Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 176
Filtrar
1.
BMC Biol ; 21(1): 84, 2023 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-37069561

RESUMO

BACKGROUND: The non-neuronal retinal pigment epithelium (RPE) functions in intimate association with retinal photoreceptors, performing a multitude of tasks critical for maintaining retinal homeostasis and collaborating with retinal glial cells to provide metabolic support and ionic buffering. Accordingly, the RPE has recently been shown to display dynamic properties mediated by an array of ion channels usually more characteristic of astrocytes and excitable cells. The recent discovery of canonical voltage-activated Na+ channels in the RPE and their importance for phagocytosis of photoreceptor outer segments raises a question about their electrogenic function. Here, we performed a detailed electrophysiological analysis related to the functioning of these channels in human embryonic stem cell (hESC)-derived RPE. RESULTS: Our studies examining the electrical properties of the hESC-RPE revealed that its membrane mainly displays passive properties in a broad voltage range, with the exception of depolarization-induced spikes caused by voltage-activated Na+ current (INa). Spike amplitude depended on the availability of INa and spike kinetics on the membrane time constant, and the spikes could be largely suppressed by TTX. Membrane resistance fluctuated rapidly and strongly, repeatedly changing over the course of recordings and causing closely correlated fluctuations in resting membrane potential. In a minority of cells, we found delayed secondary INa-like inward currents characterized by comparatively small amplitudes and slow kinetics, which produced secondary depolarizing spikes. Up to three consecutive delayed inward current waves were detected. These currents could be rapidly and reversibly augmented by applying L-type Ca2+ channel blocker nifedipine to diminish influx of calcium and thus increase gap junctional conductance. CONCLUSIONS: This work shows, for the first time, that INa and INa-mediated voltage spikes can spread laterally through gap junctions in the monolayer of cells that are traditionally considered non-excitable. Our findings support a potential role of the RPE that goes beyond giving homeostatic support to the retina.


Assuntos
Retina , Epitélio Pigmentado da Retina , Humanos , Células-Tronco Embrionárias Humanas/fisiologia
2.
Cells ; 12(6)2023 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-36980244

RESUMO

The rare SLC30A8 mutation encoding a truncating p.Arg138* variant (R138X) in zinc transporter 8 (ZnT8) is associated with a 65% reduced risk for type 2 diabetes. To determine whether ZnT8 is required for beta cell development and function, we derived human pluripotent stem cells carrying the R138X mutation and differentiated them into insulin-producing cells. We found that human pluripotent stem cells with homozygous or heterozygous R138X mutation and the null (KO) mutation have normal efficiency of differentiation towards insulin-producing cells, but these cells show diffuse granules that lack crystalline zinc-containing insulin granules. Insulin secretion is not compromised in vitro by KO or R138X mutations in human embryonic stem cell-derived beta cells (sc-beta cells). Likewise, the ability of sc-beta cells to secrete insulin and maintain glucose homeostasis after transplantation into mice was comparable across different genotypes. Interestingly, sc-beta cells with the SLC30A8 KO mutation showed increased cytoplasmic zinc, and cells with either KO or R138X mutation were resistant to apoptosis when extracellular zinc was limiting. These findings are consistent with a protective role of zinc in cell death and with the protective role of zinc in T2D.


Assuntos
Proteínas de Transporte de Cátions , Diabetes Mellitus Tipo 2 , Células-Tronco Embrionárias Humanas , Transportador 8 de Zinco , Zinco , Animais , Humanos , Camundongos , Apoptose/genética , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Células-Tronco Embrionárias Humanas/metabolismo , Células-Tronco Embrionárias Humanas/fisiologia , Insulina/metabolismo , Mutação com Perda de Função , Mutação/genética , Zinco/metabolismo , Transportador 8 de Zinco/genética , Transportador 8 de Zinco/metabolismo
3.
Nat Commun ; 12(1): 6768, 2021 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-34799555

RESUMO

Organizing centers secrete morphogens that specify the emergence of germ layers and the establishment of the body's axes during embryogenesis. While traditional experimental embryology tools have been instrumental in dissecting the molecular aspects of organizers in model systems, they are impractical in human in-vitro model systems to dissect the relationships between signaling and fate along embryonic coordinates. To systematically study human embryonic organizer centers, we devised a collection of optogenetic ePiggyBac vectors to express a photoactivatable Cre-loxP recombinase, that allows the systematic induction of organizer structures by shining blue-light on human embryonic stem cells (hESCs). We used a light stimulus to geometrically confine SHH expression in neuralizing hESCs. This led to the self-organization of mediolateral neural patterns. scRNA-seq analysis established that these structures represent the dorsal-ventral forebrain, at the end of the first month of development. Here, we show that morphogen light-stimulation is a scalable tool that induces self-organizing centers.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas Hedgehog/metabolismo , Células-Tronco Embrionárias Humanas/fisiologia , Prosencéfalo/embriologia , Linhagem da Célula/fisiologia , Embriologia/métodos , Regulação da Expressão Gênica no Desenvolvimento/efeitos da radiação , Vetores Genéticos/genética , Humanos , Integrases/genética , Luz , Optogenética/métodos , Prosencéfalo/metabolismo , RNA-Seq , Transdução de Sinais/fisiologia , Transdução de Sinais/efeitos da radiação , Análise de Célula Única
4.
Nat Commun ; 12(1): 6749, 2021 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-34799566

RESUMO

The hypothalamus regulates metabolic homeostasis by influencing behavior and endocrine systems. Given its role governing key traits, such as body weight and reproductive timing, understanding the genetic regulation of hypothalamic development and function could yield insights into disease pathogenesis. However, given its inaccessibility, studying human hypothalamic gene regulation has proven challenging. To address this gap, we generate a high-resolution chromatin architecture atlas of an established embryonic stem cell derived hypothalamic-like neuron model across three stages of in vitro differentiation. We profile accessible chromatin and identify physical contacts between gene promoters and putative cis-regulatory elements to characterize global regulatory landscape changes during hypothalamic differentiation. Next, we integrate these data with GWAS loci for various complex traits, identifying multiple candidate effector genes. Our results reveal common target genes for these traits, potentially affecting core developmental pathways. Our atlas will enable future efforts to determine hypothalamic mechanisms influencing disease susceptibility.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Células-Tronco Embrionárias Humanas/fisiologia , Hipotálamo/embriologia , Neurônios/fisiologia , Diferenciação Celular/genética , Linhagem Celular , Mapeamento Cromossômico , Estudo de Associação Genômica Ampla , Humanos , Hipotálamo/citologia , Herança Multifatorial , RNA-Seq , Elementos Reguladores de Transcrição/genética
5.
Nat Methods ; 18(9): 1046-1055, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34480151

RESUMO

Chromosome conformation capture (3C) assays are used to map chromatin interactions genome-wide. Chromatin interaction maps provide insights into the spatial organization of chromosomes and the mechanisms by which they fold. Hi-C and Micro-C are widely used 3C protocols that differ in key experimental parameters including cross-linking chemistry and chromatin fragmentation strategy. To understand how the choice of experimental protocol determines the ability to detect and quantify aspects of chromosome folding we have performed a systematic evaluation of 3C experimental parameters. We identified optimal protocol variants for either loop or compartment detection, optimizing fragment size and cross-linking chemistry. We used this knowledge to develop a greatly improved Hi-C protocol (Hi-C 3.0) that can detect both loops and compartments relatively effectively. In addition to providing benchmarked protocols, this work produced ultra-deep chromatin interaction maps using Micro-C, conventional Hi-C and Hi-C 3.0 for key cell lines used by the 4D Nucleome project.


Assuntos
Cromatina/química , Cromossomos Humanos/química , Reagentes de Ligações Cruzadas/química , Técnicas Genéticas , Linhagem Celular , Cromatina/metabolismo , Bases de Dados Factuais , Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Embrionárias Humanas/fisiologia , Humanos
6.
Int J Mol Sci ; 22(13)2021 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-34209900

RESUMO

Adult human cardiomyocytes have an extremely limited proliferative capacity, which poses a great barrier to regenerative medicine and research. Human embryonic stem cells (hESCs) have been proposed as an alternative source to generate large numbers of clinical grade cardiomyocytes (CMs) that can have potential therapeutic applications to treat cardiac diseases. Previous studies have shown that bioactive lipids are involved in diverse cellular responses including cardiogenesis. In this study, we explored the novel function of the chemically synthesized bioactive lipid O-cyclic phytosphingosine-1-phosphate (cP1P) as an inducer of cardiac differentiation. Here, we identified cP1P as a novel factor that significantly enhances the differentiation potential of hESCs into cardiomyocytes. Treatment with cP1P augments the beating colony number and contracting area of CMs. Furthermore, we elucidated the molecular mechanism of cP1P regulating SMAD1/5/8 signaling via the ALK3/BMP receptor cascade during cardiac differentiation. Our result provides a new insight for cP1P usage to improve the quality of CM differentiation for regenerative therapies.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Células-Tronco Embrionárias Humanas/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Esfingosina/análogos & derivados , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Diferenciação Celular/genética , Células Cultivadas , Regulação da Expressão Gênica/efeitos dos fármacos , Células-Tronco Embrionárias Humanas/fisiologia , Humanos , Lipídeos/química , Lipídeos/farmacologia , Miócitos Cardíacos/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Esfingosina/química , Esfingosina/farmacologia
7.
Stem Cells Transl Med ; 10(11): 1491-1499, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34302450

RESUMO

Experimental cell models are indispensable for clarifying the pathophysiology of coronavirus disease 2019 (COVID-19), which is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, and for developing therapeutic agents. To recapitulate the symptoms and drug response of COVID-19 patients in vitro, SARS-CoV-2 studies using physiologically relevant human embryonic stem (ES)/induced pluripotent stem (iPS) cell-derived somatic cells and organoids are ongoing. These cells and organoids have been used to show that SARS-CoV-2 can infect and damage various organs including the lung, heart, brain, intestinal tract, kidney, and pancreas. They are also being used to develop COVID-19 therapeutic agents, including evaluation of their antiviral efficacy and safety. The relationship between COVID-19 aggravation and human genetic backgrounds has been investigated using genetically modified ES/iPS cells and patient-derived iPS cells. This review summarizes the latest results and issues of SARS-CoV-2 research using human ES/iPS cell-derived somatic cells and organoids.


Assuntos
COVID-19 , Células-Tronco Embrionárias Humanas/fisiologia , Organoides/fisiologia , SARS-CoV-2/fisiologia , Pesquisa Biomédica/métodos , Pesquisa Biomédica/tendências , COVID-19/etiologia , COVID-19/patologia , COVID-19/terapia , Terapia Genética/métodos , Terapia Genética/tendências , Células-Tronco Embrionárias Humanas/transplante , Humanos , Células-Tronco Pluripotentes Induzidas/fisiologia , Células-Tronco Pluripotentes Induzidas/transplante , Organoides/citologia , Organoides/transplante
8.
Endocrinology ; 162(9)2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34125902

RESUMO

Gonadotropin-releasing hormone (GnRH) neurons in the hypothalamus play a key role in the regulation of reproductive function. In this study, we sought an efficient method for generating GnRH neurons from human embryonic and induced pluripotent stem cells (hESC and hiPSC, respectively). First, we found that exposure of primitive neuroepithelial cells, rather than neuroprogenitor cells, to fibroblast growth factor 8 (FGF8), was more effective in generating GnRH neurons. Second, addition of kisspeptin to FGF8 further increased the efficiency rates of GnRH neurogeneration. Third, we generated a fluorescent marker mCherry labeled human embryonic GnRH cell line (mCh-hESC) using a CRISPR-Cas9 targeting approach. Fourth, we examined physiological characteristics of GnRH (mCh-hESC) neurons: similar to GnRH neurons in vivo, they released the GnRH peptide in a pulsatile manner at ~60 min intervals; GnRH release increased in response to high potassium, kisspeptin, estradiol, and neurokinin B challenges; and injection of depolarizing current induced action potentials. Finally, we characterized developmental changes in transcriptomes of GnRH neurons using hESC, hiPSC, and mCh-hESC. The developmental pattern of transcriptomes was remarkably similar among the 3 cell lines. Collectively, human stem cell-derived GnRH neurons will be an important tool for establishing disease models to understand diseases, such as idiopathic hypothalamic hypogonadism, and testing contraceptive drugs.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Células-Tronco Embrionárias Humanas/fisiologia , Neurogênese/genética , Neurônios/fisiologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Células Cultivadas , Fator 8 de Crescimento de Fibroblasto/farmacologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Células-Tronco Embrionárias Humanas/efeitos dos fármacos , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/fisiologia , Neurogênese/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Transcriptoma/efeitos dos fármacos
9.
PLoS One ; 16(6): e0245634, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34077425

RESUMO

Self-organized patterning of mammalian embryonic stem cells on micropatterned surfaces has previously been established as an in vitro platform for early mammalian developmental studies, complimentary to in vivo studies. Traditional micropatterning methods, such as micro-contact printing (µCP), involve relatively complicated fabrication procedures, which restricts widespread adoption by biologists. Here, we demonstrate a rapid method of micropatterning by printing hydrogel micro-features onto a glass-bottomed culture vessel. The micro-features are printed using a projection stereolithography bioprinter yielding hydrogel structures that geometrically restrict the attachment of cells or proteins. Compared to traditional and physical photomasks, a digitally tunable virtual photomask is used in the projector to generate blue light patterns that enable rapid iteration with minimal cost and effort. We show that a protocol that makes use of this method together with LN521 coating, an extracellular matrix coating, creates a surface suitable for human embryonic stem cell (hESC) attachment and growth with minimal non-specific adhesion. We further demonstrate that self-patterning of hESCs following previously published gastrulation and ectodermal induction protocols achieves results comparable with those obtained with commercially available plates.


Assuntos
Células-Tronco Embrionárias Humanas/citologia , Hidrogéis/química , Microtecnologia/métodos , Estereolitografia/instrumentação , Células-Tronco Embrionárias Humanas/fisiologia , Humanos , Propriedades de Superfície
10.
Nat Commun ; 12(1): 3275, 2021 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-34045451

RESUMO

Despite advancements in human pluripotent stem cells (hPSCs) differentiation protocols to generate appropriate neuronal progenitors suitable for transplantation in Parkinson's disease, resultant grafts contain low proportions of dopamine neurons. Added to this is the tumorigenic risk associated with the potential presence of incompletely patterned, proliferative cells within grafts. Here, we utilised a hPSC line carrying a FailSafeTM suicide gene (thymidine kinase linked to cyclinD1) to selectively ablate proliferative cells in order to improve safety and purity of neural transplantation in a Parkinsonian model. The engineered FailSafeTM hPSCs demonstrated robust ventral midbrain specification in vitro, capable of forming neural grafts upon transplantation. Activation of the suicide gene within weeks after transplantation, by ganciclovir administration, resulted in significantly smaller grafts without affecting the total yield of dopamine neurons, their capacity to innervate the host brain or reverse motor deficits at six months in a rat Parkinsonian model. Within ganciclovir-treated grafts, other neuronal, glial and non-neural populations (including proliferative cells), were significantly reduced-cell types that may pose adverse or unknown influences on graft and host function. These findings demonstrate the capacity of a suicide gene-based system to improve both the standardisation and safety of hPSC-derived grafts in a rat model of Parkinsonism.


Assuntos
Engenharia Celular/métodos , Genes Transgênicos Suicidas , Doença de Parkinson Secundária/terapia , Transplante de Células-Tronco/métodos , Animais , Apoptose/genética , Diferenciação Celular , Linhagem Celular , Proliferação de Células/genética , Modelos Animais de Doenças , Neurônios Dopaminérgicos/fisiologia , Feminino , Genes bcl-1/genética , Xenoenxertos/citologia , Xenoenxertos/patologia , Células-Tronco Embrionárias Humanas/fisiologia , Humanos , Masculino , Mesencéfalo/citologia , Mesencéfalo/patologia , Oxidopamina/administração & dosagem , Oxidopamina/toxicidade , Doença de Parkinson Secundária/induzido quimicamente , Doença de Parkinson Secundária/patologia , Ratos , Transplante de Células-Tronco/efeitos adversos , Transplante de Células-Tronco/normas , Timidina Quinase/genética
11.
Nat Commun ; 12(1): 2829, 2021 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-33990559

RESUMO

Polycomb group (PcG) proteins maintain cell identity by repressing gene expression during development. Surprisingly, emerging studies have recently reported that a number of PcG proteins directly activate gene expression during cell fate determination process. However, the mechanisms by which they direct gene activation in pluripotency remain poorly understood. Here, we show that Phc1, a subunit of canonical polycomb repressive complex 1 (cPRC1), can exert its function in pluripotency maintenance via a PRC1-independent activation of Nanog. Ablation of Phc1 reduces the expression of Nanog and overexpression of Nanog partially rescues impaired pluripotency caused by Phc1 depletion. We find that Phc1 interacts with Nanog and activates Nanog transcription by stabilizing the genome-wide chromatin interactions of the Nanog locus. This adds to the already known canonical function of PRC1 in pluripotency maintenance via a PRC1-dependent repression of differentiation genes. Overall, our study reveals a function of Phc1 to activate Nanog transcription through regulating chromatin architecture and proposes a paradigm for PcG proteins to maintain pluripotency.


Assuntos
Cromatina/genética , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Proteína Homeobox Nanog/genética , Complexo Repressor Polycomb 1/genética , Complexo Repressor Polycomb 1/fisiologia , Animais , Células Cultivadas , Técnicas de Silenciamento de Genes , Técnicas de Inativação de Genes , Genoma Humano , Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Embrionárias Humanas/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Modelos Genéticos , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/fisiologia , Complexo Repressor Polycomb 1/antagonistas & inibidores , Complexo Repressor Polycomb 1/deficiência
12.
Sci Rep ; 11(1): 8242, 2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33859268

RESUMO

We identified a human embryonic stem cell subline that fails to respond to the differentiation cues needed to obtain endoderm derivatives, differentiating instead into extra-embryonic mesoderm. RNA-sequencing analysis showed that the subline has hyperactivation of the WNT and BMP4 signalling. Modulation of these pathways with small molecules confirmed them as the cause of the differentiation impairment. While activation of WNT and BMP4 in control cells resulted in a loss of endoderm differentiation and induction of extra-embryonic mesoderm markers, inhibition of these pathways in the subline restored its ability to differentiate. Karyotyping and exome sequencing analysis did not identify any changes in the genome that could account for the pathway deregulation. These findings add to the increasing evidence that different responses of stem cell lines to differentiation protocols are based on genetic and epigenetic factors, inherent to the line or acquired during cell culture.


Assuntos
Proteína Morfogenética Óssea 4/genética , Diferenciação Celular/genética , Células-Tronco Embrionárias Humanas/fisiologia , Proteínas Wnt/genética , Proteína Morfogenética Óssea 4/metabolismo , Células Cultivadas , Endoderma/citologia , Endoderma/fisiologia , Membranas Extraembrionárias/citologia , Membranas Extraembrionárias/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Embrionárias Humanas/metabolismo , Humanos , Mesoderma/citologia , Mesoderma/fisiologia , Transdução de Sinais/genética , Transcriptoma , Proteínas Wnt/metabolismo
13.
Sci China Life Sci ; 64(12): 2100-2113, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-33740188

RESUMO

A stable, rapid and effective neural differentiation method is essential for the clinical applications of human embryonic stem cells (ESCs) or induced pluripotent stem cells (iPSCs) in treating neurological disorders and diseases. Herein, we established a novel and robust monolayer differentiation method to produce functional neural progenitor cells (NPCs) from human ESC/iPSCs on Type I Collagen. The derived cells not only displayed the requisite markers, but also behaved similarly to classic NPCs both in vitro and in vivo. Upon transplantation into traumatic brain injury model, the derived NPCs facilitated recovery from injury. We also found that SMAD signaling stayed down throughout the differentiation process on Type I Collagen, and the pluripotent signals were rapidly downregulated along with raising up of neural early markers on the third day. Meanwhile, ATAC-seq data showed the related mediation of distinct transcriptome and global chromatin dynamics during NPC induction. Totally, our results thus provide a convenient way to generate NPCs from human ESC/iPSCs for neural diseases' treatment.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias Humanas/fisiologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Células-Tronco Neurais , Lesões Encefálicas Traumáticas/terapia , Técnicas de Cultura de Células , Colágeno Tipo I , Humanos , Células-Tronco Neurais/transplante , Análise de Sequência de RNA
14.
PLoS Comput Biol ; 17(3): e1008778, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33647016

RESUMO

Human pluripotent stem cells hold significant promise for regenerative medicine. However, long differentiation protocols and immature characteristics of stem cell-derived cell types remain challenges to the development of many therapeutic applications. In contrast to the slow differentiation of human stem cells in vitro that mirrors a nine-month gestation period, mouse stem cells develop according to a much faster three-week gestation timeline. Here, we tested if co-differentiation with mouse pluripotent stem cells could accelerate the differentiation speed of human embryonic stem cells. Following a six-week RNA-sequencing time course of neural differentiation, we identified 929 human genes that were upregulated earlier and 535 genes that exhibited earlier peaked expression profiles in chimeric cell cultures than in human cell cultures alone. Genes with accelerated upregulation were significantly enriched in Gene Ontology terms associated with neurogenesis, neuron differentiation and maturation, and synapse signaling. Moreover, chimeric mixed samples correlated with in utero human embryonic samples earlier than human cells alone, and acceleration was dose-dependent on human-mouse co-culture ratios. The altered gene expression patterns and developmental rates described in this report have implications for accelerating human stem cell differentiation and the use of interspecies chimeric embryos in developing human organs for transplantation.


Assuntos
Quimerismo , Células-Tronco Embrionárias Humanas , Neurogênese , Células-Tronco Pluripotentes , Animais , Células Cultivadas , Biologia Computacional , Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Embrionárias Humanas/fisiologia , Humanos , Camundongos , Neurogênese/genética , Neurogênese/fisiologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/fisiologia , Especificidade da Espécie , Transcriptoma/genética
15.
Exp Eye Res ; 202: 108305, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33080300

RESUMO

The biosafety and efficiency of transplanting retinal pigment epithelial (RPE) cells derived from both human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs) have been evaluated in phase I and phase II clinical trials. For further large-scale application, cryopreserved RPE cells must be used; thus, it is highly important to investigate the influence of cryopreservation and thawing on the biological characteristics of hESC-RPE cells and their post-transplantation vision-restoring function. Here, via immunofluorescence, qPCR, transmission electron microscopy, transepithelial electrical resistance, and enzyme-linked immunosorbent assays (ELISAs), we showed that cryopreserved hESC-RPE cells retained the specific gene expression profile, morphology, ultrastructure, and maturity-related functions of induced RPE cells. Additionally, cryopreserved hESC-RPE cells exhibited a polarized monolayer, tight junction, and gap junction structure and an in vitro nanoparticle phagocytosis capability similar to those of induced hESC-RPE cells. However, the level of pigment epithelium-derived factor (PEDF) secretion was significantly decreased in cryopreserved hESC-RPE cells. Royal College of Surgeons rats with cryopreserved hESC-RPE cells engrafted into the subretinal space exhibited a significant decrease in the b-wave amplitude compared with rats engrafted with induced hESC-RPE cells at 4 weeks post transplantation. However, the difference disappeared at 8 weeks and 12 weeks post operation. No significant difference in the outer nuclear layer (ONL) thickness was observed between the two groups. Our data showed that even after cryopreservation and thawing, cryopreserved hESC-RPE cells are still qualified as a donor cell source for cell-based therapy of retinal degenerative diseases.


Assuntos
Células-Tronco Embrionárias Humanas/fisiologia , Degeneração Retiniana/terapia , Epitélio Pigmentado da Retina/fisiologia , Transplante de Células-Tronco , Linhagem Celular , Polaridade Celular , Células Cultivadas , Criopreservação , Impedância Elétrica , Células-Tronco Embrionárias Humanas/ultraestrutura , Humanos , Microscopia Eletrônica de Transmissão , Degeneração Retiniana/metabolismo , Degeneração Retiniana/fisiopatologia , Epitélio Pigmentado da Retina/ultraestrutura
16.
Biomech Model Mechanobiol ; 20(1): 205-222, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32809130

RESUMO

Human embryonic stem cells (hESCs) can differentiate to three germ layers within biochemical and biomechanical niches. The complicated mechanical environments in vivo could have diverse effects on the fate decision and biological functions of hESCs. To globally screen mechanosensitive molecules, three typical types of mechanical stimuli, i.e., tensile stretch, shear flow, and mechanical compression, were applied in respective parameter sets of loading pattern, amplitude, frequency, and/or duration, and then, iTRAQ proteomics test was used for identifying and quantifying differentially expressed proteins in hESCs. Bioinformatics analysis identified 37, 41, and 23 proteins under stretch pattern, frequency, and duration, 13, 18, and 41 proteins under shear pattern, amplitude, and duration, and 4, 0, and 183 proteins under compression amplitude, frequency, and duration, respectively, where distinct parameters yielded the differentially weighted preferences under each stimulus. Ten mechanosensitive proteins were commonly shared between two of three mechanical stimuli, together with numerous proteins identified under single stimulus. More importantly, functional GSEA and WGCNA analyses elaborated the variations of the screened proteins with loading parameters. Common functions in protein synthesis and modification were identified among three stimuli, and specific functions were observed in skin development under stretch alone. In conclusion, mechanomics analysis is indispensable to map actual mechanosensitive proteins under physiologically mimicking mechanical environment, and sheds light on understanding the core hub proteins in mechanobiology.


Assuntos
Células-Tronco Embrionárias Humanas/fisiologia , Resistência ao Cisalhamento , Estresse Mecânico , Fenômenos Biomecânicos , Linhagem Celular , Nucléolo Celular/metabolismo , Análise por Conglomerados , Redes Reguladoras de Genes , Células-Tronco Embrionárias Humanas/metabolismo , Humanos , Mecanotransdução Celular , Fenótipo , Mapas de Interação de Proteínas , Proteínas/metabolismo , RNA Ribossômico/genética , RNA Ribossômico/metabolismo , Reprodutibilidade dos Testes , Frações Subcelulares/metabolismo , Resistência à Tração
17.
Reproduction ; 161(1): F41-F51, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33258799

RESUMO

Development of the preimplantation embryo is reliant on nutrients present in the milieu of the reproductive tract. While carbohydrates, amino acids, lipids, and micronutrients are often considered when discussing preimplantation embryo nutrition, environmental oxygen is frequently overlooked. Although oxygen is not classically considered a nutrient, it is an important component of the in vitro culture environment and a critical regulator of cellular physiology. Oxygen is required to sustain an oxidative metabolism but when oxygen becomes limited, cells mount a physiological response driven by a family of transcription factors termed 'hypoxia inducible factors' which promote expression of a multitude of oxygen sensitive genes. It is this hypoxic response that is responsible not only for the switch to a glycolytic metabolism but also for a plethora of other cellular responses. There has been much debate in recent years over which environmental oxygen tension is preferential for the culture of preimplantation embryos. The review will evaluate this question and highlights how research using human embryonic stem cells can inform our understanding of why culturing under physiological oxygen tensions may be beneficial for the development of embryos generated through clinical in vitro fertilisation.


Assuntos
Blastocisto/fisiologia , Genitália Feminina/fisiologia , Células-Tronco Embrionárias Humanas/fisiologia , Hipóxia , Oxigênio/fisiologia , Feminino , Homeostase , Humanos , Saúde Reprodutiva
18.
Methods Mol Biol ; 2258: 119-130, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33340358

RESUMO

In the developing mammalian embryo, intercellular signaling allows cells to self-organize to create spatial patterns of different cell fates. This process is challenging to study because of the difficulty of observing or manipulating embryos on the spatial and temporal scales required. In vitro models can provide a complement to in vivo systems for addressing these issues. These models are also the only windows we have into early human development. Here we provide protocols for two systems based on differentiating human pluripotent stem cells in micropatterned colonies on defined size and shape. The first model replicates the patterning of the germ layers at gastrulation, while the second replicates the medial-lateral patterning of the ectoderm. These systems allow study of how signaling underlies self-organized patterning at stages of development which are otherwise inaccessible.


Assuntos
Diferenciação Celular , Linhagem da Célula , Ectoderma/fisiologia , Gastrulação , Células-Tronco Embrionárias Humanas/fisiologia , Comunicação Celular , Forma Celular , Tamanho Celular , Células Cultivadas , Ectoderma/citologia , Imunofluorescência , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Microscopia de Fluorescência , Transdução de Sinais , Fatores de Tempo
19.
Methods Mol Biol ; 2258: 193-203, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33340362

RESUMO

Neurally differentiating human pluripotent stem cells (hPSCs) possess the ability to self-organize into structures reminiscent of the developing fetal brain. In 2- and 3D cultures, this phenomenon initiates with formation of polarized areas of neural stem cells (NSCs), known as rosettes that resemble cross-sectional slices of the embryonic neural tube, i.e., the central nervous system (CNS) anlage. Thus, neural rosettes serve as an excellent starting point for bioengineering tissue models of all CNS tissues. Here, we provide detailed methods for bioengineering controlled induction of hPSC-derived neural assemblies with a biomimetic, singular neural rosette cytoarchitecture.


Assuntos
Células-Tronco Embrionárias Humanas/fisiologia , Células-Tronco Neurais/fisiologia , Tubo Neural/fisiologia , Neurogênese , Neurônios/fisiologia , Engenharia Tecidual , Materiais Biomiméticos , Técnicas de Cultura de Células , Linhagem Celular , Células-Tronco Embrionárias Humanas/metabolismo , Humanos , Imuno-Histoquímica , Microscopia , Morfogênese , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco Neurais/metabolismo , Tubo Neural/citologia , Tubo Neural/metabolismo , Neurônios/metabolismo , Esferoides Celulares
20.
Theranostics ; 10(26): 12204-12222, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33204338

RESUMO

Rationale: Mesenchymal stem cells (MSCs) show promising therapeutic potential in treating inflammatory bowel disease (IBD) due to their immunomodulatory and trophic functions. However, their efficacy is influenced by tissue origin, donator condition, isolation, and expansion methods. Here, we generated phenotypically uniform MSCs from human embryonic stem cells (T-MSCs) and explored the molecular mechanisms involved in promoting mucosal integrity and regeneration in colitis mice. Methods: T-MSCs were injected intravenously into mice with dextran sulfate sodium (DSS)-induced colitis, and the in vivo distribution and therapeutic efficacy were evaluated. We performed serum cytokine antibody microarrays to screen potentially effective proteins and examined the therapeutic effect of insulin-like growth factor-1 (IGF-1). Colon epithelial regeneration potential was evaluated, and RNA sequencing was employed to determine the underlying molecular mechanisms. Finally, in vitro IGF-1 stimulation was performed to assess its effect on cell functions and organoid growth. Results: Intravenous administration of T-MSCs alleviated colitis in both acute and chronic DSS mouse models. Labeled T-MSCs were mainly distributed in the lungs, liver, and spleen after systemic infusion. The antibody array analysis of serum cytokines indicated that the IGF-1 level was increased in the treatment group, and serum ELISA further confirmed its elevation in the regeneration stage. Intraperitoneal injection of IGF-1 receptor inhibitors abrogated the anti-inflammatory activity of T-MSCs. The colonic epithelium of the treatment group showed greater regenerative potency than the controls and the IGF1R-PI3K-AKT pathway was up-regulated. RNA sequencing showed that T-MSC treatment contributed to colonic cell integrity and promoted xenobiotic metabolism. In vitro IGF-1 stimulation promoted the growth and proliferation of colon cells and organoids. Conclusions: Intravenous infusion of T-MSCs alleviated colitis in mice by elevating the circulating IGF-1 level. Increased IGF-1 maintained the integrity of epithelial cells and contributed to their repair and regeneration. Our study has identified T- MSCs as a potential cell resource for IBD treatment.


Assuntos
Colite Ulcerativa/terapia , Células-Tronco Embrionárias Humanas/fisiologia , Fator de Crescimento Insulin-Like I/metabolismo , Transplante de Células-Tronco Mesenquimais/métodos , Regeneração/fisiologia , Animais , Diferenciação Celular , Colite Ulcerativa/sangue , Colite Ulcerativa/induzido quimicamente , Colite Ulcerativa/imunologia , Colo/efeitos dos fármacos , Colo/imunologia , Colo/patologia , Sulfato de Dextrana/administração & dosagem , Sulfato de Dextrana/toxicidade , Modelos Animais de Doenças , Humanos , Injeções Intravenosas , Fator de Crescimento Insulin-Like I/análise , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Masculino , Células-Tronco Mesenquimais/fisiologia , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...