Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.174
Filtrar
1.
Int J Mol Sci ; 25(14)2024 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-39062912

RESUMEN

Knowledge of the molecular mechanisms that underlie the regulation of major adaptive responses to an unbalanced oxygen tension is central to understanding tissue homeostasis and disease. Hypoxia-inducible transcription factors (HIFs) coordinate changes in the transcriptome that control these adaptive responses. Here, we focused on the functional role of the transcriptional repressor basic-helix-loop-helix family member e40 (Bhlhe40), which we previously identified in a meta-analysis as one of the most consistently upregulated genes in response to hypoxia across various cell types. We investigated the role of Bhlhe40 in controlling proliferation and angiogenesis using a gene editing strategy in mouse embryonic stem cells (mESCs) that we differentiated in embryoid bodies (EBs). We observed that hypoxia-induced Bhlhe40 expression was compatible with the rapid proliferation of pluripotent mESCs under low oxygen tension. However, in EBs, hypoxia triggered a Bhlhe40-dependent cell cycle arrest in most progenitor cells and endothelial cells within vascular structures. Furthermore, Bhlhe40 knockout increased the basal vascularization of the EBs in normoxia and exacerbated the hypoxia-induced vascularization, supporting a novel role for Bhlhe40 as a negative regulator of blood vessel formation. Our findings implicate Bhlhe40 in mediating key functional adaptive responses to hypoxia, such as proliferation arrest and angiogenesis.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Hipoxia de la Célula , Proliferación Celular , Cuerpos Embrioides , Células Madre Embrionarias de Ratones , Neovascularización Fisiológica , Animales , Ratones , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Cuerpos Embrioides/metabolismo , Cuerpos Embrioides/citología , Células Madre Embrionarias de Ratones/metabolismo , Células Madre Embrionarias de Ratones/citología , Neovascularización Fisiológica/genética , Diferenciación Celular/genética , Proteínas de Homeodominio/metabolismo , Proteínas de Homeodominio/genética , Células Endoteliales/metabolismo , Angiogénesis
2.
Am J Surg Pathol ; 48(9): 1164-1176, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-38963187

RESUMEN

We investigated the frequency and associated pathology of embryoid bodies in ovarian tumors by evaluating neoplasms in which they are known to occur: 100 immature teratomas, 125 malignant mixed germ cell tumors, and 6 polyembryomas. Three immature teratomas contained a single relatively well-formed embryoid body, whereas these and 11 others showed foci we categorized as embryoid body remnants consisting of microscopic aggregates of embryonal or yolk sac-type epithelium associated with spaces consistent with yolk sac or amniotic cavity but lacking a classic embryoid body structure. Teratomas with these foci were all high grade. A well-formed embryoid body was found in only 1 malignant mixed tumor, but embryoid body remnants were present in 25%, invariably associated with foci of immature teratoma (100%) and often with yolk sac tumor (97%), embryonal carcinoma (35%), or both (32%). These foci usually took the form of round to oval aggregates, often well-circumscribed, for which the term "polyembryoma background" has been proposed. The polyembryomas were typically grossly hemorrhagic and occurred in patients from 9 to 43 years of age. The embryoid bodies in them generally grew in lobules within an edematous to occasionally myxoid stroma. Four tumors contained liver-like cells, 4 numerous glands likely recapitulating the allantois, 3 syncytiotrophoblast cells, 2 prominent cysts, and 2 striking vascular proliferations. This study indicates that (1) typical embryoid bodies are rare in immature teratomas but about 14% of them have embryoid body remnants. (2) Embryoid body remnants are seen in 25% of malignant mixed germ cell tumors with a teratomatous component and often proliferate to form yolk sac tumor and embryonal carcinoma. (3) Well-formed embryoid bodies growing in a confluent manner (polyembryoma) are rare, and minor foci of teratoma, yolk sac tumor, or embryonal carcinoma are almost always present, indicating that these are fundamentally malignant mixed germ cell tumors but the polyembryoma component is dominant and distinctive which, in our opinion, justifies its own nomenclature. (4) Embryoid bodies are not a feature of other germ cell tumors.


Asunto(s)
Proliferación Celular , Cuerpos Embrioides , Neoplasias de Células Germinales y Embrionarias , Neoplasias Ováricas , Teratoma , Neoplasias Ováricas/patología , Femenino , Humanos , Neoplasias de Células Germinales y Embrionarias/patología , Cuerpos Embrioides/patología , Adolescente , Adulto , Niño , Teratoma/patología , Adulto Joven , Persona de Mediana Edad , Tumor del Seno Endodérmico/patología , Anciano
3.
Development ; 151(13)2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38940293

RESUMEN

Generation of hematopoietic stem and progenitor cells (HSPCs) ex vivo and in vivo, especially the generation of safe therapeutic HSPCs, still remains inefficient. In this study, we have identified compound BF170 hydrochloride as a previously unreported pro-hematopoiesis molecule, using the differentiation assays of primary zebrafish blastomere cell culture and mouse embryoid bodies (EBs), and we demonstrate that BF170 hydrochloride promoted definitive hematopoiesis in vivo. During zebrafish definitive hematopoiesis, BF170 hydrochloride increases blood flow, expands hemogenic endothelium (HE) cells and promotes HSPC emergence. Mechanistically, the primary cilia-Ca2+-Notch/NO signaling pathway, which is downstream of the blood flow, mediated the effects of BF170 hydrochloride on HSPC induction in vivo. Our findings, for the first time, reveal that BF170 hydrochloride is a compound that enhances HSPC induction and may be applied to the ex vivo expansion of HSPCs.


Asunto(s)
Diferenciación Celular , Hematopoyesis , Células Madre Hematopoyéticas , Pez Cebra , Animales , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Ratones , Diferenciación Celular/efectos de los fármacos , Hematopoyesis/efectos de los fármacos , Receptores Notch/metabolismo , Transducción de Señal/efectos de los fármacos , Cuerpos Embrioides/citología , Cuerpos Embrioides/efectos de los fármacos , Cuerpos Embrioides/metabolismo , Cilios/metabolismo , Cilios/efectos de los fármacos , Blastómeros/citología , Blastómeros/metabolismo , Blastómeros/efectos de los fármacos , Células Cultivadas
4.
Methods Mol Biol ; 2794: 157-167, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38630227

RESUMEN

There is a high demand for the development of in vitro models for human brain development and diseases due to the inaccessibility of human brain tissues. The human iPSC-derived brain organoids provide a promising in vitro model for studying human brain development and disorders. However, it is challenging to generate a large number of brain organoids with high consistency for modeling human neurological diseases. Here, we describe a method for generating high-yield brain organoids with high consistency by combining large-scale embryoid body (EB) generation and incorporating a quality control screening step during differentiation. The method described in this chapter provides a robust way to generate brain organoids for studying human brain development and modeling neurological diseases.


Asunto(s)
Células Madre Pluripotentes Inducidas , Humanos , Encéfalo , Cuerpos Embrioides , Organoides , Control de Calidad
5.
Int J Mol Sci ; 25(3)2024 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-38338722

RESUMEN

Within the last decade, a wide variety of protocols have emerged for the generation of retinal organoids. A subset of studies have compared protocols based on stem cell source, the physical features of the microenvironment, and both internal and external signals, all features that influence embryoid body and retinal organoid formation. Most of these comparisons have focused on the effect of signaling pathways on retinal organoid development. In this study, our aim is to understand whether starting cell conditions, specifically those involved in embryoid body formation, affect the development of retinal organoids in terms of differentiation capacity and reproducibility. To investigate this, we used the popular 3D floating culture method to generate retinal organoids from stem cells. This method starts with either small clumps of stem cells generated from larger clones (clumps protocol, CP) or with an aggregation of single cells (single cells protocol, SCP). Using histological analysis and gene-expression comparison, we found a retention of the pluripotency capacity on embryoid bodies generated through the SCP compared to the CP. Nonetheless, these early developmental differences seem not to impact the final retinal organoid formation, suggesting a potential compensatory mechanism during the neurosphere stage. This study not only facilitates an in-depth exploration of embryoid body development but also provides valuable insights for the selection of the most suitable protocol in order to study retinal development and to model inherited retinal disorders in vitro.


Asunto(s)
Cuerpos Embrioides , Retina , Reproducibilidad de los Resultados , Retina/metabolismo , Organoides , Diferenciación Celular
6.
Arch Toxicol ; 98(4): 1209-1224, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38311648

RESUMEN

To meet the growing demand for developmental toxicity assessment of chemicals, New Approach Methodologies (NAMs) are needed. Previously, we developed two 3D in vitro assays based on human-induced pluripotent stem cells (hiPSC) and cardiomyocyte differentiation: the PluriBeat assay, based on assessment of beating differentiated embryoid bodies, and the PluriLum assay, a reporter gene assay based on the expression of the early cardiac marker NKX2.5; both promising assays for predicting embryotoxic effects of chemicals and drugs. In this work, we aimed to further describe the predictive power of the PluriLum assay and compare its sensitivity with PluriBeat and similar human stem cell-based assays developed by others. For this purpose, we assessed the toxicity of a panel of ten chemicals from different chemical classes, consisting of the known developmental toxicants 5-fluorouracil, all-trans retinoic acid and valproic acid, as well as the negative control compounds ascorbic acid and folic acid. In addition, the fungicides epoxiconazole and prochloraz, and three perfluoroalkyl substances (PFAS), PFOS, PFOA and GenX were tested. Generally, the PluriLum assay displayed higher sensitivity when compared to the PluriBeat assay. For several compounds the luminescence readout of the PluriLum assay showed effects not detected by the PluriBeat assay, including two PFAS compounds and the two fungicides. Overall, we find that the PluriLum assay has the potential to provide a fast and objective detection of developmental toxicants and has a level of sensitivity that is comparable to or higher than other in vitro assays also based on human stem cells and cardiomyocyte differentiation for assessment of developmental toxicity.


Asunto(s)
Fluorocarburos , Fungicidas Industriales , Células Madre Pluripotentes Inducidas , Humanos , Pruebas de Toxicidad/métodos , Cuerpos Embrioides , Diferenciación Celular , Sustancias Peligrosas
7.
J Vet Med Sci ; 86(3): 247-257, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38171744

RESUMEN

Canine induced pluripotent stem cells (ciPSCs) can provide useful insights into novel therapies in both veterinary and medical fields. However, limited accessibility to the present culture medium and requirement of considerable time, effort, and cost for routine ciPSC maintenance restrict advancement in ciPSC research. In addition, it is unknown whether ciPSC culture conditions influence differentiation propensity. We investigated the availability of the common human pluripotent stem cells (hPSCs) culture systems for ciPSC maintenance and the differentiation propensities of the ciPSCs maintained in these culture systems. StemFlex and mTeSR Plus supported PSC-like colony formation and pluripotency markers expression in ciPSCs even after five passages. Additionally, ciPSCs were maintained under weekend-free culture conditions with a stable growth rate, pluripotency marker expression, and differentiation abilities using vitronectin (VTN-N) and Geltrex. Following maintenance of spontaneously differentiated ciPSCs under various conditions by embryoid body formation, there were few differences in the differentiation propensities of ciPSCs among the tested culture conditions. Thus, ciPSCs were successfully cultured under weekend-free conditions for ciPSC maintenance using StemFlex or mTeSR Plus with VTN-N or Geltrex. The present study offers simpler and more effort-, time-, and cost-saving options for ciPSC culture systems, which may lead to further development in research using ciPSCs.


Asunto(s)
Células Madre Pluripotentes Inducidas , Células Madre Pluripotentes , Animales , Perros , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes/metabolismo , Diferenciación Celular , Cuerpos Embrioides
8.
STAR Protoc ; 5(1): 102827, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38219151

RESUMEN

Here, we present a protocol to differentiate induced pluripotent stem cell (iPSC) into adherent hematopoietic progenitors that release floating CD14+ CD45+ monocytes into the culture medium. We describe steps for iPSC expansion, embryoid body (EB) formation, suspension culture, plating EBs, and recurring harvests of monocytes, a.k.a. "monocyte factory." We then describe detailed procedures for freezing/thawing of monocytes and differentiation into polarized M1 and M2 macrophages. This protocol provides foundation to study iPSC monocytes and their progenies such as macrophages, microglial, and dendritic cells. For complete details on the use and execution of this protocol, please refer to Karlson et al.1 and Panicker et al.2.


Asunto(s)
Células Madre Pluripotentes Inducidas , Monocitos , Humanos , Macrófagos , Diferenciación Celular , Cuerpos Embrioides
9.
In Vitro Cell Dev Biol Anim ; 60(5): 521-534, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38169039

RESUMEN

Trisomy 12 is one of the most frequent chromosomal abnormalities in cultured human pluripotent stem cells (hPSCs). Although potential oncogenic properties and augmented cell cycle caused by trisomy 12 have been reported, the consequences of trisomy 12 in terms of cell differentiation, which is the basis for regenerative medicine, drug development, and developmental biology studies, have not yet been investigated. Here, we report that trisomy 12 compromises the mesendodermal differentiation of hPSCs. We identified sublines of hPSCs carrying trisomy 12 after their prolonged culture. Transcriptome analysis revealed that these hPSC sublines carried abnormal gene expression patterns in specific signaling pathways in addition to cancer-related cell cycle pathways. These hPSC sublines showed a lower propensity for mesendodermal differentiation in embryoid bodies cultured in a serum-free medium. BMP4-induced exit from the self-renewal state was impaired in the trisomy 12 hPSC sublines, with less upregulation of key transcription factor gene expression. As a consequence, the differentiation efficiency of hematopoietic and hepatic lineages was also impaired in the trisomy 12 hPSC sublines. We reveal that trisomy 12 disrupts the genome-wide expression patterns that are required for proper mesendodermal differentiation.


Asunto(s)
Diferenciación Celular , Cromosomas Humanos Par 12 , Células Madre Pluripotentes , Trisomía , Humanos , Diferenciación Celular/genética , Trisomía/genética , Cromosomas Humanos Par 12/genética , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/citología , Mesodermo/citología , Endodermo/citología , Endodermo/metabolismo , Proteína Morfogenética Ósea 4/metabolismo , Proteína Morfogenética Ósea 4/genética , Cuerpos Embrioides/citología , Cuerpos Embrioides/metabolismo , Línea Celular , Transducción de Señal/genética
10.
Methods Mol Biol ; 2753: 217-230, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38285341

RESUMEN

Animal-based test systems have traditionally been used to screen for the potential teratogenic activity of drugs. Still, their deficits in predicting precise human-specific outcomes and ethical concerns have led to a need for alternative approaches. In vitro, teratogenicity testing using cell cultures or other in vitro systems is a potential alternative. Of the different in vitro platforms, the mouse embryonic stem cell test (mEST) is currently the most widely used and validated in vitro test for assessing the potential effects of teratogens on early embryonic development. The mEST involves exposing mouse embryonic stem cells to the test compound and monitoring their differentiation for several days.Nevertheless, its predictive ability was comparatively lower when distinguishing weak developmental toxicants from non-toxic substances. Since then, several modifications and adaptations of the mEST protocol have been developed. This chapter describes an alternative method based on molecular approaches to predict embryotoxicity. This method, originated from the mEST, analyzes the expression of differentiation genes involved in the development of mesoderm, endoderm, and stoderm and allows screening embryo-toxicants with different mechanisms of action. The hanging drops embryoid bodies used in the original mEST protocol have been replaced with monolayer culture, and thus the process has been shortened. In general, the method shows higher predictability compared with the traditional ones.


Asunto(s)
Células Madre Embrionarias , Teratogénesis , Femenino , Embarazo , Humanos , Animales , Ratones , Células Madre Embrionarias de Ratones , Teratógenos/toxicidad , Cuerpos Embrioides , Sustancias Peligrosas
11.
Cell Prolif ; 57(6): e13595, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38185785

RESUMEN

Basement membrane (BM) component deposition is closely linked to the establishment of cell polarity. Previously, we showed that Prickle1 is crucial for BM deposition and cell polarity events in tear duct elongation. To gain a deeper understanding of the intimate relationship between BM formation and cell polarity, we generated induced pluripotent stem cells (iPSCs)-derived embryoid bodies (EBs) with a basement membrane separating the visceral endoderm (VE) and inner EB cell mass. We found that Prickle1 was highly expressed in VE of the normal EBs, and the Prickle1 mutant EBs displayed severely impaired BM. Notably, the formation of the basement membrane appeared to rely on the proper microtubule network of the VE cells, which was disrupted in the Prickle1 mutant EBs. Moreover, disruption of vesicle trafficking in the VE hindered BM secretion. Furthermore, reintroducing Prickle1 in the mutant EBs completely rescued BM formation but not the apicobasal cell polarity of the VE. Our data, in conjunction with studies by others, highlight the conserved role of Prickle1 in directing the secretion of BM components of the VE cells during embryonic germ layer differentiation, even in the absence of established general polarity machinery. Our study introduces a novel system based on iPSCs-derived EBs for investigating cellular and molecular events associated with cell polarity.


Asunto(s)
Membrana Basal , Diferenciación Celular , Polaridad Celular , Cuerpos Embrioides , Células Madre Pluripotentes Inducidas , Proteínas con Dominio LIM , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/citología , Cuerpos Embrioides/metabolismo , Cuerpos Embrioides/citología , Membrana Basal/metabolismo , Proteínas con Dominio LIM/metabolismo , Proteínas con Dominio LIM/genética , Animales , Ratones , Endodermo/metabolismo , Endodermo/citología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética
12.
Nature ; 626(7998): 367-376, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38092041

RESUMEN

Implantation of the human embryo begins a critical developmental stage that comprises profound events including axis formation, gastrulation and the emergence of haematopoietic system1,2. Our mechanistic knowledge of this window of human life remains limited due to restricted access to in vivo samples for both technical and ethical reasons3-5. Stem cell models of human embryo have emerged to help unlock the mysteries of this stage6-16. Here we present a genetically inducible stem cell-derived embryoid model of early post-implantation human embryogenesis that captures the reciprocal codevelopment of embryonic tissue and the extra-embryonic endoderm and mesoderm niche with early haematopoiesis. This model is produced from induced pluripotent stem cells and shows unanticipated self-organizing cellular programmes similar to those that occur in embryogenesis, including the formation of amniotic cavity and bilaminar disc morphologies as well as the generation of an anterior hypoblast pole and posterior domain. The extra-embryonic layer in these embryoids lacks trophoblast and shows advanced multilineage yolk sac tissue-like morphogenesis that harbours a process similar to distinct waves of haematopoiesis, including the emergence of erythroid-, megakaryocyte-, myeloid- and lymphoid-like cells. This model presents an easy-to-use, high-throughput, reproducible and scalable platform to probe multifaceted aspects of human development and blood formation at the early post-implantation stage. It will provide a tractable human-based model for drug testing and disease modelling.


Asunto(s)
Desarrollo Embrionario , Estratos Germinativos , Hematopoyesis , Saco Vitelino , Humanos , Implantación del Embrión , Endodermo/citología , Endodermo/embriología , Estratos Germinativos/citología , Estratos Germinativos/embriología , Saco Vitelino/citología , Saco Vitelino/embriología , Mesodermo/citología , Mesodermo/embriología , Células Madre Pluripotentes Inducidas/citología , Amnios/citología , Amnios/embriología , Cuerpos Embrioides/citología , Linaje de la Célula , Biología Evolutiva/métodos , Biología Evolutiva/tendencias
13.
Curr Protoc ; 3(12): e948, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38148714

RESUMEN

The patterning of excitatory cortical neurons from human pluripotent stem cells (hPSCs) is a desired technique for the study of neurodevelopmental disorders, as neurons can be created and compared from control hPSC lines, hPSC lines generated from patients, and CRISPR-modified hPSC lines. Therefore, this technique allows for the examination of disease phenotypes and assists in the development of potential new therapeutics for neurodevelopmental disorders. Many protocols, however, are optimized for use with specific hPSC lines or within a single laboratory, and they often provide insufficient guidance on how to identify positive stages in the differentiation or how to troubleshoot. Here, we present an efficient and reproducible directed differentiation protocol to generate two-dimensional cultures of hPSC-derived excitatory cortical neurons without intermediary embryoid body formation. This novel protocol is supported by our data generated with five independent hPSC lines and in two independent laboratories. Importantly, as neuronal differentiations follow a long time course to reach maturity, we provide extensive guidance regarding morphological and flow cytometry checkpoints allowing for early indications of successful differentiation. We also include extensive troubleshooting tips and support protocols to assist the operator. The goal of this protocol is to assist others in the successful differentiation of excitatory cortical neurons from hPSCs. © 2023 Wiley Periodicals LLC. Basic Protocol: Directed differentiation of hPSCs into excitatory cortical neurons Support Protocol 1: Harvesting and fixing cells for flow cytometry analyses Support Protocol 2: Performing flow cytometry analyses Support Protocol 3: Thawing NPCs from a cryopreserved stock Alternate Protocol 1: Continuing Expansion of NPCs Alternate Protocol 2: Treatment of neurons with Ara-C to ablate radial glia Support Protocol 4: Experimental methods for validation of excitatory cortical neurons.


Asunto(s)
Técnicas de Cultivo de Célula , Células Madre Pluripotentes , Humanos , Técnicas de Cultivo de Célula/métodos , Células Madre Pluripotentes/fisiología , Neuronas/fisiología , Diferenciación Celular/fisiología , Cuerpos Embrioides
14.
Stem Cell Res Ther ; 14(1): 371, 2023 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-38110996

RESUMEN

BACKGROUND: Morbidity and mortality associated with cardiovascular diseases, such as myocardial infarction, stem from the inability of terminally differentiated cardiomyocytes to regenerate, and thus repair the damaged myocardial tissue structure. The molecular biological mechanisms behind the lack of regenerative capacity for those cardiomyocytes remains to be fully elucidated. Recent studies have shown that c-Jun serves as a cell cycle regulator for somatic cell fates, playing a key role in multiple molecular pathways, including the inhibition of cellular reprogramming, promoting angiogenesis, and aggravation of cardiac hypertrophy, but its role in cardiac development is largely unknown. This study aims to delineate the role of c-Jun in promoting early-stage cardiac differentiation. METHODS: The c-Jun gene in mouse embryonic stem cells (mESCs) was knocked out with CRISPR-Cas9, and the hanging drop method used to prepare the resulting embryoid bodies. Cardiac differentiation was evaluated up to 9 days after c-Jun knockout (ko) via immunofluorescence, flow cytometric, and qPCR analyses. RESULTS: Compared to the wild-type control group, obvious beating was observed among the c-Jun-ko mESCs after 6 days, which was also associated with significant increases in myocardial marker expression. Additionally, markers associated with mesoderm and endoderm cell layer development, essential for further differentiation of ESCs into cardiomyocytes, were also up-regulated in the c-Jun-ko cell group. CONCLUSIONS: Knocking out c-Jun directs ESCs toward a meso-endodermal cell lineage fate, in turn leading to generation of beating myocardial cells. Thus, c-Jun plays an important role in regulating early cardiac cell development.


Asunto(s)
Cuerpos Embrioides , Miocitos Cardíacos , Proteínas Proto-Oncogénicas c-jun , Animales , Ratones , Diferenciación Celular , Linaje de la Célula , Células Madre Embrionarias de Ratones , Miocardio , Miocitos Cardíacos/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo
15.
J Vis Exp ; (200)2023 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-37929996

RESUMEN

Mesenchymal stromal cells (MSCs) are adult pluripotent stem cells which have been widely used in regenerative medicine. As somatic tissue-derived MSCs are restricted by limited donation, quality variations, and biosafety, the past 10 years have seen a great rise in efforts to generate MSCs from human induced pluripotent stem cells (hiPSCs). Past and recent efforts in the differentiation of hiPSCs into MSCs have been centered around two culture methodologies: (1) the formation of embryoid bodies (EBs) and (2) the use of monolayer culture. This protocol describes these two representative methods in deriving MSC from hiPSCs. Each method presents its advantages and disadvantages, including time, cost, cell proliferation ability, the expression of MSC markers, and their capability of differentiation in vitro. This protocol demonstrates that both methods can derive mature and functional MSCs from hiPSCs. The monolayer method is characterized by lower cost, simpler operation, and easier osteogenic differentiation, while the EB method is characterized by lower time consumption.


Asunto(s)
Células Madre Pluripotentes Inducidas , Células Madre Mesenquimatosas , Adulto , Humanos , Osteogénesis , Diferenciación Celular , Cuerpos Embrioides
16.
Biochem Biophys Res Commun ; 686: 149197, 2023 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-37924668

RESUMEN

Various extracellular matrix (ECM) in the lungs regulate tissue development and homeostasis, as well as provide support for cell structures. However, few studies regarding the effects of lung cell differentiation using lung-derived ECM (LM) alone have been reported. The present study investigated the capability of lung-derived matrix sheets (LMSs) to induce lung cell differentiation using mouse embryonic stem (ES) cells. Expressions of lung-related cell markers were significantly upregulated in ES-derived embryoid bodies (EBs) cultured on an LMS for two weeks. Moreover, immunohistochemical analysis of EBs grown on LMSs revealed differentiation of various lung-related cells. These results suggest that an LMS can be used to promote differentiation of stem cells into lung cells.


Asunto(s)
Cuerpos Embrioides , Células Madre Embrionarias , Animales , Ratones , Diferenciación Celular/fisiología , Células Cultivadas , Pulmón
17.
Biofabrication ; 16(1)2023 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-37972398

RESUMEN

Embryoid bodies (EBs) and self-organizing organoids derived from human pluripotent stem cells (hPSCs) recapitulate tissue development in a dish and hold great promise for disease modeling and drug development. However, current protocols are hampered by cellular stress and apoptosis during cell aggregation, resulting in variability and impaired cell differentiation. Here, we demonstrate that EBs and various organoid models (e.g., brain, gut, kidney) can be optimized by using the small molecule cocktail named CEPT (chroman 1, emricasan, polyamines, trans-ISRIB), a polypharmacological approach that ensures cytoprotection and cell survival. Application of CEPT for just 24 h during cell aggregation has long-lasting consequences affecting morphogenesis, gene expression, cellular differentiation, and organoid function. Various qualification methods confirmed that CEPT treatment enhanced experimental reproducibility and consistently improved EB and organoid fitness as compared to the widely used ROCK inhibitor Y-27632. Collectively, we discovered that stress-free cell aggregation and superior cell survival in the presence of CEPT are critical quality control determinants that establish a robust foundation for bioengineering complex tissue and organ models.


Asunto(s)
Cuerpos Embrioides , Células Madre Pluripotentes , Humanos , Cuerpos Embrioides/metabolismo , Reproducibilidad de los Resultados , Organoides , Diferenciación Celular
18.
Rev. bioét. derecho ; (59): 5-29, Nov. 2023. tab
Artículo en Español | IBECS | ID: ibc-226611

RESUMEN

En los últimos años el desarrollo de modelos in vitro con células madre humanas que simulan el desarrollo embrionario temprano ha vivido un gran progreso. Las dificultades para acceder a embriones humanos, la escasez de material embrionario y los desafíos técnicos, legales y éticos existentes sobre la investigación y experimentación con embriones humanos in vitro siguen siendo una barrera para avanzar en el conocimiento de la embriogénesis tras la gastrulación. Por ello distintos mecanismos celulares subyacentes a la formación de las líneas celulares en los seres humanos siguen siendo desconocidos. En el presente trabajo intentaremos reflejar varios de los aspectos que son motivo de incertidumbres jurídicas internacionales en relación con la investigación con embrioides como modelo experimental.(AU)


En els últims anys el desenvolupament de models in vitroamb cèl·lules mare humanes que simulen el desenvolupament embrionari primerenc ha viscut un gran progrés. Les dificultats per a accedir a embrions humans, l'escassetat de material embrionari i els desafiaments tècnics, legals i ètics existents sobre la recerca i experimentació amb embrions humans in vitrocontinuen sent una barrera per a avançar en el coneixement de la embriogènesi després de la gastrulació. Per això diferents mecanismes cel·lulars subjacents a la formació de les línies cel·lulars en els éssers humans continuen sent desconeguts. En el present treball intentaré reflectir diversos dels aspectes que són motiu d'incerteses jurídiques internacionals en relació amb la recerca amb embrioides com a model experimental.(AU)


In recent years, the development of in vitromodels with human stem cells that simulate early embryonic development has experienced great progress. The difficulties in accessing human embryos, the scarcity of embryonic material, and the existing technical, legal, and ethical challenges regarding research and experimentation with in vitrohuman embryos still represents a barrier to advancing in the knowledge of post-gastrulation embryogenesis. Therefore, different cellular mechanisms underlying the formation of cell lines in humans remain unknown. In the present work Iwill try to reflect several of the aspects that are the cause of internationallegal uncertainties in relation to research with embryoids as an experimental model.(AU)


Asunto(s)
Humanos , Cuerpos Embrioides , Células Madre , Técnicas In Vitro , Investigaciones con Embriones , Desarrollo Embrionario , Bioética , Discusiones Bioéticas , Fertilización In Vitro , Fertilización
19.
Nat Commun ; 14(1): 6685, 2023 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-37865642

RESUMEN

Organoids are engineered 3D miniature tissues that are defined by their organ-like structures, which drive a fundamental understanding of human development. However, current organoid generation methods are associated with low production throughputs and poor control over size and function including due to organoid merging, which limits their clinical and industrial translation. Here, we present a microfluidic platform for the mass production of lumenogenic embryoid bodies and functional cardiospheres. Specifically, we apply triple-jet in-air microfluidics for the ultra-high-throughput generation of hollow, thin-shelled, hydrogel microcapsules that can act as spheroid-forming bioreactors in a cytocompatible, oil-free, surfactant-free, and size-controlled manner. Uniquely, we show that microcapsules generated by in-air microfluidics provide a lumenogenic microenvironment with near 100% efficient cavitation of spheroids. We demonstrate that upon chemical stimulation, human pluripotent stem cell-derived spheroids undergo cardiomyogenic differentiation, effectively resulting in the mass production of homogeneous and functional cardiospheres that are responsive to external electrical stimulation. These findings drive clinical and industrial adaption of stem cell technology in tissue engineering and drug testing.


Asunto(s)
Cuerpos Embrioides , Células Madre Pluripotentes , Humanos , Cápsulas , Ingeniería de Tejidos/métodos , Organoides , Esferoides Celulares
20.
Lab Chip ; 23(20): 4378-4389, 2023 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-37695312

RESUMEN

Embryonic stem cell (ESC)-derived aggregates, called embryoid bodies (EBs), are powerful in vitro models used to study human development and disease. However, the cost-effective and large-scale production of homogeneous EBs still remains a challenge. Here, we report a rapid, straightforward method for fabricating closely arrayed deep concave microwells, enabling the mass production of uniform EBs from single cell suspensions. By simply combining micromilling, caramel replica molding, and thermal reflow, we generate convex micromolds with high aspect ratios and excellent surface smoothness. Benefitting from the nature of reflow, this method can produce rounded bottom polydimethylsiloxane (PDMS) microwells, which are not easily achieved with standard soft lithography techniques but critical to producing spherical EBs. To achieve optimal concave microwells, we investigated the effect of thermal reflow temperature and time on the surface smoothness and roundness of the finished microwells. In addition, to further improve the utility of this method, we also investigated the effect of microwell aspect ratio (AR) on the loss of EBs during medium manipulation. The capability of this deep concave microwell system was validated by rapidly generating a large number of human embryonic stem cell (hESC)-derived EBs and then efficiently differentiating them into a cardiac lineage. The proposed fabrication method and deep concave microwell platform are highly practical, and thus will benefit the mass production of EBs for potential tissue regeneration and cell therapy applications.


Asunto(s)
Cuerpos Embrioides , Células Madre Embrionarias , Humanos , Corazón , Diferenciación Celular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA