Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Rep ; 43(4): 113999, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38554281

RESUMO

Motor neuron (MN) demise is a hallmark of several neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). Post-transcriptional gene regulation can control RNA's fate, and defects in RNA processing are critical determinants of MN degeneration. N6-methyladenosine (m6A) is a post-transcriptional RNA modification that controls diverse aspects of RNA metabolism. To assess the m6A requirement in MNs, we depleted the m6A methyltransferase-like 3 (METTL3) in cells and mice. METTL3 depletion in embryonic stem cell-derived MNs has profound and selective effects on survival and neurite outgrowth. Mice with cholinergic neuron-specific METTL3 depletion display a progressive decline in motor behavior, accompanied by MN loss and muscle denervation, culminating in paralysis and death. Reader proteins convey m6A effects, and their silencing phenocopies METTL3 depletion. Among the m6A targets, we identified transactive response DNA-binding protein 43 (TDP-43) and discovered that its expression is under epitranscriptomic control. Thus, impaired m6A signaling disrupts MN homeostasis and triggers neurodegeneration conceivably through TDP-43 deregulation.


Assuntos
Neurônios Colinérgicos , Metiltransferases , Doenças Neuromusculares , Animais , Humanos , Camundongos , Adenosina/metabolismo , Adenosina/análogos & derivados , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/patologia , Esclerose Lateral Amiotrófica/genética , Neurônios Colinérgicos/metabolismo , Neurônios Colinérgicos/patologia , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/genética , Metiltransferases/metabolismo , Metiltransferases/genética , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Doenças Neuromusculares/metabolismo , Doenças Neuromusculares/patologia
2.
Nature ; 622(7983): 562-573, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37673118

RESUMO

The ability to study human post-implantation development remains limited owing to ethical and technical challenges associated with intrauterine development after implantation1. Embryo-like models with spatially organized morphogenesis and structure of all defining embryonic and extra-embryonic tissues of the post-implantation human conceptus (that is, the embryonic disc, the bilaminar disc, the yolk sac, the chorionic sac and the surrounding trophoblast layer) remain lacking1,2. Mouse naive embryonic stem cells have recently been shown to give rise to embryonic and extra-embryonic stem cells capable of self-assembling into post-gastrulation structured stem-cell-based embryo models with spatially organized morphogenesis (called SEMs)3. Here we extend those findings to humans using only genetically unmodified human naive embryonic stem cells (cultured in human enhanced naive stem cell medium conditions)4. Such human fully integrated and complete SEMs recapitulate the organization of nearly all known lineages and compartments of post-implantation human embryos, including the epiblast, the hypoblast, the extra-embryonic mesoderm and the trophoblast layer surrounding the latter compartments. These human complete SEMs demonstrated developmental growth dynamics that resemble key hallmarks of post-implantation stage embryogenesis up to 13-14 days after fertilization (Carnegie stage 6a). These include embryonic disc and bilaminar disc formation, epiblast lumenogenesis, polarized amniogenesis, anterior-posterior symmetry breaking, primordial germ-cell specification, polarized yolk sac with visceral and parietal endoderm formation, extra-embryonic mesoderm expansion that defines a chorionic cavity and a connecting stalk, and a trophoblast-surrounding compartment demonstrating syncytium and lacunae formation. This SEM platform will probably enable the experimental investigation of previously inaccessible windows of human early post implantation up to peri-gastrulation development.


Assuntos
Implantação do Embrião , Embrião de Mamíferos , Desenvolvimento Embrionário , Células-Tronco Embrionárias Humanas , Humanos , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Fertilização , Gastrulação , Camadas Germinativas/citologia , Camadas Germinativas/embriologia , Células-Tronco Embrionárias Humanas/citologia , Trofoblastos/citologia , Saco Vitelino/citologia , Saco Vitelino/embriologia , Células Gigantes/citologia
3.
Mol Cell ; 2023 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-37657444

RESUMO

N6-methyladenosine (m6A) RNA modification plays important roles in the governance of gene expression and is temporally regulated in different cell states. In contrast to global m6A profiling in bulk sequencing, single-cell technologies for analyzing m6A heterogeneity are not extensively established. Here, we developed single-nucleus m6A-CUT&Tag (sn-m6A-CT) for simultaneous profiling of m6A methylomes and transcriptomes within a single nucleus using mouse embryonic stem cells (mESCs). m6A-CT is capable of enriching m6A-marked RNA molecules in situ, without isolating RNAs from cells. We adapted m6A-CT to the droplet-based single-cell omics platform and demonstrated high-throughput performance in analyzing nuclei isolated from thousands of cells from various cell types. We show that sn-m6A-CT profiling is sufficient to determine cell identity and allows the generation of cell-type-specific m6A methylome landscapes from heterogeneous populations. These indicate that sn-m6A-CT provides additional dimensions to multimodal datasets and insights into epitranscriptomic landscape in defining cell fate identity and states.

4.
Nat Immunol ; 24(8): 1256-1264, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37400674

RESUMO

Innate lymphoid cells (ILCs) can quickly switch from a quiescent state to an active state and rapidly produce effector molecules that provide critical early immune protection. How the post-transcriptional machinery processes different stimuli and initiates robust gene expression in ILCs is poorly understood. Here, we show that deletion of the N6-methyladenosine (m6A) writer protein METTL3 has little impact on ILC homeostasis or cytokine-induced ILC1 or ILC3 responses but significantly diminishes ILC2 proliferation, migration and effector cytokine production and results in impaired antihelminth immunity. m6A RNA modification supports an increase in cell size and transcriptional activity in activated ILC2s but not in ILC1s or ILC3s. Among other transcripts, the gene encoding the transcription factor GATA3 is highly m6A methylated in ILC2s. Targeted m6A demethylation destabilizes nascent Gata3 mRNA and abolishes the upregulation of GATA3 and ILC2 activation. Our study suggests a lineage-specific requirement of m6A for ILC2 responses.


Assuntos
Imunidade Inata , Linfócitos , Citocinas/metabolismo , Homeostase , Imunidade Inata/genética , Imunidade Inata/imunologia , Linfócitos/imunologia , RNA/metabolismo , Animais , Camundongos
5.
Nat Commun ; 14(1): 3293, 2023 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-37280197

RESUMO

Lissencephaly-1 (LIS1) is associated with neurodevelopmental diseases and is known to regulate the molecular motor cytoplasmic dynein activity. Here we show that LIS1 is essential for the viability of mouse embryonic stem cells (mESCs), and it governs the physical properties of these cells. LIS1 dosage substantially affects gene expression, and we uncovered an unexpected interaction of LIS1 with RNA and RNA-binding proteins, most prominently the Argonaute complex. We demonstrate that LIS1 overexpression partially rescued the extracellular matrix (ECM) expression and mechanosensitive genes conferring stiffness to Argonaute null mESCs. Collectively, our data transforms the current perspective on the roles of LIS1 in post-transcriptional regulation underlying development and mechanosensitive processes.


Assuntos
1-Alquil-2-acetilglicerofosfocolina Esterase , Proteínas Argonautas , Células-Tronco Embrionárias , Proteínas Associadas aos Microtúbulos , Animais , Camundongos , 1-Alquil-2-acetilglicerofosfocolina Esterase/metabolismo , Blastocisto/citologia , Blastocisto/metabolismo , Sobrevivência Celular , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Células-Tronco Pluripotentes , Mapas de Interação de Proteínas , Proteínas Argonautas/metabolismo
6.
Stem Cell Reports ; 17(11): 2484-2500, 2022 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-36270280

RESUMO

The recent derivation of human trophoblast stem cells (TSCs) from placental cytotrophoblasts and blastocysts opened opportunities for studying the development and function of the human placenta. Recent reports have suggested that human naïve, but not primed, pluripotent stem cells (PSCs) retain an exclusive potential to generate TSCs. Here we report that, in the absence of WNT stimulation, transforming growth factor ß (TGF-ß) pathway inhibition leads to direct and robust conversion of primed human PSCs into TSCs. The resulting primed PSC-derived TSC lines exhibit self-renewal, can differentiate into the main trophoblast lineages, and present RNA and epigenetic profiles that are indistinguishable from recently established TSC lines derived from human placenta, blastocysts, or isogenic human naïve PSCs expanded under human enhanced naïve stem cell medium (HENSM) conditions. Activation of nuclear Yes-associated protein (YAP) signaling is sufficient for this conversion and necessary for human TSC maintenance. Our findings underscore a residual plasticity in primed human PSCs that allows their in vitro conversion into extra-embryonic trophoblast lineages.


Assuntos
Células-Tronco Pluripotentes , Trofoblastos , Feminino , Humanos , Gravidez , Blastocisto , Diferenciação Celular , Placenta , Células-Tronco Pluripotentes/metabolismo
7.
Curr Opin Genet Dev ; 77: 101988, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36179582

RESUMO

Research on early postimplantation mammalian development has been limited by the small size and intrauterine confinement of the developing embryos. Owing to the inability to observe and manipulate living embryos at these stages in utero, the establishment of robust ex utero embryo-culture systems that capture prolonged periods of mouse development has been an important research goal. In the last few years, these methods have been significantly improved by the optimization and enhancement of in vitro culture systems sustaining embryo development during peri-implantation stages for several species, and more recently, proper growth of natural mouse embryos from pregastrulation to late organogenesis stages and of embryonic stem cell (ES)-derived synthetic embryo models until early organogenesis stages. Here, we discuss the most recent ex utero embryo-culture systems established to date for rodents, nonhuman primates, and humans. We emphasize their technical aspects and developmental timeframe and provide insights into the new opportunities that these methods will contribute to the study of natural and synthetic mammalian embryogenesis and the stem-cell field.


Assuntos
Embrião de Mamíferos , Desenvolvimento Embrionário , Humanos , Camundongos , Animais , Desenvolvimento Embrionário/genética , Implantação do Embrião , Organogênese , Roedores
8.
Nature ; 610(7930): 143-153, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36007540

RESUMO

Embryonic stem (ES) cells can undergo many aspects of mammalian embryogenesis in vitro1-5, but their developmental potential is substantially extended by interactions with extraembryonic stem cells, including trophoblast stem (TS) cells, extraembryonic endoderm stem (XEN) cells and inducible XEN (iXEN) cells6-11. Here we assembled stem cell-derived embryos in vitro from mouse ES cells, TS cells and iXEN cells and showed that they recapitulate the development of whole natural mouse embryo in utero up to day 8.5 post-fertilization. Our embryo model displays headfolds with defined forebrain and midbrain regions and develops a beating heart-like structure, a trunk comprising a neural tube and somites, a tail bud containing neuromesodermal progenitors, a gut tube, and primordial germ cells. This complete embryo model develops within an extraembryonic yolk sac that initiates blood island development. Notably, we demonstrate that the neurulating embryo model assembled from Pax6-knockout ES cells aggregated with wild-type TS cells and iXEN cells recapitulates the ventral domain expansion of the neural tube that occurs in natural, ubiquitous Pax6-knockout embryos. Thus, these complete embryoids are a powerful in vitro model for dissecting the roles of diverse cell lineages and genes in development. Our results demonstrate the self-organization ability of ES cells and two types of extraembryonic stem cells to reconstitute mammalian development through and beyond gastrulation to neurulation and early organogenesis.


Assuntos
Embrião de Mamíferos , Gastrulação , Modelos Biológicos , Neurulação , Organogênese , Animais , Linhagem da Célula , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Células-Tronco Embrionárias/citologia , Endoderma/citologia , Endoderma/embriologia , Coração/embriologia , Mesencéfalo/embriologia , Camundongos , Tubo Neural/embriologia , Fator de Transcrição PAX6/deficiência , Fator de Transcrição PAX6/genética , Prosencéfalo/embriologia , Somitos/embriologia
9.
Cell ; 185(17): 3070-3072, 2022 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-35985282

RESUMO

Cell asked LGBTQ+ scientists around the world about how their identity shapes their experiences in STEM. Here we share six unique perspectives of researchers highlighting how their area of expertise, research focus, institutions, and geographical location have played a role in this regard. We thank them for sharing their voices and continued efforts toward making science more inclusive.


Assuntos
Pesquisadores , Humanos
10.
Cell ; 185(18): 3290-3306.e25, 2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-35988542

RESUMO

In vitro cultured stem cells with distinct developmental capacities can contribute to embryonic or extraembryonic tissues after microinjection into pre-implantation mammalian embryos. However, whether cultured stem cells can independently give rise to entire gastrulating embryo-like structures with embryonic and extraembryonic compartments remains unknown. Here, we adapt a recently established platform for prolonged ex utero growth of natural embryos to generate mouse post-gastrulation synthetic whole embryo models (sEmbryos), with both embryonic and extraembryonic compartments, starting solely from naive ESCs. This was achieved by co-aggregating non-transduced ESCs, with naive ESCs transiently expressing Cdx2 or Gata4 to promote their priming toward trophectoderm and primitive endoderm lineages, respectively. sEmbryos adequately accomplish gastrulation, advance through key developmental milestones, and develop organ progenitors within complex extraembryonic compartments similar to E8.5 stage mouse embryos. Our findings highlight the plastic potential of naive pluripotent cells to self-organize and functionally reconstitute and model the entire mammalian embryo beyond gastrulation.


Assuntos
Células-Tronco Embrionárias , Gastrulação , Animais , Diferenciação Celular/fisiologia , Embrião de Mamíferos/fisiologia , Desenvolvimento Embrionário , Endoderma , Mamíferos , Camundongos
11.
Cell Rep ; 39(5): 110778, 2022 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-35508130

RESUMO

Antibody-mediated immunity is initiated by B cell differentiation into multiple cell subsets, including plasmablast, memory, and germinal center (GC) cells. B cell differentiation trajectories are determined by transcription factors, yet very few mechanisms that specifically determine early B cell fates have been described. Here, we report a post-transcriptional mechanism that suppresses the plasmablast genetic program and promotes GC B cell fate commitment. Single-cell RNA-sequencing analysis reveals that antigen-specific B cell precursors at the pre-GC stage upregulate YTHDF2, which enhances the decay of methylated transcripts. Ythdf2-deficient B cells exhibit intact proliferation and activation, whereas differentiation into GC B cells is blocked. Mechanistically, B cells require YTHDF2 to attenuate the plasmablast genetic program during GC seeding, and transcripts of key plasmablast-regulating genes are methylated and bound by YTHDF2. Collectively, this study reveals how post-transcriptional suppression of gene expression directs appropriate B cell fate commitment during initiation of the adaptive immune response.


Assuntos
Centro Germinativo , Plasmócitos , Linfócitos B , Ativação Linfocitária , Fatores de Transcrição/metabolismo
12.
Mol Cell ; 82(1): 106-122.e9, 2022 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-34875212

RESUMO

The fidelity of the early embryonic program is underlined by tight regulation of the chromatin. Yet, how the chromatin is organized to prohibit the reversal of the developmental program remains unclear. Specifically, the totipotency-to-pluripotency transition marks one of the most dramatic events to the chromatin, and yet, the nature of histone alterations underlying this process is incompletely characterized. Here, we show that linker histone H1 is post-translationally modulated by SUMO2/3, which facilitates its fixation onto ultra-condensed heterochromatin in embryonic stem cells (ESCs). Upon SUMOylation depletion, the chromatin becomes de-compacted and H1 is evicted, leading to totipotency reactivation. Furthermore, we show that H1 and SUMO2/3 jointly mediate the repression of totipotent elements. Lastly, we demonstrate that preventing SUMOylation on H1 abrogates its ability to repress the totipotency program in ESCs. Collectively, our findings unravel a critical role for SUMOylation of H1 in facilitating chromatin repression and desolation of the totipotent identity.


Assuntos
Blastocisto/metabolismo , Linhagem da Célula , Montagem e Desmontagem da Cromatina , Cromatina/metabolismo , Histonas/metabolismo , Células-Tronco Embrionárias Murinas/metabolismo , Animais , Blastocisto/citologia , Cromatina/genética , Técnicas de Cultura Embrionária , Desenvolvimento Embrionário , Regulação da Expressão Gênica no Desenvolvimento , Células HEK293 , Histonas/genética , Humanos , Camundongos , Fenótipo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/genética , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Sumoilação , Ubiquitinas/genética , Ubiquitinas/metabolismo
13.
Cell Rep ; 37(11): 110124, 2021 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-34910919

RESUMO

Regulatory T (Treg) cells play crucial roles in suppressing deleterious immune response. Here, we investigate how Treg cells are mechanistically induced in vitro (iTreg) and stabilized via transcriptional regulation of Treg lineage-specifying factor Foxp3. We find that acetylation of histone tails at the Foxp3 promoter is required for inducing Foxp3 transcription. Upon induction, histone acetylation signals via bromodomain-containing proteins, particularly targets of inhibitor JQ1, and sustains Foxp3 transcription via a global or trans effect. Subsequently, Tet-mediated DNA demethylation of Foxp3 cis-regulatory elements, mainly enhancer CNS2, increases chromatin accessibility and protein binding, stabilizing Foxp3 transcription and obviating the need for the histone acetylation signal. These processes transform stochastic iTreg induction into a stable cell fate, with the former sensitive and the latter resistant to genetic and environmental perturbations. Thus, sequential histone acetylation and DNA demethylation in Foxp3 induction and maintenance reflect stepwise mechanical switches governing iTreg cell lineage specification.


Assuntos
Desmetilação do DNA , Proteínas de Ligação a DNA/fisiologia , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica , Histonas/química , Proteínas Proto-Oncogênicas/fisiologia , Linfócitos T Reguladores/imunologia , Acetilação , Animais , Diferenciação Celular , Metilação de DNA , Feminino , Fatores de Transcrição Forkhead/genética , Histonas/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Regiões Promotoras Genéticas , Sequências Reguladoras de Ácido Nucleico
14.
J Vis Exp ; (176)2021 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-34747408

RESUMO

Postimplantation mammalian embryo culture methods have been generally inefficient and limited to brief periods after dissection out of the uterus. Platforms have been recently developed for highly robust and prolonged ex utero culture of mouse embryos from egg-cylinder stages until advanced organogenesis. These platforms enable appropriate and faithful development of pregastrulating embryos (E5.5) until the hind limb formation stage (E11). Late gastrulating embryos (E7.5) are grown in rotating bottles in these settings, while extended culture from pregastrulation stages (E5.5 or E6.5) requires a combination of static and rotating bottle cultures. In addition, sensitive regulation of O2 and CO2 concentration, gas pressure, glucose levels, and the use of a specific ex utero culture medium are critical for proper embryo development. Here, a detailed step-by-step protocol for extended ex utero mouse embryo culture is provided. The ability to grow normal mouse embryos ex utero from gastrulation to organogenesis represents a valuable tool for characterizing the effect of different experimental perturbations during embryonic development.


Assuntos
Técnicas de Cultura Embrionária , Organogênese , Animais , Técnicas de Cultura Embrionária/métodos , Embrião de Mamíferos/fisiologia , Desenvolvimento Embrionário , Feminino , Gastrulação , Mamíferos , Camundongos , Gravidez
15.
Mol Cell ; 81(19): 3949-3964.e7, 2021 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-34450044

RESUMO

Immunoglobulin heavy chain (IgH) locus-associated G-rich long noncoding RNA (SµGLT) is important for physiological and pathological B cell DNA recombination. We demonstrate that the METTL3 enzyme-catalyzed N6-methyladenosine (m6A) RNA modification drives recognition and 3' end processing of SµGLT by the RNA exosome, promoting class switch recombination (CSR) and suppressing chromosomal translocations. The recognition is driven by interaction of the MPP6 adaptor protein with nuclear m6A reader YTHDC1. MPP6 and YTHDC1 promote CSR by recruiting AID and the RNA exosome to actively transcribe SµGLT. Direct suppression of m6A modification of SµGLT or of m6A reader YTHDC1 reduces CSR. Moreover, METTL3, an essential gene for B cell development in the bone marrow and germinal center, suppresses IgH-associated aberrant DNA breaks and prevents genomic instability. Taken together, we propose coordinated and central roles for MPP6, m6A modification, and m6A reader proteins in controlling long noncoding RNA processing, DNA recombination, and development in B cells.


Assuntos
Adenosina/análogos & derivados , Linfócitos B/metabolismo , Complexo Multienzimático de Ribonucleases do Exossomo/metabolismo , Cadeias Pesadas de Imunoglobulinas/metabolismo , Processamento de Terminações 3' de RNA , RNA Longo não Codificante/metabolismo , Recombinação Genética , Adenosina/metabolismo , Animais , Linfócitos B/imunologia , Citidina Desaminase/genética , Citidina Desaminase/metabolismo , Complexo Multienzimático de Ribonucleases do Exossomo/genética , Feminino , Instabilidade Genômica , Células HEK293 , Humanos , Switching de Imunoglobulina , Cadeias Pesadas de Imunoglobulinas/genética , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Metilação , Metiltransferases/genética , Metiltransferases/metabolismo , Camundongos Knockout , RNA Longo não Codificante/genética , RNA não Traduzido/genética , RNA não Traduzido/metabolismo
16.
J Exp Med ; 218(10)2021 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-34402854

RESUMO

Long-lasting immunity depends on the generation of protective antibodies through the germinal center (GC) reaction. N6-methyladenosine (m6A) modification of mRNAs by METTL3 activity modulates transcript lifetime primarily through the function of m6A readers; however, the physiological role of this molecular machinery in the GC remains unknown. Here, we show that m6A modifications by METTL3 are required for GC maintenance through the differential functions of m6A readers. Mettl3-deficient GC B cells exhibited reduced cell-cycle progression and decreased expression of proliferation- and oxidative phosphorylation-related genes. The m6A binder, IGF2BP3, was required for stabilization of Myc mRNA and expression of its target genes, whereas the m6A reader, YTHDF2, indirectly regulated the expression of the oxidative phosphorylation gene program. Our findings demonstrate how two independent gene networks that support critical GC functions are modulated by m6A through distinct mRNA binders.


Assuntos
Centro Germinativo/fisiologia , Metiltransferases/metabolismo , RNA/metabolismo , Adenosina/análogos & derivados , Adenosina/genética , Adenosina/metabolismo , Animais , Linfócitos B/patologia , Ciclo Celular/genética , Regulação da Expressão Gênica , Genes myc , Centro Germinativo/patologia , Metilação , Metiltransferases/genética , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fosforilação Oxidativa , RNA/genética , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Smegmamorpha , Baço/patologia
17.
EMBO Mol Med ; 13(8): e13610, 2021 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-34268881

RESUMO

Developmental and epileptic encephalopathies (DEE) are a group of disorders associated with intractable seizures, brain development, and functional abnormalities, and in some cases, premature death. Pathogenic human germline biallelic mutations in tumor suppressor WW domain-containing oxidoreductase (WWOX) are associated with a relatively mild autosomal recessive spinocerebellar ataxia-12 (SCAR12) and a more severe early infantile WWOX-related epileptic encephalopathy (WOREE). In this study, we generated an in vitro model for DEEs, using the devastating WOREE syndrome as a prototype, by establishing brain organoids from CRISPR-engineered human ES cells and from patient-derived iPSCs. Using these models, we discovered dramatic cellular and molecular CNS abnormalities, including neural population changes, cortical differentiation malfunctions, and Wnt pathway and DNA damage response impairment. Furthermore, we provide a proof of concept that ectopic WWOX expression could potentially rescue these phenotypes. Our findings underscore the utility of modeling childhood epileptic encephalopathies using brain organoids and their use as a unique platform to test possible therapeutic intervention strategies.


Assuntos
Encefalopatias , Espasmos Infantis , Encéfalo , Criança , Humanos , Mutação , Organoides
18.
Cell Stem Cell ; 28(9): 1549-1565.e12, 2021 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-33915080

RESUMO

Isolating human MEK/ERK signaling-independent pluripotent stem cells (PSCs) with naive pluripotency characteristics while maintaining differentiation competence and (epi)genetic integrity remains challenging. Here, we engineer reporter systems that allow the screening for defined conditions that induce molecular and functional features of human naive pluripotency. Synergistic inhibition of WNT/ß-CATENIN, protein kinase C (PKC), and SRC signaling consolidates the induction of teratoma-competent naive human PSCs, with the capacity to differentiate into trophoblast stem cells (TSCs) and extraembryonic naive endodermal (nEND) cells in vitro. Divergent signaling and transcriptional requirements for boosting naive pluripotency were found between mouse and human. P53 depletion in naive hPSCs increased their contribution to mouse-human cross-species chimeric embryos upon priming and differentiation. Finally, MEK/ERK inhibition can be substituted with the inhibition of NOTCH/RBPj, which induces alternative naive-like hPSCs with a diminished risk for deleterious global DNA hypomethylation. Our findings set a framework for defining the signaling foundations of human naive pluripotency.


Assuntos
Células-Tronco Pluripotentes , Animais , Diferenciação Celular , Embrião de Mamíferos , Humanos , Camundongos , Transdução de Sinais , Trofoblastos
19.
Nature ; 593(7857): 119-124, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33731940

RESUMO

The mammalian body plan is established shortly after the embryo implants into the maternal uterus, and our understanding of post-implantation developmental processes remains limited. Although pre- and peri-implantation mouse embryos are routinely cultured in vitro1,2, approaches for the robust culture of post-implantation embryos from egg cylinder stages until advanced organogenesis remain to be established. Here we present highly effective platforms for the ex utero culture of post-implantation mouse embryos, which enable the appropriate development of embryos from before gastrulation (embryonic day (E) 5.5) until the hindlimb formation stage (E11). Late gastrulating embryos (E7.5) are grown in three-dimensional rotating bottles, whereas extended culture from pre-gastrulation stages (E5.5 or E6.5) requires a combination of static and rotating bottle culture platforms. Histological, molecular and single-cell RNA sequencing analyses confirm that the ex utero cultured embryos recapitulate in utero development precisely. This culture system is amenable to the introduction of a variety of embryonic perturbations and micro-manipulations, the results of which can be followed ex utero for up to six days. The establishment of a system for robustly growing normal mouse embryos ex utero from pre-gastrulation to advanced organogenesis represents a valuable tool for investigating embryogenesis, as it eliminates the uterine barrier and allows researchers to mechanistically interrogate post-implantation morphogenesis and artificial embryogenesis in mammals.


Assuntos
Técnicas de Cultura Embrionária , Embrião de Mamíferos/embriologia , Desenvolvimento Embrionário , Técnicas In Vitro , Organogênese , Animais , Técnicas de Cultura Embrionária/métodos , Embrião de Mamíferos/citologia , Feminino , Gastrulação , Masculino , Camundongos , Fatores de Tempo , Útero
20.
Methods Mol Biol ; 2195: 125-145, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32852762

RESUMO

Primordial germ cells (PGCs) are common ancestors of all germline cells. In mammals, PGCs emerge in early-stage embryos around the timing of gastrulation at or near epiblast, and specification of PGCs from their precursor cells involves multiple growth factors secreted by adjacent cells. Recent advancements in germline stem cell biology have made it possible to generate PGC-like cell culture models (PGCLCs for PGC-like cells) from human and mouse pluripotent stem cells by mimicking the embryonic growth factor environment in vitro. Here we describe a method of producing human PGCLCs from primed-pluripotency induced pluripotent stem cells (iPSCs) via temporal conversion to naive pluripotency followed by formation of embryoid bodies (EBs) using the spin-EB method.


Assuntos
Técnicas de Cultura de Células , Gástrula/citologia , Células Germinativas/citologia , Células Germinativas/metabolismo , Camadas Germinativas/citologia , Imuno-Histoquímica , Imunofenotipagem , Biomarcadores , Técnicas de Cultura de Células/métodos , Diferenciação Celular , Autorrenovação Celular , Células Cultivadas , Corpos Embrioides/citologia , Humanos , Imuno-Histoquímica/métodos , Imunofenotipagem/métodos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...