Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
EMBO J ; 40(21): e107277, 2021 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-34558085

RESUMEN

The dorsal and ventral human telencephalons contain different neuronal subtypes, including glutamatergic, GABAergic, and cholinergic neurons, and how these neurons are generated during early development is not well understood. Using scRNA-seq and stringent validations, we reveal here a developmental roadmap for human telencephalic neurons. Both dorsal and ventral telencephalic radial glial cells (RGs) differentiate into neurons via dividing intermediate progenitor cells (IPCs_div) and early postmitotic neuroblasts (eNBs). The transcription factor ASCL1 plays a key role in promoting fate transition from RGs to IPCs_div in both regions. RGs from the regionalized neuroectoderm show heterogeneity, with restricted glutamatergic, GABAergic, and cholinergic differentiation potencies. During neurogenesis, IPCs_div gradually exit the cell cycle and branch into sister eNBs to generate distinct neuronal subtypes. Our findings highlight a general RGs-IPCs_div-eNBs developmental scheme for human telencephalic progenitors and support that the major neuronal fates of human telencephalon are predetermined during dorsoventral regionalization with neuronal diversity being further shaped during neurogenesis and neural circuit integration.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Linaje de la Célula/genética , Regulación del Desarrollo de la Expresión Génica , Neurogénesis/genética , Neuronas/metabolismo , Telencéfalo/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Ciclo Celular/genética , Diferenciación Celular , Colina/metabolismo , Proteína Doblecortina/genética , Proteína Doblecortina/metabolismo , Feto , Ontología de Genes , Glutamato Descarboxilasa/genética , Glutamato Descarboxilasa/metabolismo , Ácido Glutámico/metabolismo , Humanos , Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Anotación de Secuencia Molecular , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neuroglía/citología , Neuroglía/metabolismo , Neuronas/clasificación , Neuronas/citología , Factores de Transcripción SOXC/genética , Factores de Transcripción SOXC/metabolismo , Transducción de Señal , Estatmina/genética , Estatmina/metabolismo , Telencéfalo/citología , Telencéfalo/crecimiento & desarrollo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Ácido gamma-Aminobutírico/metabolismo
2.
Stem Cells ; 38(11): 1423-1437, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32930470

RESUMEN

Allogeneic immune rejection is a major barrier for the application of human pluripotent stem cells (hPSCs) in regenerative medicine. A broad spectrum of immune cells, including T cells, natural killer (NK) cells, and antigen-presenting cells, which either cause direct cell killing or constitute an immunogenic environment, are involved in allograft immune rejection. A strategy to protect donor cells from cytotoxicity while decreasing the secretion of inflammatory cytokines of lymphocytes is still lacking. Here, we engineered hPSCs with no surface expression of classical human leukocyte antigen (HLA) class I proteins via beta-2 microglobulin (B2M) knockout or biallelic knockin of HLA-G1 within the frame of endogenous B2M loci. Elimination of the surface expression of HLA class I proteins protected the engineered hPSCs from cytotoxicity mediated by T cells. However, this lack of surface expression also resulted in missing-self response and NK cell activation, which were largely compromised by expression of ß2m-HLA-G1 fusion proteins. We also proved that the engineered ß2m-HLA-G5 fusion proteins were soluble, secretable, and capable of safeguarding low immunogenic environments by lowering inflammatory cytokines secretion in allografts. Our current study reveals a novel strategy that may offer unique advantages to construct hypoimmunogenic hPSCs via the expression of membrane-bound and secreted ß2m-HLA-G fusion proteins. These engineered hPSCs are expected to serve as an unlimited cell source for generating universally compatible "off-the-shelf" cell grafts in the future.


Asunto(s)
Proteínas de la Fusión de la Membrana/metabolismo , Células Madre Pluripotentes/metabolismo , Animales , Técnicas de Cultivo de Célula , Humanos , Masculino , Ratones
3.
Cereb Cortex ; 27(5): 2941-2954, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-27226442

RESUMEN

The anteroposterior patterning of the central nervous system follows an activation/transformation model, which proposes that a prospective telencephalic fate will be activated by default during the neural induction stage, while this anterior fate could be transformed posteriorly according to caudalization morphogens. Although both extrinsic signals and intrinsic transcription factors have been implicated in dorsoventral (DV) specification of vertebrate telencephalon, the DV patterning model remains elusive. This is especially true in human considering its evolutionary trait and uniqueness of gene regulatory networks during neural induction. Here, we point to a model that human forebrain DV patterning also follows an activation/transformation paradigm. Human neuroectoderm (NE) will activate a forebrain dorsal fate automatically and this default anterior dorsal fate does not depend on Wnts activation or Pax6 expression. Forced expression of Pax6 in human NE hinders its ventralization even under sonic hedgehog (Shh) treatment, suggesting that the ventral fate is repressed by dorsal genes. Genetic manipulation of Nkx2.1, a key gene for forebrain ventral progenitors, shows that Nkx2.1 is neither necessary nor sufficient for Shh-driven ventralization. We thus propose that Shh represses dorsal genes of human NE and subsequently transforms the primitively activated dorsal fate ventrally in a repression release manner.


Asunto(s)
Tipificación del Cuerpo/fisiología , Diferenciación Celular/fisiología , Modelos Biológicos , Prosencéfalo/fisiología , Transducción de Señal/fisiología , Línea Celular , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Células Madre Embrionarias , Regulación del Desarrollo de la Expresión Génica/fisiología , Humanos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Prosencéfalo/citología , Prosencéfalo/embriología , ARN Mensajero/metabolismo , Factores de Tiempo , Transfección
4.
Cancer Med ; 12(17): 18078-18097, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37563971

RESUMEN

BACKGROUND: Ubiquitin-conjugating enzyme E2S (UBE2S), an E2 enzyme, is associated with the development of various tumors and exerts oncogenic activities. UBE2S is overexpressed in tumors, including hepatocellular carcinoma (HCC). However, the key molecular mechanisms of UBE2S in HCC still need additional research. The aim of this study was to explore the role of UBE2S in HCC. METHODS: The expression levels of UBE2S in HCC tissues and cells were detected by western blot analysis, quantitative real-time polymerase chain reaction analysis (qRT-PCR), and immunohistochemistry (IHC). A 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assay, wound healing assay, colony formation assay transwell assay, and animal models were used to detect the proliferation and migration ability of HCC cells. Western blot analysis, qRT-PCR, immunofluorescence, small-interfering RNA (siRNA), and plasmid transfection and coimmunoprecipitation (Co-IP) assays were performed to detect the interaction among UBE2S, von Hippel-Lindau (VHL), hypoxia-inducible factor 1-alpha (HIF-1α), Janus kinase-2 (JAK2), and signal transducer and activator of transcription 3 (STAT3). RESULTS: In this study, we found that high UBE2S expression was associated with poor prognosis in HCC patients. In addition, UBE2S expression was upregulated in HCC tissues and cell lines. Knockdown of UBE2S inhibited the proliferation and migration of HCC cells in vitro and in vivo by directly interacting with VHL to downregulate the HIF-1α and JAK2/STAT3 signaling pathways. Accordingly, overexpression of UBE2S significantly enhanced the proliferation and migration of HCC cells in vitro via VHL to upregulate HIF-1α and JAK2/STAT3 signaling pathways. Furthermore, we found that downregulation of UBE2S expression enhanced the sensitivity of HCC cells to sorafenib in vivo and in vitro. CONCLUSION: UBE2S enhances malignant properties via the VHL/HIF-1α and VHL/JAK2/STAT3 signaling pathways and reduces sensitivity to sorafenib in HCC. The findings of this study may open a new approach for HCC diagnosis and provide a potential option for the treatment of HCC.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Animales , Humanos , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Sorafenib/farmacología , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo
5.
Neurosci Bull ; 38(1): 81-94, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34460072

RESUMEN

Congenital hydrocephalus is a major neurological disorder with high rates of morbidity and mortality; however, the underlying cellular and molecular mechanisms remain largely unknown. Reproducible animal models mirroring both embryonic and postnatal hydrocephalus are also limited. Here, we describe a new mouse model of congenital hydrocephalus through knockout of ß-catenin in Nkx2.1-expressing regional neural progenitors. Progressive ventriculomegaly and an enlarged brain were consistently observed in knockout mice from embryonic day 12.5 through to adulthood. Transcriptome profiling revealed severe dysfunctions in progenitor maintenance in the ventricular zone and therefore in cilium biogenesis after ß-catenin knockout. Histological analyses also revealed an aberrant neuronal layout in both the ventral and dorsal telencephalon in hydrocephalic mice at both embryonic and postnatal stages. Thus, knockout of ß-catenin in regional neural progenitors leads to congenital hydrocephalus and provides a reproducible animal model for studying pathological changes and developing therapeutic interventions for this devastating disease.


Asunto(s)
Hidrocefalia , beta Catenina , Animales , Modelos Animales de Enfermedad , Hidrocefalia/genética , Ratones , Ratones Noqueados , Neuronas , beta Catenina/genética
6.
iScience ; 24(1): 101926, 2021 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-33385119

RESUMEN

Understanding the biological processes that determine the entry of three germ layers of human pluripotent stem cells (hPSCs) is a central question in developmental and stem cell biology. Here, we genetically engineered hPSCs with the germ layer reporter and inducible CRISPR/Cas9 knockout system, and a genome-scale screening was performed to define pathways restricting germ layer specification. Genes clustered in the key biological processes, including embryonic development, mRNA processing, metabolism, and epigenetic regulation, were centered in the governance of pluripotency and lineage development. Other than typical pluripotent transcription factors and signaling molecules, loss of function of mesendodermal specifiers resulted in advanced neuroectodermal differentiation, given their inter-germ layer antagonizing effect. Regarding the epigenetic superfamily, microRNAs enriched in hPSCs showed clear germ layer-targeting specificity. The cholesterol synthesis pathway maintained hPSCs via retardation of neuroectoderm specification. Thus, in this study, we identified a full landscape of genetic wiring and biological processes that control hPSC self-renewal and trilineage specification.

7.
Stem Cell Reports ; 12(5): 934-949, 2019 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-31056478

RESUMEN

Medial ganglionic eminence (MGE)-like cells yielded from human pluripotent stem cells (hPSCs) hold great potentials for cell therapies of related neurological disorders. However, cues that orchestrate the maintenance versus differentiation of human MGE progenitors, and ways for large-scale expansion of these cells have not been investigated. Here, we report that WNT/CTNNB1 signaling plays an essential role in maintaining MGE-like cells derived from hPSCs. Ablation of CTNNB1 in MGE cells led to precocious cell-cycle exit and advanced neuronal differentiation. Activation of WNT signaling through genetic or chemical approach was sufficient to maintain MGE cells in an expandable manner with authentic neuronal differentiation potencies through activation of endogenous NOTCH signaling. Our findings reveal that WNT/NOTCH signaling cascade is a key player in governing the maintenance versus terminal differentiation of MGE progenitors in humans. Large-scale expansion of functional MGE progenitors for cell therapies can therefore be achieved by modifying WNT/NOTCH pathway.


Asunto(s)
Proliferación Celular/fisiología , Eminencia Media/citología , Células Madre Pluripotentes/citología , Receptores Notch/metabolismo , Vía de Señalización Wnt/fisiología , Ciclo Celular/genética , Ciclo Celular/fisiología , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Línea Celular , Proliferación Celular/genética , Neuronas GABAérgicas/citología , Neuronas GABAérgicas/metabolismo , Perfilación de la Expresión Génica/métodos , Redes Reguladoras de Genes , Humanos , Interneuronas/citología , Interneuronas/metabolismo , Células Madre Pluripotentes/metabolismo , Receptores Notch/genética , Vía de Señalización Wnt/genética , beta Catenina/genética , beta Catenina/metabolismo
8.
Stem Cell Reports ; 11(5): 1257-1271, 2018 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-30449321

RESUMEN

It is highly desirable to specify human developmental principles in an appropriate human model with advanced genetic tools. However, genetically engineering human cells with lineage-tracing systems has not been achieved. Here we introduce strategies to construct lineage-tracing systems in human embryonic stem cells (hESCs). The AAVS1 locus was suitable for the integration of the conditional reporter. The Cre-LoxP and Flp-FRT systems were highly sensitive, which may cause inaccurate lineage labeling in human cells. The recombination sensitivity and tracing fidelity could be finely tuned by modification of the LoxP recombination site. Moreover, tamoxifen-controllable CreERT2-LoxP and FlpERT2-FRT systems showed compelling advantages in tightly tracing human lineages temporally. In proof-of-principle experiments, we traced human PAX6+ neuroectoderm cells and revealed their full neural lineage differentiation potency both in vitro and in vivo. Devising and optimizing of lineage-tracing systems in hESCs will thus set up a solid foundation for human developmental studies.


Asunto(s)
Linaje de la Célula , Ingeniería Genética , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/metabolismo , Animales , Secuencia de Bases , Línea Celular , Ectodermo/metabolismo , Sitios Genéticos , Humanos , Integrasas/metabolismo , Ratones Endogámicos NOD , Ratones SCID , Mutación/genética , Células-Madre Neurales/metabolismo , Factor de Transcripción PAX6/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Factores de Tiempo
9.
Cell Res ; 27(9): 1083-1099, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28675158

RESUMEN

Both environmental cues and intracellular bioenergetic states profoundly affect intracellular pH (pHi). How a cell responds to pHi changes to maintain bioenergetic homeostasis remains elusive. Here we show that Smad5, a well-characterized downstream component of bone morphogenetic protein (BMP) signaling responds to pHi changes. Cold, basic or hypertonic conditions increase pHi, which in turn dissociates protons from the charged amino acid clusters within the MH1 domain of Smad5, prompting its relocation from the nucleus to the cytoplasm. On the other hand, heat, acidic or hypotonic conditions decrease pHi, blocking the nuclear export of Smad5, and thus causing its nuclear accumulation. Active nucleocytoplasmic shuttling of Smad5 induced by environmental changes and pHi fluctuation is independent of BMP signaling, carboxyl terminus phosphorylation and Smad4. In addition, ablation of Smad5 causes chronic and irreversible dysregulation of cellular bioenergetic homeostasis and disrupted normal neural developmental processes as identified in a differentiation model of human pluripotent stem cells. Importantly, these metabolic and developmental deficits in Smad5-deficient cells could be rescued only by cytoplasmic Smad5. Cytoplasmic Smad5 physically interacts with hexokinase 1 and accelerates glycolysis. Together, our findings indicate that Smad5 acts as a pHi messenger and maintains the bioenergetic homeostasis of cells by regulating cytoplasmic metabolic machinery.


Asunto(s)
Metabolismo Energético , Homeostasis , Espacio Intracelular/metabolismo , Proteína Smad5/metabolismo , Transporte Activo de Núcleo Celular , Aminoácidos/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular , Núcleo Celular/metabolismo , Respiración de la Célula , Regulación hacia Abajo , Técnicas de Inactivación de Genes , Glucólisis , Células HEK293 , Hexoquinasa/metabolismo , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/metabolismo , Células Madre Embrionarias Humanas/ultraestructura , Humanos , Concentración de Iones de Hidrógeno , Carioferinas/metabolismo , Mitocondrias/metabolismo , Concentración Osmolar , Unión Proteica , Dominios Proteicos , Receptores Citoplasmáticos y Nucleares/metabolismo , Transducción de Señal , Proteína Smad5/química , Proteína Smad5/deficiencia , Relación Estructura-Actividad , Temperatura , Proteína Exportina 1
10.
Stem Cell Reports ; 7(5): 941-954, 2016 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-27720902

RESUMEN

Embryoid body (EB) formation and adherent culture (AD) paradigms are equivalently thought to be applicable for neural specification of human pluripotent stem cells. Here, we report that sonic hedgehog-induced ventral neuroprogenitors under EB conditions are fated to medial ganglionic eminence (MGE), while the AD cells mostly adopt a floor-plate (FP) fate. The EB-MGE later on differentiates into GABA and cholinergic neurons, while the AD-FP favors dopaminergic neuron specification. Distinct developmental, metabolic, and adhesion traits in AD and EB cells may potentially account for their differential patterning potency. Gene targeting combined with small-molecule screening experiments identified that concomitant inhibition of Wnts, STAT3, and p38 pathways (3i) could largely convert FP to MGE under AD conditions. Thus, differentiation paradigms and signaling regulators can be integrated together to specify distinct neuronal subtypes for studying and treating related neurological diseases, such as epilepsy, Alzheimer's disease, and Parkinson's disease.


Asunto(s)
Diferenciación Celular , Células-Madre Neurales/citología , Neuronas/citología , Células Madre Pluripotentes/citología , Biomarcadores , Proteínas Morfogenéticas Óseas/antagonistas & inhibidores , Adhesión Celular , Técnicas de Cultivo de Célula , Diferenciación Celular/genética , Línea Celular , Células Cultivadas , Metabolismo Energético , Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/metabolismo , Humanos , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Fenotipo , Células Madre Pluripotentes/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Transcripción Genética , Transcriptoma , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
11.
Sci Rep ; 6: 21602, 2016 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-26905010

RESUMEN

The transforming growth factor beta (TGFß) related signaling is one of the most important signaling pathways regulating early developmental events. Smad2 and Smad3 are structurally similar and it is mostly considered that they are equally important in mediating TGFß signals. Here, we show that Smad3 is an insensitive TGFß transducer as compared with Smad2. Smad3 preferentially localizes within the nucleus and is thus sequestered from membrane signaling. The ability of Smad3 in oligomerization with Smad4 upon agonist stimulation is also impaired given its unique linker region. Smad2 mediated TGFß signaling plays a crucial role in epiblast development and patterning of three germ layers. However, signaling unrelated nuclear localized Smad3 is dispensable for TGFß signaling-mediated epiblast specification, but important for early neural development, an event blocked by TGFß/Smad2 signaling. Both Smad2 and Smad3 bind to the conserved Smads binding element (SBE), but they show nonoverlapped target gene binding specificity and differential transcriptional activity. We conclude that Smad2 and Smad3 possess differential sensitivities in relaying TGFß signaling and have distinct roles in regulating early developmental events.


Asunto(s)
Proteína Smad2/fisiología , Proteína smad3/fisiología , Factor de Crecimiento Transformador beta/fisiología , Transporte Activo de Núcleo Celular , Animales , Secuencia de Bases , Tipificación del Cuerpo , Carcinogénesis/metabolismo , Linaje de la Célula , Núcleo Celular/metabolismo , Secuencia Conservada , Células HEK293 , Células Madre Embrionarias Humanas/metabolismo , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Unión Proteica , Multimerización de Proteína , Elementos de Respuesta , Transducción de Señal , Teratoma/metabolismo , Teratoma/patología
12.
Stem Cell Reports ; 7(3): 496-507, 2016 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-27594587

RESUMEN

Loss-of-function studies in human pluripotent stem cells (hPSCs) require efficient methodologies for lesion of genes of interest. Here, we introduce a donor-free paired gRNA-guided CRISPR/Cas9 knockout strategy (paired-KO) for efficient and rapid gene ablation in hPSCs. Through paired-KO, we succeeded in targeting all genes of interest with high biallelic targeting efficiencies. More importantly, during paired-KO, the cleaved DNA was repaired mostly through direct end joining without insertions/deletions (precise ligation), and thus makes the lesion product predictable. The paired-KO remained highly efficient for one-step targeting of multiple genes and was also efficient for targeting of microRNA, while for long non-coding RNA over 8 kb, cleavage of a short fragment of the core promoter region was sufficient to eradicate downstream gene transcription. This work suggests that the paired-KO strategy is a simple and robust system for loss-of-function studies for both coding and non-coding genes in hPSCs.


Asunto(s)
Sistemas CRISPR-Cas , Técnicas de Inactivación de Genes , Células Madre Pluripotentes/metabolismo , Secuencia de Bases , Diferenciación Celular , Línea Celular , Células Madre Embrionarias , Marcación de Gen , Humanos , Células Madre Pluripotentes/citología , ARN Guía de Kinetoplastida , ARN Largo no Codificante/genética , Proteína smad3/deficiencia , Proteína smad3/genética , beta Catenina/deficiencia , beta Catenina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA