Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 18(5): 552-562, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28346408

RESUMEN

Gut dysbiosis might underlie the pathogenesis of type 1 diabetes. In mice of the non-obese diabetic (NOD) strain, we found that key features of disease correlated inversely with blood and fecal concentrations of the microbial metabolites acetate and butyrate. We therefore fed NOD mice specialized diets designed to release large amounts of acetate or butyrate after bacterial fermentation in the colon. Each diet provided a high degree of protection from diabetes, even when administered after breakdown of immunotolerance. Feeding mice a combined acetate- and butyrate-yielding diet provided complete protection, which suggested that acetate and butyrate might operate through distinct mechanisms. Acetate markedly decreased the frequency of autoreactive T cells in lymphoid tissues, through effects on B cells and their ability to expand populations of autoreactive T cells. A diet containing butyrate boosted the number and function of regulatory T cells, whereas acetate- and butyrate-yielding diets enhanced gut integrity and decreased serum concentration of diabetogenic cytokines such as IL-21. Medicinal foods or metabolites might represent an effective and natural approach for countering the numerous immunological defects that contribute to T cell-dependent autoimmune diseases.


Asunto(s)
Acetatos/metabolismo , Linfocitos B/inmunología , Butiratos/metabolismo , Colon/metabolismo , Diabetes Mellitus Tipo 1/dietoterapia , Disbiosis/dietoterapia , Linfocitos T Reguladores/inmunología , Animales , Autoinmunidad , Linfocitos B/microbiología , Células Cultivadas , Colon/patología , Dietoterapia , Microbioma Gastrointestinal , Interleucinas/sangre , Ratones , Ratones Endogámicos NOD , Linfocitos T Reguladores/microbiología
3.
Nature ; 620(7975): 863-872, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37587336

RESUMEN

Cells undergo a major epigenome reconfiguration when reprogrammed to human induced pluripotent stem cells (hiPS cells). However, the epigenomes of hiPS cells and human embryonic stem (hES) cells differ significantly, which affects hiPS cell function1-8. These differences include epigenetic memory and aberrations that emerge during reprogramming, for which the mechanisms remain unknown. Here we characterized the persistence and emergence of these epigenetic differences by performing genome-wide DNA methylation profiling throughout primed and naive reprogramming of human somatic cells to hiPS cells. We found that reprogramming-induced epigenetic aberrations emerge midway through primed reprogramming, whereas DNA demethylation begins early in naive reprogramming. Using this knowledge, we developed a transient-naive-treatment (TNT) reprogramming strategy that emulates the embryonic epigenetic reset. We show that the epigenetic memory in hiPS cells is concentrated in cell of origin-dependent repressive chromatin marked by H3K9me3, lamin-B1 and aberrant CpH methylation. TNT reprogramming reconfigures these domains to a hES cell-like state and does not disrupt genomic imprinting. Using an isogenic system, we demonstrate that TNT reprogramming can correct the transposable element overexpression and differential gene expression seen in conventional hiPS cells, and that TNT-reprogrammed hiPS and hES cells show similar differentiation efficiencies. Moreover, TNT reprogramming enhances the differentiation of hiPS cells derived from multiple cell types. Thus, TNT reprogramming corrects epigenetic memory and aberrations, producing hiPS cells that are molecularly and functionally more similar to hES cells than conventional hiPS cells. We foresee TNT reprogramming becoming a new standard for biomedical and therapeutic applications and providing a novel system for studying epigenetic memory.


Asunto(s)
Reprogramación Celular , Epigénesis Genética , Células Madre Pluripotentes Inducidas , Humanos , Cromatina/genética , Cromatina/metabolismo , Desmetilación del ADN , Metilación de ADN , Elementos Transponibles de ADN , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/metabolismo , Lamina Tipo B
4.
Cell ; 151(7): 1617-32, 2012 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-23260147

RESUMEN

Factor-induced reprogramming of somatic cells into induced pluripotent stem cells (iPSCs) is inefficient, complicating mechanistic studies. Here, we examined defined intermediate cell populations poised to becoming iPSCs by genome-wide analyses. We show that induced pluripotency elicits two transcriptional waves, which are driven by c-Myc/Klf4 (first wave) and Oct4/Sox2/Klf4 (second wave). Cells that become refractory to reprogramming activate the first but fail to initiate the second transcriptional wave and can be rescued by elevated expression of all four factors. The establishment of bivalent domains occurs gradually after the first wave, whereas changes in DNA methylation take place after the second wave when cells acquire stable pluripotency. This integrative analysis allowed us to identify genes that act as roadblocks during reprogramming and surface markers that further enrich for cells prone to forming iPSCs. Collectively, our data offer new mechanistic insights into the nature and sequence of molecular events inherent to cellular reprogramming.


Asunto(s)
Reprogramación Celular , Técnicas Citológicas/métodos , Células Madre Pluripotentes Inducidas/citología , Animales , Estudio de Asociación del Genoma Completo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Factor 4 Similar a Kruppel , Ratones , Factores de Transcripción/metabolismo
5.
Mol Cell ; 74(6): 1148-1163.e7, 2019 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-31005419

RESUMEN

Self-renewal and pluripotency of the embryonic stem cell (ESC) state are established and maintained by multiple regulatory networks that comprise transcription factors and epigenetic regulators. While much has been learned regarding transcription factors, the function of epigenetic regulators in these networks is less well defined. We conducted a CRISPR-Cas9-mediated loss-of-function genetic screen that identified two epigenetic regulators, TAF5L and TAF6L, components or co-activators of the GNAT-HAT complexes for the mouse ESC (mESC) state. Detailed molecular studies demonstrate that TAF5L/TAF6L transcriptionally activate c-Myc and Oct4 and their corresponding MYC and CORE regulatory networks. Besides, TAF5L/TAF6L predominantly regulate their target genes through H3K9ac deposition and c-MYC recruitment that eventually activate the MYC regulatory network for self-renewal of mESCs. Thus, our findings uncover a role of TAF5L/TAF6L in directing the MYC regulatory network that orchestrates gene expression programs to control self-renewal for the maintenance of mESC state.


Asunto(s)
Células Madre Embrionarias/metabolismo , Redes Reguladoras de Genes , Células Madre Pluripotentes Inducidas/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Factores Asociados con la Proteína de Unión a TATA/genética , Animales , Sistemas CRISPR-Cas , Ciclo Celular/genética , Proliferación Celular , Reprogramación Celular , Embrión de Mamíferos , Células Madre Embrionarias/citología , Epigénesis Genética , Fibroblastos/citología , Fibroblastos/metabolismo , Edición Génica , Regulación de la Expresión Génica , Células HEK293 , Histonas/genética , Histonas/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Ratones , Cultivo Primario de Células , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Transducción de Señal , Factores Asociados con la Proteína de Unión a TATA/metabolismo
6.
Nature ; 586(7827): 101-107, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32939092

RESUMEN

The reprogramming of human somatic cells to primed or naive induced pluripotent stem cells recapitulates the stages of early embryonic development1-6. The molecular mechanism that underpins these reprogramming processes remains largely unexplored, which impedes our understanding and limits rational improvements to reprogramming protocols. Here, to address these issues, we reconstruct molecular reprogramming trajectories of human dermal fibroblasts using single-cell transcriptomics. This revealed that reprogramming into primed and naive pluripotency follows diverging and distinct trajectories. Moreover, genome-wide analyses of accessible chromatin showed key changes in the regulatory elements of core pluripotency genes, and orchestrated global changes in chromatin accessibility over time. Integrated analysis of these datasets revealed a role for transcription factors associated with the trophectoderm lineage, and the existence of a subpopulation of cells that enter a trophectoderm-like state during reprogramming. Furthermore, this trophectoderm-like state could be captured, which enabled the derivation of induced trophoblast stem cells. Induced trophoblast stem cells are molecularly and functionally similar to trophoblast stem cells derived from human blastocysts or first-trimester placentas7. Our results provide a high-resolution roadmap for the transcription-factor-mediated reprogramming of human somatic cells, indicate a role for the trophectoderm-lineage-specific regulatory program during this process, and facilitate the direct reprogramming of somatic cells into induced trophoblast stem cells.


Asunto(s)
Reprogramación Celular/genética , Regulación de la Expresión Génica , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Trofoblastos/citología , Trofoblastos/metabolismo , Adulto , Cromatina/genética , Cromatina/metabolismo , Ectodermo/citología , Ectodermo/metabolismo , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Transcripción Genética
8.
PLoS Biol ; 19(9): e3001394, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34550965

RESUMEN

The ZEB2 transcription factor has been demonstrated to play important roles in hematopoiesis and leukemic transformation. ZEB1 is a close family member of ZEB2 but has remained more enigmatic concerning its roles in hematopoiesis. Here, we show using conditional loss-of-function approaches and bone marrow (BM) reconstitution experiments that ZEB1 plays a cell-autonomous role in hematopoietic lineage differentiation, particularly as a positive regulator of monocyte development in addition to its previously reported important role in T-cell differentiation. Analysis of existing single-cell (sc) RNA sequencing (RNA-seq) data of early hematopoiesis has revealed distinctive expression differences between Zeb1 and Zeb2 in hematopoietic stem and progenitor cell (HSPC) differentiation, with Zeb2 being more highly and broadly expressed than Zeb1 except at a key transition point (short-term HSC [ST-HSC]➔MPP1), whereby Zeb1 appears to be the dominantly expressed family member. Inducible genetic inactivation of both Zeb1 and Zeb2 using a tamoxifen-inducible Cre-mediated approach leads to acute BM failure at this transition point with increased long-term and short-term hematopoietic stem cell numbers and an accompanying decrease in all hematopoietic lineage differentiation. Bioinformatics analysis of RNA-seq data has revealed that ZEB2 acts predominantly as a transcriptional repressor involved in restraining mature hematopoietic lineage gene expression programs from being expressed too early in HSPCs. ZEB1 appears to fine-tune this repressive role during hematopoiesis to ensure hematopoietic lineage fidelity. Analysis of Rosa26 locus-based transgenic models has revealed that Zeb1 as well as Zeb2 cDNA-based overexpression within the hematopoietic system can drive extramedullary hematopoiesis/splenomegaly and enhance monocyte development. Finally, inactivation of Zeb2 alone or Zeb1/2 together was found to enhance survival in secondary MLL-AF9 acute myeloid leukemia (AML) models attesting to the oncogenic role of ZEB1/2 in AML.


Asunto(s)
Linaje de la Célula , Células Madre Hematopoyéticas/metabolismo , Leucemia Mieloide Aguda/metabolismo , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo , Animales , Células de la Médula Ósea/patología , Diferenciación Celular , Regulación Neoplásica de la Expresión Génica , Hematopoyesis , Células Madre Hematopoyéticas/patología , Leucemia Mieloide Aguda/patología , Ratones , Ratones Transgénicos , RNA-Seq , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética
9.
Nucleic Acids Res ; 50(15): e87, 2022 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-35716123

RESUMEN

Genome wide association studies provide statistical measures of gene-trait associations that reveal how genetic variation influences phenotypes. This study develops an unsupervised dimensionality reduction method called UnTANGLeD (Unsupervised Trait Analysis of Networks from Gene Level Data) which organizes 16,849 genes into discrete gene programs by measuring the statistical association between genetic variants and 1,393 diverse complex traits. UnTANGLeD reveals 173 gene clusters enriched for protein-protein interactions and highly distinct biological processes governing development, signalling, disease, and homeostasis. We identify diverse gene networks with robust interactions but not associated with known biological processes. Analysis of independent disease traits shows that UnTANGLeD gene clusters are conserved across all complex traits, providing a simple and powerful framework to predict novel gene candidates and programs influencing orthogonal disease phenotypes. Collectively, this study demonstrates that gene programs co-ordinately orchestrating cell functions can be identified without reliance on prior knowledge, providing a method for use in functional annotation, hypothesis generation, machine learning and prediction algorithms, and the interpretation of diverse genomic data.


Asunto(s)
Redes Reguladoras de Genes , Estudio de Asociación del Genoma Completo , Enfermedad/genética , Estudio de Asociación del Genoma Completo/métodos , Genómica/métodos , Fenotipo , Polimorfismo de Nucleótido Simple
10.
Nat Methods ; 14(11): 1055-1062, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28945704

RESUMEN

Recent reports on the characteristics of naive human pluripotent stem cells (hPSCs) obtained using independent methods differ. Naive hPSCs have been mainly derived by conversion from primed hPSCs or by direct derivation from human embryos rather than by somatic cell reprogramming. To provide an unbiased molecular and functional reference, we derived genetically matched naive hPSCs by direct reprogramming of fibroblasts and by primed-to-naive conversion using different naive conditions (NHSM, RSeT, 5iLAF and t2iLGöY). Our results show that hPSCs obtained in these different conditions display a spectrum of naive characteristics. Furthermore, our characterization identifies KLF4 as sufficient for conversion of primed hPSCs into naive t2iLGöY hPSCs, underscoring the role that reprogramming factors can play for the derivation of bona fide naive hPSCs.


Asunto(s)
Reprogramación Celular , Células Madre Pluripotentes/citología , Diferenciación Celular , Fibroblastos/citología , Inestabilidad Genómica , Humanos , Factor 4 Similar a Kruppel
11.
EMBO J ; 34(10): 1319-35, 2015 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-25759216

RESUMEN

Snail family members regulate epithelial-to-mesenchymal transition (EMT) during invasion of intestinal tumours, but their role in normal intestinal homeostasis is unknown. Studies in breast and skin epithelia indicate that Snail proteins promote an undifferentiated state. Here, we demonstrate that conditional knockout of Snai1 in the intestinal epithelium results in apoptotic loss of crypt base columnar stem cells and bias towards differentiation of secretory lineages. In vitro organoid cultures derived from Snai1 conditional knockout mice also undergo apoptosis when Snai1 is deleted. Conversely, ectopic expression of Snai1 in the intestinal epithelium in vivo results in the expansion of the crypt base columnar cell pool and a decrease in secretory enteroendocrine and Paneth cells. Following conditional deletion of Snai1, the intestinal epithelium fails to produce a proliferative response following radiation-induced damage indicating a fundamental requirement for Snai1 in epithelial regeneration. These results demonstrate that Snai1 is required for regulation of lineage choice, maintenance of CBC stem cells and regeneration of the intestinal epithelium following damage.


Asunto(s)
Mucosa Intestinal/metabolismo , Intestinos/citología , Factores de Transcripción/metabolismo , Animales , Apoptosis/genética , Apoptosis/fisiología , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Linaje de la Célula , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Noqueados , Transducción de Señal/genética , Transducción de Señal/fisiología , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética
12.
Stem Cells ; 36(6): 822-833, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29396901

RESUMEN

The canonical Wnt/ß-catenin pathway is crucial for early embryonic patterning, tissue homeostasis, and regeneration. While canonical Wnt/ß-catenin stimulation has been used extensively to modulate pluripotency and differentiation of pluripotent stem cells (PSCs), the mechanism of these two seemingly opposing roles has not been fully characterized and is currently largely attributed to activation of nuclear Wnt target genes. Here, we show that low levels of Wnt stimulation via ectopic expression of Wnt1 or administration of glycogen synthase kinase-3 inhibitor CHIR99021 significantly increases PSC differentiation into neurons, cardiomyocytes and early endodermal intermediates. Our data indicate that enhanced differentiation outcomes are not mediated through activation of traditional Wnt target genes but by ß-catenin's secondary role as a binding partner of membrane bound cadherins ultimately leading to the activation of developmental genes. In summary, fine-tuning of Wnt signaling to subthreshold levels for detectable nuclear ß-catenin function appears to act as a switch to enhance differentiation of PSCs into multiple lineages. Our observations highlight a mechanism by which Wnt/ß-catenin signaling can achieve dosage dependent dual roles in regulating self-renewal and differentiation. Stem Cells 2018;36:822-833.


Asunto(s)
Células Madre Pluripotentes/metabolismo , Vía de Señalización Wnt/genética , beta Catenina/genética , Animales , Diferenciación Celular , Humanos , Ratones , Transducción de Señal
13.
Stem Cells ; 35(3): 626-640, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28009074

RESUMEN

The study and application of human pluripotent stem cells (hPSCs) will be enhanced by the availability of well-characterized monoclonal antibodies (mAbs) detecting cell-surface epitopes. Here, we report generation of seven new mAbs that detect cell surface proteins present on live and fixed human ES cells (hESCs) and human iPS cells (hiPSCs), confirming our previous prediction that these proteins were present on the cell surface of hPSCs. The mAbs all show a high correlation with POU5F1 (OCT4) expression and other hPSC surface markers (TRA-160 and SSEA-4) in hPSC cultures and detect rare OCT4 positive cells in differentiated cell cultures. These mAbs are immunoreactive to cell surface protein epitopes on both primed and naive state hPSCs, providing useful research tools to investigate the cellular mechanisms underlying human pluripotency and states of cellular reprogramming. In addition, we report that subsets of the seven new mAbs are also immunoreactive to human bone marrow-derived mesenchymal stem cells (MSCs), normal human breast subsets and both normal and tumorigenic colorectal cell populations. The mAbs reported here should accelerate the investigation of the nature of pluripotency, and enable development of robust cell separation and tracing technologies to enrich or deplete for hPSCs and other human stem and somatic cell types. Stem Cells 2017;35:626-640.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Proteínas de la Membrana/inmunología , Células Madre Pluripotentes/metabolismo , Animales , Antígenos de Superficie/metabolismo , Técnicas de Cultivo de Célula , Diferenciación Celular , Autorrenovación de las Células , Regulación hacia Abajo/genética , Cuerpos Embrioides/citología , Cuerpos Embrioides/metabolismo , Citometría de Flujo , Células Madre Hematopoyéticas/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , Ratones , Factor 3 de Transcripción de Unión a Octámeros/metabolismo
14.
Differentiation ; 87(5): 193-9, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25015842

RESUMEN

Induced pluripotent stem cells (iPSCs) are characterised by their ability to differentiate into any cell type of the body. Accordingly, iPSCs possess immense potential for disease modelling, pharmaceutical screening and autologous cell therapies. The most common source of iPSCs derivation is skin fibroblasts. However, from a clinical point of view, skin fibroblasts may not be ideal, as invasive procedures such as skin biopsies are required for their extraction. Moreover, fibroblasts are highly heterogeneous with a poorly defined developmental pathway, which makes studying reprogramming mechanistics difficult. Granulocytes, on the other hand, are easily obtainable, their developmental pathway has been extensively studied and fluorescence activated cell sorting allows for the isolation of these cells at high purity; thus iPSCs derivation from granulocytes could provide an alternative to fibroblast-derived iPSCs. Previous studies succeeded in producing iPSC colonies from mouse granulocytes but with the use of a mitotically inactivated feeder layer, restricting their use for studying reprogramming mechanistics. As granulocytes display poor survival under culture conditions, we investigated the influence of haematopoietic cytokines to stabilise this cell type in vitro and allow for reprogramming in the absence of a feeder layer. Our results show that treatment with MEF-conditioned media and/or initial exposure to GM-CSF allows for reprogramming of granulocytes under feeder-free conditions. This work can serve as a basis for future work aimed at dissecting the reprogramming mechanism as well as obtaining large numbers of iPSCs from a clinically relevant cell source.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Granulocitos/citología , Células Madre Pluripotentes Inducidas/citología , Animales , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Reprogramación Celular/efectos de los fármacos , Medios de Cultivo Condicionados , Fibroblastos/efectos de los fármacos , Granulocitos/efectos de los fármacos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Ratones
15.
Stem Cells ; 31(4): 703-16, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23307500

RESUMEN

Mitochondrial DNA haplotypes are associated with various phenotypes, such as altered susceptibility to disease, environmental adaptations, and aging. Accumulating evidence suggests that mitochondrial DNA is essential for cell differentiation and the cell phenotype. However, the effects of different mitochondrial DNA haplotypes on differentiation and development remain to be determined. Using embryonic stem cell lines possessing the same Mus musculus chromosomes but harboring one of Mus musculus, Mus spretus, or Mus terricolor mitochondrial DNA haplotypes, we have determined the effects of different mitochondrial DNA haplotypes on chromosomal gene expression, differentiation, and mitochondrial metabolism. In undifferentiated and differentiating embryonic stem cells, we observed mitochondrial DNA haplotype-specific expression of genes involved in pluripotency, differentiation, mitochondrial energy metabolism, and DNA methylation. These mitochondrial DNA haplotypes also influenced the potential of embryonic stem cells to produce spontaneously beating cardiomyocytes. The differences in gene expression patterns and cardiomyocyte production were independent of ATP content, oxygen consumption, and respiratory capacity, which until now have been considered to be the primary roles of mitochondrial DNA. Differentiation of embryonic stem cells harboring the different mitochondrial DNA haplotypes in a 3D environment significantly increased chromosomal gene expression for all haplotypes during differentiation. However, haplotype-specific differences in gene expression patterns were maintained in this environment. Taken together, these results provide significant insight into the phenotypic consequences of mitochondrial DNA haplotypes and demonstrate their influence on differentiation and development. We propose that mitochondrial DNA haplotypes play a pivotal role in the process of differentiation and mediate the fate of the cell.


Asunto(s)
ADN Mitocondrial/genética , Células Madre Embrionarias/metabolismo , Células Madre Pluripotentes/metabolismo , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Línea Celular , Células Madre Embrionarias/citología , Expresión Génica/genética , Expresión Génica/fisiología , Haplotipos/genética , Células Madre Pluripotentes/citología
16.
Dev Cell ; 59(6): 705-722.e8, 2024 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-38354738

RESUMEN

Wnt signaling is a critical determinant of cell lineage development. This study used Wnt dose-dependent induction programs to gain insights into molecular regulation of stem cell differentiation. We performed single-cell RNA sequencing of hiPSCs responding to a dose escalation protocol with Wnt agonist CHIR-99021 during the exit from pluripotency to identify cell types and genetic activity driven by Wnt stimulation. Results of activated gene sets and cell types were used to build a multiple regression model that predicts the efficiency of cardiomyocyte differentiation. Cross-referencing Wnt-associated gene expression profiles to the Connectivity Map database, we identified the small-molecule drug, tranilast. We found that tranilast synergistically activates Wnt signaling to promote cardiac lineage differentiation, which we validate by in vitro analysis of hiPSC differentiation and in vivo analysis of developing quail embryos. Our study provides an integrated workflow that links experimental datasets, prediction models, and small-molecule databases to identify drug-like compounds that control cell differentiation.


Asunto(s)
Miocitos Cardíacos , Vía de Señalización Wnt , ortoaminobenzoatos , Miocitos Cardíacos/metabolismo , Diferenciación Celular/genética , Linaje de la Célula/genética , Vía de Señalización Wnt/genética , Mesodermo
17.
Stem Cells ; 30(7): 1349-61, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22495882

RESUMEN

LIM homeobox transcription factor 1 alpha (Lmx1a) is required for the development of midbrain dopaminergic neurons, roof plate formation, and cortical hem development. We generated a reporter embryonic stem cell (ESC) line for Lmx1a and used it to track differentiation and extract neural progenitors from differentiating mouse ESCs. Lmx1a(+) cells gave rise to functional cortical upper layer GABAergic neurons or dopaminergic neurons depending on the culture conditions used for differentiation. Under chemically defined neurobasal conditions, ESC differentiation resulted in widespread and transient expression of Lmx1a, without the addition of exogenous factors such as sonic hedgehog (Shh), Wnts, and/or bone morphogenic proteins (BMPs). Under neutral conditions, Lmx1a(+) cells express genes known to be downstream of Lmx1a and cortical hem markers Wnt3a and p73. The majority of these cells did not express the ventral midbrain dopaminergic marker Foxa2 or dorsal roof plate marker BMP-2. Lmx1a(+) -Foxa2(-) cells were primed to become SatB2(+) GABAergic neurons and appeared to be resistant to dopaminergic patterning cues. PA6 coculture produced a substantial population of Lmx1a(+) progenitors that also expressed Foxa2 and on further differentiation gave rise to dopaminergic neurons at high frequency. We conclude that Lmx1a is a useful marker for the extraction of progenitors of GABAergic or dopaminergic neurons. We caution against the assumption that it indicates dopaminergic commitment during in vitro differentiation of ESCs. Indeed, in monolayer culture under neurobasal conditions, with or without the addition of Shh and fibroblast growth factor 8 (FGF8), Lmx1a(+) cells were predominantly progenitors of forebrain GABAergic neurons. We obtained dopaminergic cells in large numbers only by coculture with PA6 cells.


Asunto(s)
Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Neuronas GABAérgicas/citología , Neuronas GABAérgicas/metabolismo , Proteínas con Homeodominio LIM/metabolismo , Factor de Transcripción MSX1/metabolismo , Factores de Transcripción/metabolismo , Animales , Diferenciación Celular/fisiología , Línea Celular , Citometría de Flujo , Inmunohistoquímica , Proteínas con Homeodominio LIM/genética , Factor de Transcripción MSX1/genética , Ratones , Reacción en Cadena de la Polimerasa , Factores de Transcripción/genética
18.
Sci Signal ; 16(782): eabq1366, 2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-37098119

RESUMEN

Macrophages are key cellular contributors to the pathogenesis of COVID-19, the disease caused by the virus SARS-CoV-2. The SARS-CoV-2 entry receptor ACE2 is present only on a subset of macrophages at sites of SARS-CoV-2 infection in humans. Here, we investigated whether SARS-CoV-2 can enter macrophages, replicate, and release new viral progeny; whether macrophages need to sense a replicating virus to drive cytokine release; and, if so, whether ACE2 is involved in these mechanisms. We found that SARS-CoV-2 could enter, but did not replicate within, ACE2-deficient human primary macrophages and did not induce proinflammatory cytokine expression. By contrast, ACE2 overexpression in human THP-1-derived macrophages permitted SARS-CoV-2 entry, processing and replication, and virion release. ACE2-overexpressing THP-1 macrophages sensed active viral replication and triggered proinflammatory, antiviral programs mediated by the kinase TBK-1 that limited prolonged viral replication and release. These findings help elucidate the role of ACE2 and its absence in macrophage responses to SARS-CoV-2 infection.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , SARS-CoV-2/fisiología , Enzima Convertidora de Angiotensina 2/genética , Citocinas , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/metabolismo , Macrófagos/metabolismo , Virión/metabolismo
19.
Nat Commun ; 14(1): 2099, 2023 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-37055407

RESUMEN

Megakaryocytes (MK) generate platelets. Recently, we and others, have reported MK also regulate hematopoietic stem cells (HSC). Here we show high ploidy large cytoplasmic megakaryocytes (LCM) are critical negative regulators of HSC and critical for platelet formation. Using a mouse knockout model (Pf4-Srsf3Δ/Δ) with normal MK numbers, but essentially devoid of LCM, we demonstrate a pronounced increase in BM HSC concurrent with endogenous mobilization and extramedullary hematopoiesis. Severe thrombocytopenia is observed in animals with diminished LCM, although there is no change in MK ploidy distribution, uncoupling endoreduplication and platelet production. When HSC isolated from a microenvironment essentially devoid of LCM reconstitute hematopoiesis in lethally irradiated mice, the absence of LCM increases HSC in BM, blood and spleen, and the recapitulation of thrombocytopenia. In contrast, following a competitive transplant using minimal numbers of WT HSC together with HSC from a microenvironment with diminished LCM, sufficient WT HSC-generated LCM regulates a normal HSC pool and prevents thrombocytopenia. Importantly, LCM are conserved in humans.


Asunto(s)
Megacariocitos , Trombocitopenia , Humanos , Animales , Megacariocitos/metabolismo , Células Madre Hematopoyéticas/metabolismo , Plaquetas , Trombopoyesis/genética , Hematopoyesis/genética , Trombocitopenia/metabolismo , Modelos Animales de Enfermedad , Ploidias , Factores de Empalme Serina-Arginina/metabolismo
20.
Stem Cells ; 29(6): 928-39, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21472823

RESUMEN

Investigation of serotonergic neuronal activity and its relationship to disease has been limited by a lack of physiologically relevant in vitro cell models. Serotonergic neurons derived from embryonic stem cells (ESCs) could provide a platform for such studies and provide models for use in drug discovery. Here, we report enhancement of serotonergic differentiation using a genetic approach. Expression of Gata2 increased the yield of serotonergic neurons. Enhancement was only achieved when Gata2 was expressed under the control of the tissue-specific promoter of the transcription factor Nkx6.1. High levels of Gata2 expression in ESCs compromised pluripotency and induced non-neuronal differentiation. Combined directed expression of Gata2, proneural gene Mash1, and forkhead transcription factor Foxa2 further enhanced serotonergic neural differentiation, resulting in a 10-fold increase in serotonin content. These neurons were also capable of depolarization (KCl, 30 mM)-induced elevations of intracellular Ca(2+) . The presence of sonic hedgehog during differentiation produced a further modest increase in numbers (1.5-fold). Transgene expression did not influence the number of tyrosine hydroxylase positive neurons in the cultures after 20 days, implying that Gata2, Mash1, and Foxa2 modulate in vitro differentiation at a time beyond the decision-point for dopaminergic or nondopaminergic commitment. This study demonstrates that the directed expression of specific transcription factors enhances serotonergic neuron differentiation in vitro and highlights the importance of transgene expression at the right stage of ESC differentiation to effect the generation of a desired neural subtype.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular , Células Madre Embrionarias/citología , Factor de Transcripción GATA2/metabolismo , Factor Nuclear 3-beta del Hepatocito/metabolismo , Neuronas/metabolismo , Serotonina/biosíntesis , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Clonación Molecular , Células Madre Embrionarias/metabolismo , Factor de Transcripción GATA2/genética , Factor de Transcripción GATA3/genética , Factor de Transcripción GATA3/metabolismo , Genes Reporteros , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Proteínas Hedgehog/farmacología , Factor Nuclear 3-beta del Hepatocito/genética , Proteínas de Homeodominio/genética , Ratones , Factor 1 de Elongación Peptídica/genética , Fenotipo , Regiones Promotoras Genéticas , Biosíntesis de Proteínas/efectos de los fármacos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , beta-Lactamasas/biosíntesis , beta-Lactamasas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA