different neural cells and their applicability for robust in vitro disease modeling and drug screening platforms for most common neurodegenerative conditions.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Enfermedades Neurodegenerativas/tratamiento farmacológico , Neuronas/citología , Células Madre Pluripotentes/citología , Animales , Sistemas CRISPR-Cas , Edición Génica/métodos , Humanos , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Neurogénesis/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/patología
18.
Curr Protoc Stem Cell Biol ; 49(1): e85, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30995363

RESUMEN

The production of human organs inside human-animal interspecies chimeras might one day comprise a viable strategy for generating patient-specific organs, but such experiments will require human chimera-competent pluripotent stem (PS) cells. The stabilization of PS cell self-renewal in serum-free medium and ERK blockade might be critical for capturing primate chimera-competent pluripotency. It has recently been shown that shielding primate cells from the activation of ERK, WNT, and PKC signaling is crucial for deriving African green monkey ERK-independent PS cells. Here, I show that this principle is generalizable to human cells. In this chapter, methods are provided to reset conventional human PS cells to ERK-independence using histone deacetylase inhibitors and PGCX media comprised of N2B27 medium supplemented with LIF, PD0325901, Go6983, CHIR99021, and XAV939. The novel stem cells exhibit higher levels of KLF4 and manifest increased mitochondrial membrane depolarization. However, the author observed that not all PS cell lines are amenable to small molecule-mediated resetting. The ERK-independent PS cells described herein will provide a useful resource for testing interspecies organogenesis strategies. © 2019 by John Wiley & Sons, Inc.


Asunto(s)
Diferenciación Celular/fisiología , Reprogramación Celular/fisiología , Células Madre Pluripotentes/citología , Animales , Quimera/fisiología , Chlorocebus aethiops , Humanos , Factor 4 Similar a Kruppel , Sistema de Señalización de MAP Quinasas/fisiología
19.
Ann Endocrinol (Paris) ; 80(2): 128-133, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30833018

RESUMEN

In the modern world, type-2 diabetes mellitus has become a leading public healthcare problem, due to major risks of morbidity and mortality. Prevalence has increased significantly in recent decades. Treatment involves oral hypoglycemic agents or insulin replacement therapy. Development is ongoing for cell-based diabetes therapies using stem cells with the potential to differentiate into insulin-producing cells (IPCs): embryonic stem cells (ESCs), mesenchymal stem cells (MSCs), induced pluripotent stem cells (iPSCs), and stem cells from adult pancreas, liver, central nervous system, bone marrow and adipose tissue. Successful induction of iPSCs, however, depends on the quantity and quality of available stem cells and the development of adapted protocols determining the environment of extrinsic factors and involvement of small molecules. Validating such new cell therapies must be founded on this experimental rationale.


Asunto(s)
Factores Biológicos/farmacología , Diferenciación Celular/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Células Madre Pluripotentes/efectos de los fármacos , Animales , Factores Biológicos/análisis , Factores Biológicos/aislamiento & purificación , Técnicas de Cultivo de Célula , Reprogramación Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos/métodos , Humanos , Insulina/metabolismo , Secreción de Insulina/efectos de los fármacos , Células Secretoras de Insulina/fisiología , Páncreas/citología , Páncreas/efectos de los fármacos , Páncreas/fisiología , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/fisiología , Bibliotecas de Moléculas Pequeñas/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA