Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 110
Filtrar
1.
Dev Biol ; 478: 102-121, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34181916

RESUMO

Human organoids stand at the forefront of basic and translational research, providing experimentally tractable systems to study human development and disease. These stem cell-derived, in vitro cultures can generate a multitude of tissue and organ types, including distinct brain regions and sensory systems. Neural organoid systems have provided fundamental insights into molecular mechanisms governing cell fate specification and neural circuit assembly and serve as promising tools for drug discovery and understanding disease pathogenesis. In this review, we discuss several human neural organoid systems, how they are generated, advances in 3D imaging and bioengineering, and the impact of organoid studies on our understanding of the human nervous system.


Assuntos
Encefalopatias , Encéfalo , Organoides , Retina , Encéfalo/citologia , Encéfalo/embriologia , Encéfalo/crescimento & desenvolvimento , Diferenciação Celular , Biologia do Desenvolvimento/métodos , Corpos Embrioides/fisiologia , Indução Embrionária , Humanos , Células-Tronco Neurais/fisiologia , Neurobiologia/métodos , Neurogênese , Retina/citologia , Retina/embriologia , Retina/crescimento & desenvolvimento , Técnicas de Cultura de Tecidos
3.
Stem Cell Reports ; 16(5): 1039-1050, 2021 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-33979592

RESUMO

Embryonic development has been traditionally seen as an inductive process directed by exogenous maternal inputs and extra-embryonic signals. Increasing evidence, however, is showing that, in addition to exogenous signals, the development of the embryo involves endogenous self-organization. Recently, this self-organizing potential has been highlighted by a number of stem cell models known as embryoids that can recapitulate different aspects of embryogenesis in vitro. Here, we review the self-organizing behaviors observed in different embryoid models and seek to reconcile this new evidence with classical knowledge of developmental biology. This analysis leads to reexamine embryonic development as a guided self-organizing process, where patterning and morphogenesis are controlled by a combination of exogenous signals and endogenous self-organization. Finally, we discuss the multidisciplinary approach required to investigate the genetic and cellular basis of self-organization.


Assuntos
Embrião de Mamíferos/fisiologia , Corpos Embrioides/fisiologia , Desenvolvimento Embrionário , Animais , Bioengenharia , Células-Tronco Embrionárias/metabolismo , Camadas Germinativas/citologia , Humanos
4.
J Vis Exp ; (169)2021 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-33749682

RESUMO

Organoids provide a promising platform to study disease mechanism and treatments, directly in the context of human tissue with the versatility and throughput of cell culture. Mature human retinal organoids are utilized to screen potential pharmaceutical treatments for the age-related retinal degenerative disease macular telangiectasia type 2 (MacTel). We have recently shown that MacTel can be caused by elevated levels of an atypical lipid species, deoxysphingolipids (deoxySLs). These lipids are toxic to the retina and may drive the photoreceptor loss that occurs in MacTel patients. To screen drugs for their ability to prevent deoxySL photoreceptor toxicity, we generated human retinal organoids from a non-MacTel induced pluripotent stem cell (iPSC) line and matured them to a post-mitotic age where they develop all of the neuronal lineage-derived cells of the retina, including functionally mature photoreceptors. The retinal organoids were treated with a deoxySL metabolite and apoptosis was measured within the photoreceptor layer using immunohistochemistry. Using this toxicity model, pharmacological compounds that prevent deoxySL-induced photoreceptor death were screened. Using a targeted candidate approach, we determined that fenofibrate, a drug commonly prescribed for the treatment of high cholesterol and triglycerides, can also prevent deoxySL toxicity in the cells of the retina. The toxicity screen successfully identified an FDA-approved drug that can prevent photoreceptor death. This is a directly actionable finding owing to the highly disease-relevant model tested. This platform can be easily modified to test any number of metabolic stressors and potential pharmacological interventions for future treatment discovery in retinal diseases.


Assuntos
Descoberta de Drogas , Organoides/fisiologia , Retina/fisiologia , Testes de Toxicidade , Apoptose/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Corpos Embrioides/efeitos dos fármacos , Corpos Embrioides/fisiologia , Fenofibrato/toxicidade , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Organoides/efeitos dos fármacos , Células Fotorreceptoras/efeitos dos fármacos , Células Fotorreceptoras/metabolismo , Retina/citologia , Retina/efeitos dos fármacos , Esfingosina/análogos & derivados , Esfingosina/toxicidade
5.
Arch Toxicol ; 94(11): 3831-3846, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32700165

RESUMO

There is a great need for novel in vitro methods to predict human developmental toxicity to comply with the 3R principles and to improve human safety. Human-induced pluripotent stem cells (hiPSC) are ideal for the development of such methods, because they are easy to retrieve by conversion of adult somatic cells and can differentiate into most cell types of the body. Advanced three-dimensional (3D) cultures of these cells, so-called embryoid bodies (EBs), moreover mimic the early developing embryo. We took advantage of this to develop a novel human toxicity assay to predict chemically induced developmental toxicity, which we termed the PluriBeat assay. We employed three different hiPSC lines from male and female donors and a robust microtiter plate-based method to produce EBs. We differentiated the cells into cardiomyocytes and introduced a scoring system for a quantitative readout of the assay-cardiomyocyte contractions in the EBs observed on day 7. Finally, we tested the three compounds thalidomide (2.3-36 µM), valproic acid (25-300 µM), and epoxiconazole (1.3-20 µM) on beating and size of the EBs. We were able to detect the human-specific teratogenicity of thalidomide and found the rodent toxicant epoxiconazole as more potent than thalidomide in our assay. We conclude that the PluriBeat assay is a novel method for predicting chemicals' adverse effects on embryonic development.


Assuntos
Bioensaio/métodos , Corpos Embrioides/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Células-Tronco Pluripotentes/efeitos dos fármacos , Teratogênicos/toxicidade , Testes de Toxicidade/métodos , Linhagem Celular , Biologia do Desenvolvimento , Corpos Embrioides/fisiologia , Compostos de Epóxi/toxicidade , Feminino , Humanos , Masculino , Miócitos Cardíacos/fisiologia , Oxazinas/metabolismo , Células-Tronco Pluripotentes/fisiologia , Teratogênese , Talidomida/toxicidade , Triazóis/toxicidade , Ácido Valproico/toxicidade , Xantenos/metabolismo
6.
J Neurosci Methods ; 330: 108486, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31706928

RESUMO

BACKGROUND: Neural differentiation from embryonic stem cells (ESCs) is an excellent model for elucidating the key mechanisms involved in neurogenesis, and also provides an unlimited source of progenitors for cell-based nerve regeneration. However, the existing protocols such as small molecule substances, 3D matrix, co-culture technique and transgenic method, are complicated and difficult to operate, thus are limited by laboratory conditions. Looking for an easy-to-operate protocol with easily gained material and high induction efficiency has always been a hot issue in neuroscience research. NEW METHODS: This paper established an optimized method for embryonic neurogenesis using a strategy of "combinatorial screening". In our study, the whole process of embryonic neurogenesis was divided into two phases, and the differentiation efficiency of seven experimental protocols in phase I and three protocols in phase II were systematically evaluated in A2lox and 129 ESCs. RESULTS: In phase I differentiation, "2-day embryoid bodies formation + 6-day retinoic acid induction" (Phase I-protocol 3) could effectively induce the differentiation of ESCs into neural precursor cells (NPCs). Furthermore, in phase II, N2B27 medium II (Phase II-protocol 3) could better support the subsequent differentiation from NPCs into neurons. COMPARISON WITH EXISTING METHOD(S): Such a combinational method (phase I-protocol 3 and phase II-protocol 3) can realize embryonic neurogenesis with high efficiency, easy implementation and low-cost, and is suitable for promotion in most laboratories. CONCLUSIONS: Through "combinatorial screening" strategy, we established an optimized method for embryonic neurogenesis in vitro, which is expected to be a powerful tool for neuroscience research.


Assuntos
Diferenciação Celular/fisiologia , Técnicas de Cultura/métodos , Células-Tronco Embrionárias/fisiologia , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Neurociências/métodos , Animais , Corpos Embrioides/fisiologia , Camundongos , Camundongos Knockout
7.
Stem Cells Dev ; 28(21): 1434-1450, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31507235

RESUMO

Embryonic stem (ES) cells have been utilized as an excellent model for the study of neural development. However, the dynamic changes of ES cell-derived neural stem cells (ES-NSCs), under the effects of prolonged cell culture and hypoxic conditions, are still obscured. In the present study, using the combination of serum-free culture of embryoid body-like aggregates (SFEB) culture and cell sorting by Sox-1, the ES-NSCs were easily isolated and showed in vitro temporal neural specification, which resulted in distinct cell fates after neural differentiation. Early passaged ES-NSCs gave rise to neurons, whereas late-passaged ES-NSCs gave rise to glial cells, similar to the in vivo dynamic changes during the neural development. Remarkably, hypoxia treatment induced the neural differentiation of ES-NSCs but did not affect the cell fate. Under hypoxic conditions, early passaged ES-NSCs showed the upregulation of neuronal markers, whereas late-passaged ES-NSCs showed the upregulation of a glial marker. In addition, the knockdown of the hypoxia-inducible factor 1α expression impaired the neuronal differentiation of early passaged ES-NSCs under hypoxic conditions. These data demonstrated the distinct effects of prolonged culture and hypoxic stimuli on the neural differentiation of ES-NSCs; prolonged culture was involved in the cell fate after neural differentiation, while hypoxia treatment efficiently promoted neural differentiation.


Assuntos
Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Neurais/citologia , Neurogênese/fisiologia , Neuroglia/citologia , Neurônios/citologia , Animais , Hipóxia Celular/fisiologia , Células Cultivadas , Corpos Embrioides/fisiologia , Citometria de Fluxo/métodos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Camundongos , Células-Tronco Neurais/fisiologia , Interferência de RNA , RNA Interferente Pequeno/genética , Fatores de Transcrição SOXB1/metabolismo
8.
Methods Mol Biol ; 2048: 107-119, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31396935

RESUMO

Human natural killer (NK) cell-based adoptive anticancer immunotherapy has gained intense interest with many clinical trials actively recruiting patients to treat a variety of both hematological malignancies and solid tumors. Most of these trials use primary NK cells isolated either from peripheral blood (PB-NK cells) or umbilical cord blood (UCB-NK cells), though these sources require NK cell collection for each patient leading to donor variability and heterogeneity in the NK cell populations. In contrast, NK cells derived human embryonic stem cells (hESC-NK cells) or induced pluripotent stem cells (hiPSC-NK cells) provide more homogeneous cell populations that can be grown at clinical scale, and genetically engineered if needed. These characteristics make hESC-/iPSC-derived NK cells an ideal cell population for developing standardized, "off-the-shelf" immunotherapy products. Additionally, production of NK cells from undifferentiated human pluripotent stem cells enables studies to better define pathways that regulate human NK cell development and function. Our group previously has established a stromal-free, two-stage culture system to derive NK cells from hESC/hiPSC in vitro followed by clinical-scale expansion of these cells using interleukin (IL)-21 expressing artificial antigen-presenting cells. However, prior to differentiation, this method requires single-cell adaptation of hESCs/hiPSCs which takes months. Recently we optimized this method by adapting the mouse embryonic fibroblast-dependent hESC/hiPSC to feeder-free culture conditions. These feeder-free hESCs/hiPSCs are directly used to form embryoid body (EB) to generate hemato-endothelial precursor cells. This new method produces mature, functional NK cells with higher efficiency to enable rapid production of an essentially unlimited number of homogenous NK cells that can be used for standardized, targeted immunotherapy for the treatment of refractory cancers and infectious diseases.


Assuntos
Técnicas de Cultura de Células/métodos , Diferenciação Celular , Células-Tronco Embrionárias Humanas/fisiologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Células Matadoras Naturais/fisiologia , Animais , Técnicas de Cultura de Células/instrumentação , Linhagem Celular , Proliferação de Células , Meios de Cultura/metabolismo , Corpos Embrioides/fisiologia , Citometria de Fluxo/instrumentação , Citometria de Fluxo/métodos , Humanos , Imunoterapia Adotiva/métodos , Células Matadoras Naturais/transplante , Camundongos , Neoplasias/imunologia , Neoplasias/terapia
9.
Methods Mol Biol ; 2048: 245-257, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31396942

RESUMO

Human pluripotent stem cells (PSCs) have the potential to provide a virtually unlimited supply of cells for transplantation therapy. When combined with recent advances in genome editing technologies, human PSCs could offer various approaches that enable gene therapy, drug discovery, disease modeling, and in vitro modeling of human development. De novo generation of hematopoietic stem cells (HSCs) from human PSCs is an important focus in the field, since it enables autologous HSC transplantation to treat many blood disorders and malignancies. Although culture conditions have been established to generate a broad spectrum of hematopoietic progenitors from human PSCs, it remains a significant challenge to generate bona fide HSCs that possess sustained self-renewal and multilineage differentiation capacities upon transplantation. In this review, recent promising advances in the efforts to generate HSCs and hematopoietic progenitors from human PSCs in vitro and in vivo or from somatic cells are discussed.


Assuntos
Diferenciação Celular , Células-Tronco Hematopoéticas/fisiologia , Células-Tronco Pluripotentes/fisiologia , Cultura Primária de Células/métodos , Animais , Autorrenovação Celular , Células Cultivadas , Técnicas de Cocultura/instrumentação , Técnicas de Cocultura/métodos , Meios de Cultura/metabolismo , Corpos Embrioides/fisiologia , Transplante de Células-Tronco Hematopoéticas/métodos , Humanos , Células-Tronco Mesenquimais , Camundongos , Camundongos SCID , Cultura Primária de Células/instrumentação , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Quimeras de Transplante , Transplante Autólogo/métodos , Transplante Heterólogo/métodos
10.
Reprod Toxicol ; 85: 26-33, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30659931

RESUMO

Benzo[a]pyrene (B[a]P) is one of the most common polycyclic aromatic hydrocarbons. In utero B[a]P exposure exerts multiple adverse effects on embryo development, although the underlying molecular mechanisms have still not been clearly elucidated. In the present study, we used human embryonic stem cell derived embryoid body (EB) as an in vitro model to investigate the embryotoxicity effects of B[a]P. EBs were exposed to B[a]P for 14 days, and the morphology, viability and differentiation of the cells were analyzed, in addition to the molecular changes. The results showed that B[a]P exposure repressed cell growth, impaired the morphology, and triggered apoptosis in the EBs. In addition, B[a]P reduced the gene expression levels of the ectoderm, mesoderm and endoderm biomarkers. Finally, B[a]P inhibited the epithelial-mesenchymal transition (EMT) process and the Akt/GSK-3ß signaling pathway. Taken together, B[a]P-induced aberrant EB development and apoptosis were related to EMT process and the Akt/GSK-3ß signaling pathway modulation.


Assuntos
Benzo(a)pireno/toxicidade , Corpos Embrioides/efeitos dos fármacos , Teratogênicos/toxicidade , Apoptose/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Corpos Embrioides/fisiologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Glicogênio Sintase Quinase 3 beta/metabolismo , Células-Tronco Embrionárias Humanas , Humanos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos
11.
Stem Cell Res ; 31: 201-215, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30118958

RESUMO

Directed differentiation of pluripotent stem cells provides an accessible system to model development. However, the distinct cell types that emerge, their dynamics, and their relationship to progenitors in the early embryo has been difficult to decipher because of the cellular heterogeneity inherent to differentiation. Here, we used a combination of bulk RNA-Seq, single cell RNA-Seq, and bioinformatics analyses to dissect the cell types that emerge during directed differentiation of mouse embryonic stem cells as embryoid bodies and we compared them to spatially and temporally resolved transcriptional profiles of early embryos. Our single cell analyses of the day 4 embryoid bodies revealed three populations which had retained related yet distinct pluripotent signatures that resemble the pre- or post-implantation epiblast, one population of presumptive neuroectoderm, one population of mesendoderm, and four populations of neural progenitors. By day 6, the neural progenitors predominated the embryoid bodies, but both a small population of pluripotent-like cells and an anterior mesoderm-like Brachyury-expressing population were present. By comparing the day 4 and day 6 populations, we identified candidate differentiation paths, transcription factors, and signaling pathways that mark the in vitro correlate of the transition from the mid-to-late primitive streak stage.


Assuntos
Corpos Embrioides/fisiologia , Desenvolvimento Embrionário/fisiologia , Mesoderma/metabolismo , Diferenciação Celular , Humanos
12.
PLoS One ; 13(5): e0196817, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29734353

RESUMO

Recent studies have suggested a role for the Cyclin Dependent Kinase-2 Associated Protein 1 (CDK2AP1) in stem cell differentiation and self-renewal. In studies with mouse embryonic stem cells (mESCs) derived from generated mice embryos with targeted deletion of the Cdk2ap1 gene, CDK2AP1 was shown to be required for epigenetic silencing of Oct4 during differentiation, with deletion resulting in persistent self-renewal and reduced differentiation potential. Differentiation capacity was restored in these cells following the introduction of a non-phosphorylatible form of the retinoblastoma protein (pRb) or exogenous Cdk2ap1. In this study, we investigated the role of CDK2AP1 in human embryonic stem cells (hESCs). Using a shRNA to reduce its expression in hESCs, we found that CDK2AP1 knockdown resulted in a significant reduction in the expression of the pluripotency genes, OCT4 and NANOG. We also found that CDK2AP1 knockdown increased the number of embryoid bodies (EBs) formed when differentiation was induced. In addition, the generated EBs had significantly higher expression of markers of all three germ layers, indicating that CDK2AP1 knockdown enhanced differentiation. CDK2AP1 knockdown also resulted in reduced proliferation and reduced the percentage of cells in the S phase and increased cells in the G2/M phase of the cell cycle. Further investigation revealed that a higher level of p53 protein was present in the CDK2AP1 knockdown hESCs. In hESCs in which p53 and CDK2AP1 were simultaneously downregulated, OCT4 and NANOG expression was not affected and percentage of cells in the S phase of the cell cycle was not reduced. Taken together, our results indicate that the knockdown of CDK2AP1 in hESCs results in increased p53 and enhances differentiation and favors it over a self-renewal fate.


Assuntos
Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Células-Tronco Embrionárias Humanas/fisiologia , Proteínas Supressoras de Tumor/genética , Animais , Ciclo Celular/genética , Proliferação de Células/genética , Corpos Embrioides/fisiologia , Técnicas de Silenciamento de Genes/métodos , Camadas Germinativas/fisiologia , Humanos , Camundongos , Proteína Homeobox Nanog/genética , Fator 3 de Transcrição de Octâmero/genética , RNA Interferente Pequeno/genética , Proteína do Retinoblastoma/genética , Proteína Supressora de Tumor p53/genética
13.
Stem Cell Reports ; 10(3): 1088-1101, 2018 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-29456180

RESUMO

The transition from naive to primed state of pluripotent stem cells is hallmarked by epithelial-mesenchymal transition, metabolic switch from oxidative phosphorylation to aerobic glycolysis, and changes in the epigenetic landscape. Since these changes are also seen as putative hallmarks of neoplastic cell transformation, we hypothesized that oncogenic pathways may be involved in this process. We report that the activity of RAS is repressed in the naive state of mouse embryonic stem cells (ESCs) and that all three RAS isoforms are significantly activated upon early differentiation induced by LIF withdrawal, embryoid body formation, or transition to the primed state. Forced expression of active RAS and RAS inhibition have shown that RAS regulates glycolysis, CADHERIN expression, and the expression of repressive epigenetic marks in pluripotent stem cells. Altogether, this study indicates that RAS is located at a key junction of early ESC differentiation controlling key processes in priming of naive cells.


Assuntos
Células-Tronco Pluripotentes/metabolismo , Células-Tronco Pluripotentes/fisiologia , Proteínas ras/metabolismo , Animais , Biomarcadores/metabolismo , Diferenciação Celular/fisiologia , Células Cultivadas , Corpos Embrioides/metabolismo , Corpos Embrioides/fisiologia , Epigênese Genética/fisiologia , Camundongos , Células-Tronco Embrionárias Murinas/metabolismo , Células-Tronco Embrionárias Murinas/fisiologia , Isoformas de Proteínas/metabolismo , Transdução de Sinais/fisiologia
14.
Stem Cells ; 36(5): 655-670, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29314416

RESUMO

MicroRNAs (miRNAs) are known regulators of various cellular processes, including pluripotency and differentiation of embryonic stem cells (ESCs). We analyzed differentiation of two ESC lines-D3 and B8, and observed significant differences in the expression of miRNAs and genes involved in pluripotency and differentiation. We also examined if transient miRNA overexpression could serve as a sufficient impulse modulating differentiation of mouse ESCs. ESCs were transfected with miRNA Mimics and differentiated in embryoid bodies and embryoid body outgrowths. miRNAs involved in differentiation of mesodermal lineages, such as miR145 and miR181, as well as miRNAs regulating myogenesis (MyomiRs)-miR1, miR133a, miR133b, and miR206 were tested. Using such approach, we proved that transient overexpression of molecules selected by us modulated differentiation of mouse ESCs. Increase in miR145 levels upregulated Pax3, Pax7, Myod1, Myog, and MyHC2, while miR181 triggered the expression of such crucial myogenic factors as Myf5 and MyHC2. As a result, the ability of ESCs to initiate myogenic differentiation and form myotubes was enhanced. Premature expression of MyomiRs had, however, an adverse effect on myogenic differentiation of ESCs. Stem Cells 2018;36:655-670.


Assuntos
Células-Tronco Embrionárias/citologia , MicroRNAs/genética , Desenvolvimento Muscular/genética , Mioblastos/citologia , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Corpos Embrioides/fisiologia , Camundongos , Desenvolvimento Muscular/fisiologia
15.
Reprod Sci ; 25(5): 712-726, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-28854867

RESUMO

Embryoid bodies (EBs) can serve as a system for evaluating pluripotency, cellular differentiation, and tissue morphogenesis. In this study, we use EBs derived from mouse embryonic stem cells (mESCs) and human amniocyte-derived induced pluripotent stem cells (hAdiPSCs) as a model for ovarian granulosa cell (GC) development and steroidogenic cell commitment. We demonstrated that spontaneously differentiated murine EBs (mEBs) and human EBs (hEBs) displayed ovarian GC markers, such as aromatase (CYP19A1), FOXL2, AMHR2, FSHR, and GJA1. Comparative microarray analysis identified both shared and unique gene expression between mEBs and the maturing mouse ovary. Gene sets related to gonadogenesis, lipid metabolism, and ovarian development were significantly overrepresented in EBs. Of the 29 genes, 15 that were differentially regulated in steroidogenic mEBs displayed temporal expression changes between embryonic, postnatal, and mature ovarian tissues by polymerase chain reaction. Importantly, both mEBs and hEBs were capable of gonadotropin-responsive estradiol (E2) synthesis in vitro (217-759 pg/mL). Live fluorescence-activated cell sorting-sorted AMHR2+ granulosa-like cells from mEBs continued to produce E2 after purification (15.3 pg/mL) and secreted significantly more E2 than AMHR2- cells (8.6 pg/mL, P < .05). We conclude that spontaneously differentiated EBs of both mESC and hAdiPSC origin can serve as a biologically relevant model for ovarian GC differentiation and steroidogenic cell commitment. These cells should be further investigated for therapeutic uses, such as stem cell-based hormone replacement therapy and in vitro maturation of oocytes.


Assuntos
Corpos Embrioides/fisiologia , Células da Granulosa/fisiologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Esteroides/biossíntese , Animais , Corpos Embrioides/metabolismo , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/fisiologia , Feminino , Expressão Gênica , Células da Granulosa/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos
16.
J Vis Exp ; (125)2017 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-28784957

RESUMO

Although a number of in vitro disease models have been developed using hiPSCs, one limitation is that these two-dimensional (2-D) systems may not represent the underlying cytoarchitectural and functional complexity of the affected individuals carrying suspected disease variants. Conventional 2-D models remain incomplete representations of in vivo-like structures and do not adequately capture the complexity of the brain. Thus, there is an emerging need for more 3-D hiPSC-based models that can better recapitulate the cellular interactions and functions seen in an in vivo system. Here we report a protocol to develop a 3-D system from undifferentiated hiPSCs based on the serum free embryoid body (SFEB). This 3-D model mirrors aspects of a developing ventralized neocortex and allows for studies into functions integral to living neural cells and intact tissue such as migration, connectivity, communication, and maturation. Specifically, we demonstrate that the SFEBs using our protocol can be interrogated using physiologically relevant and high-content cell based assays such as calcium imaging, and multi-electrode array (MEA) recordings without cryosectioning. In the case of MEA recordings, we demonstrate that SFEBs increase both spike activity and network-level bursting activity during long-term culturing. This SFEB protocol provides a robust and scalable system for the study of developing network formation in a 3-D model that captures aspects of early cortical development.


Assuntos
Técnicas de Cultura de Células/métodos , Técnicas de Cocultura/métodos , Corpos Embrioides/fisiologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Animais , Diferenciação Celular , Humanos , Camundongos , Neurônios/fisiologia
17.
Ann Otol Rhinol Laryngol ; 126(7): 517-524, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28604083

RESUMO

OBJECTIVES: Compared with using autologous tissue, the use of artificial materials in the regeneration of tracheal defects is minimally invasive. However, this technique requires early epithelialization on the inner side of the artificial trachea. After differentiation from induced pluripotent stem cells (iPSCs), tracheal epithelial tissues may be used to produce artificial tracheas. Herein, we aimed to demonstrate that after differentiation from fluorescent protein-labeled iPSCs, tracheal epithelial tissues survived in nude rats with tracheal defects. METHODS: Red fluorescent tdTomato protein was electroporated into mouse iPSCs to produce tdTomato-labeled iPSCs. Embryoid bodies derived from these iPSCs were then cultured in differentiation medium supplemented with growth factors, followed by culture on air-liquid interfaces for further differentiation into tracheal epithelium. The cells were implanted with artificial tracheas into nude rats with tracheal defects on day 26 of cultivation. On day 7 after implantation, the tracheas were exposed and examined histologically. RESULTS: Tracheal epithelial tissue derived from tdTomato-labeled iPSCs survived in the tracheal defects. Moreover, immunochemical analyses showed that differentiated tissues had epithelial structures similar to those of proximal tracheal tissues. CONCLUSIONS: After differentiation from iPSCs, tracheal epithelial tissues survived in rat bodies, warranting the use of iPSCs for epithelial regeneration in tracheal defects.


Assuntos
Células Epiteliais/fisiologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Engenharia Tecidual/métodos , Traqueia/citologia , Animais , Diferenciação Celular , Células Cultivadas , Corpos Embrioides/fisiologia , Corantes Fluorescentes , Proteínas Luminescentes , Masculino , Ratos Nus , Regeneração , Alicerces Teciduais , Traqueia/fisiologia , Proteína Vermelha Fluorescente
18.
Development ; 144(6): 976-985, 2017 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-28292844

RESUMO

Cells have an intrinsic ability to self-assemble and self-organize into complex and functional tissues and organs. By taking advantage of this ability, embryoids, organoids and gastruloids have recently been generated in vitro, providing a unique opportunity to explore complex embryological events in a detailed and highly quantitative manner. Here, we examine how such approaches are being used to answer fundamental questions in embryology, such as how cells self-organize and assemble, how the embryo breaks symmetry, and what controls timing and size in development. We also highlight how further improvements to these exciting technologies, based on the development of quantitative platforms to precisely follow and measure subcellular and molecular events, are paving the way for a more complete understanding of the complex events that help build the human embryo.


Assuntos
Corpos Embrioides/fisiologia , Desenvolvimento Embrionário , Gástrula/embriologia , Organoides/embriologia , Animais , Tamanho Corporal , Humanos , Fatores de Tempo
19.
Reprod Toxicol ; 70: 102-115, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28192181

RESUMO

A major limitation to culturing tissues and organs is the lack of a functional vascular network in vitro. The zebrafish possess many useful properties which makes it a promising model for such studies. Unfortunately, methods of culturing endothelial cells from this species are not well characterised. Here, we tried two methods (embryoid body culture and organ explants from transgenic zebrafish kdrl:GFP embryos) to develop in vitro vascular networks. In the kdrl:GFP line, endothelial cells expresses green fluorescent protein, which allows to track the vascular development in live cultures. We found that embryoid bodies showed significantly longer and wider branches of connected endothelial cells when grown in a microfluidic system than in static culture. Similarly, sprouting of kdrl:GFP+ cells from the tissue explants was observed in a 3D hydrogel matrix. This study is a step towards the development of zebrafish vascular networks in vitro.


Assuntos
Embrião não Mamífero/irrigação sanguínea , Corpos Embrioides/fisiologia , Células Endoteliais/fisiologia , Neovascularização Fisiológica , Animais , Animais Geneticamente Modificados , Células Cultivadas , Proteínas de Fluorescência Verde/genética , Coração/embriologia , Fígado/citologia , Fígado/embriologia , Miocárdio/citologia , Peixe-Zebra
20.
Artigo em Inglês | MEDLINE | ID: mdl-27911036

RESUMO

Embryoid bodies (EBs) have been popular in vitro differentiation models for pluripotent stem cells for more than five decades. Initially, defined as aggregates formed by embryonal carcinoma cells, EBs gained more prominence after the derivation of karyotypically normal embryonic stem cells from early mouse blastocysts. In many cases, formation of EBs constitutes an important initial step in directed differentiation protocols aimed at generated specific cell types from undifferentiated stem cells. Indeed state-of-the-art protocols for directed differentiation of cardiomyocytes still rely on this initial EB step. Analyses of spontaneous differentiation of embryonic stem cells in EBs have yielded important insights into the molecules that direct primitive endoderm differentiation and many of the lessons we have learned about the signals and transcription factors governing this differentiation event is owed to EB models, which later were extensively validated in studies of early mouse embryos. EBs show a degree of self-organization that mimics some aspects of early embryonic development, but with important exceptions. Recent studies that employ modern signaling reporters and tracers of lineage commitment have revealed both the strengths and the weaknesses of EBs as a model of embryonic axis formation. In this review, we discuss the history, application, and future potential of EBs as an experimental model. WIREs Dev Biol 2017, 6:e259. doi: 10.1002/wdev.259 For further resources related to this article, please visit the WIREs website.


Assuntos
Diferenciação Celular , Linhagem da Célula , Corpos Embrioides/citologia , Corpos Embrioides/fisiologia , Desenvolvimento Embrionário/fisiologia , Animais , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...