Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 627(8004): 594-603, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38383780

RESUMEN

Although KDM5C is one of the most frequently mutated genes in X-linked intellectual disability1, the exact mechanisms that lead to cognitive impairment remain unknown. Here we use human patient-derived induced pluripotent stem cells and Kdm5c knockout mice to conduct cellular, transcriptomic, chromatin and behavioural studies. KDM5C is identified as a safeguard to ensure that neurodevelopment occurs at an appropriate timescale, the disruption of which leads to intellectual disability. Specifically, there is a developmental window during which KDM5C directly controls WNT output to regulate the timely transition of primary to intermediate progenitor cells and consequently neurogenesis. Treatment with WNT signalling modulators at specific times reveal that only a transient alteration of the canonical WNT signalling pathway is sufficient to rescue the transcriptomic and chromatin landscapes in patient-derived cells and to induce these changes in wild-type cells. Notably, WNT inhibition during this developmental period also rescues behavioural changes of Kdm5c knockout mice. Conversely, a single injection of WNT3A into the brains of wild-type embryonic mice cause anxiety and memory alterations. Our work identifies KDM5C as a crucial sentinel for neurodevelopment and sheds new light on KDM5C mutation-associated intellectual disability. The results also increase our general understanding of memory and anxiety formation, with the identification of WNT functioning in a transient nature to affect long-lasting cognitive function.


Asunto(s)
Cognición , Embrión de Mamíferos , Desarrollo Embrionario , Histona Demetilasas , Vía de Señalización Wnt , Animales , Humanos , Ratones , Ansiedad , Cromatina/efectos de los fármacos , Cromatina/genética , Cromatina/metabolismo , Embrión de Mamíferos/metabolismo , Perfilación de la Expresión Génica , Histona Demetilasas/genética , Histona Demetilasas/metabolismo , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Discapacidad Intelectual/genética , Memoria , Ratones Noqueados , Mutación , Neurogénesis/genética , Vía de Señalización Wnt/efectos de los fármacos
3.
Genet Med ; 23(6): 1158-1162, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33531666

RESUMEN

PURPOSE: The endoplasmic reticulum membrane complex (EMC) is a highly conserved, multifunctional 10-protein complex related to membrane protein biology. In seven families, we identified 13 individuals with highly overlapping phenotypes who harbor a single identical homozygous frameshift variant in EMC10. METHODS: Using exome, genome, and Sanger sequencing, a recurrent frameshift EMC10 variant was identified in affected individuals in an international cohort of consanguineous families. Multiple families were independently identified and connected via Matchmaker Exchange and internal databases. We assessed the effect of the frameshift variant on EMC10 RNA and protein expression and evaluated EMC10 expression in normal human brain tissue using immunohistochemistry. RESULTS: A homozygous variant EMC10 c.287delG (Refseq NM_206538.3, p.Gly96Alafs*9) segregated with affected individuals in each family, who exhibited a phenotypic spectrum of intellectual disability (ID) and global developmental delay (GDD), variable seizures and variable dysmorphic features (elongated face, curly hair, cubitus valgus, and arachnodactyly). The variant arose on two founder haplotypes and results in significantly reduced EMC10 RNA expression and an unstable truncated EMC10 protein. CONCLUSION: We propose that a homozygous loss-of-function variant in EMC10 causes a novel syndromic neurodevelopmental phenotype. Remarkably, the recurrent variant is likely the result of a hypermutable site and arose on distinct founder haplotypes.


Asunto(s)
Discapacidades del Desarrollo , Discapacidad Intelectual , Niño , Discapacidades del Desarrollo/genética , Mutación del Sistema de Lectura , Homocigoto , Humanos , Discapacidad Intelectual/genética , Proteínas de la Membrana/genética , Linaje , Fenotipo , Convulsiones/genética
4.
Nature ; 523(7561): 468-71, 2015 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-26201599

RESUMEN

Haematopoietic stem and progenitor cell (HSPC) transplant is a widely used treatment for life-threatening conditions such as leukaemia; however, the molecular mechanisms regulating HSPC engraftment of the recipient niche remain incompletely understood. Here we develop a competitive HSPC transplant method in adult zebrafish, using in vivo imaging as a non-invasive readout. We use this system to conduct a chemical screen, and identify epoxyeicosatrienoic acids (EETs) as a family of lipids that enhance HSPC engraftment. The pro-haematopoietic effects of EETs were conserved in the developing zebrafish embryo, where 11,12-EET promoted HSPC specification by activating a unique activator protein 1 (AP-1) and runx1 transcription program autonomous to the haemogenic endothelium. This effect required the activation of the phosphatidylinositol-3-OH kinase (PI(3)K) pathway, specifically PI(3)Kγ. In adult HSPCs, 11,12-EET induced transcriptional programs, including AP-1 activation, which modulate several cellular processes, such as migration, to promote engraftment. Furthermore, we demonstrate that the EET effects on enhancing HSPC homing and engraftment are conserved in mammals. Our study establishes a new method to explore the molecular mechanisms of HSPC engraftment, and discovers a previously unrecognized, evolutionarily conserved pathway regulating multiple haematopoietic generation and regeneration processes. EETs may have clinical application in marrow or cord blood transplantation.


Asunto(s)
Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Hematopoyesis , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Pez Cebra/embriología , Ácido 8,11,14-Eicosatrienoico/metabolismo , Animales , Línea Celular , Movimiento Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Femenino , Regulación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana , Humanos , Riñón/citología , Masculino , Ratones , Fosfatidilinositol 3-Quinasas , Factor de Transcripción AP-1/metabolismo , Transcripción Genética
5.
Nature ; 491(7426): 769-73, 2012 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-23103873

RESUMEN

In the course of primary infection with herpes simplex virus 1 (HSV-1), children with inborn errors of toll-like receptor 3 (TLR3) immunity are prone to HSV-1 encephalitis (HSE). We tested the hypothesis that the pathogenesis of HSE involves non-haematopoietic CNS-resident cells. We derived induced pluripotent stem cells (iPSCs) from the dermal fibroblasts of TLR3- and UNC-93B-deficient patients and from controls. These iPSCs were differentiated into highly purified populations of neural stem cells (NSCs), neurons, astrocytes and oligodendrocytes. The induction of interferon-ß (IFN-ß) and/or IFN-λ1 in response to stimulation by the dsRNA analogue polyinosinic:polycytidylic acid (poly(I:C)) was dependent on TLR3 and UNC-93B in all cells tested. However, the induction of IFN-ß and IFN-λ1 in response to HSV-1 infection was impaired selectively in UNC-93B-deficient neurons and oligodendrocytes. These cells were also much more susceptible to HSV-1 infection than control cells, whereas UNC-93B-deficient NSCs and astrocytes were not. TLR3-deficient neurons were also found to be susceptible to HSV-1 infection. The rescue of UNC-93B- and TLR3-deficient cells with the corresponding wild-type allele showed that the genetic defect was the cause of the poly(I:C) and HSV-1 phenotypes. The viral infection phenotype was rescued further by treatment with exogenous IFN-α or IFN-ß ( IFN-α/ß) but not IFN-λ1. Thus, impaired TLR3- and UNC-93B-dependent IFN-α/ß intrinsic immunity to HSV-1 in the CNS, in neurons and oligodendrocytes in particular, may underlie the pathogenesis of HSE in children with TLR3-pathway deficiencies.


Asunto(s)
Sistema Nervioso Central/patología , Herpesvirus Humano 1/inmunología , Células Madre Pluripotentes Inducidas/citología , Receptor Toll-Like 3/deficiencia , Astrocitos/inmunología , Astrocitos/virología , Biomarcadores , Diferenciación Celular , Linaje de la Célula , Separación Celular , Células Cultivadas , Sistema Nervioso Central/citología , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/virología , Niño , Susceptibilidad a Enfermedades , Encefalitis por Herpes Simple/inmunología , Encefalitis por Herpes Simple/metabolismo , Encefalitis por Herpes Simple/patología , Encefalitis por Herpes Simple/virología , Herpesvirus Humano 1/patogenicidad , Humanos , Inmunidad Innata , Células Madre Pluripotentes Inducidas/virología , Interferones/inmunología , Proteínas de Transporte de Membrana/deficiencia , Proteínas de Transporte de Membrana/genética , Células-Madre Neurales/inmunología , Células-Madre Neurales/virología , Neuronas/inmunología , Neuronas/patología , Neuronas/virología , Oligodendroglía/inmunología , Oligodendroglía/patología , Oligodendroglía/virología , Receptor Toll-Like 3/genética
6.
Mol Ther ; 25(9): 1999-2013, 2017 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-28619647

RESUMEN

Patient-derived induced pluripotent stem cells (iPSCs) hold great promise for autologous cell replacement. However, for many inherited diseases, treatment will likely require genetic repair pre-transplantation. Genome editing technologies are useful for this application. The purpose of this study was to develop CRISPR-Cas9-mediated genome editing strategies to target and correct the three most common types of disease-causing variants in patient-derived iPSCs: (1) exonic, (2) deep intronic, and (3) dominant gain of function. We developed a homology-directed repair strategy targeting a homozygous Alu insertion in exon 9 of male germ cell-associated kinase (MAK) and demonstrated restoration of the retinal transcript and protein in patient cells. We generated a CRISPR-Cas9-mediated non-homologous end joining (NHEJ) approach to excise a major contributor to Leber congenital amaurosis, the IVS26 cryptic-splice mutation in CEP290, and demonstrated correction of the transcript and protein in patient iPSCs. Lastly, we designed allele-specific CRISPR guides that selectively target the mutant Pro23His rhodopsin (RHO) allele, which, following delivery to both patient iPSCs in vitro and pig retina in vivo, created a frameshift and premature stop that would prevent transcription of the disease-causing variant. The strategies developed in this study will prove useful for correcting a wide range of genetic variants in genes that cause inherited retinal degeneration.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Marcación de Gen , Células Madre Pluripotentes Inducidas/metabolismo , Degeneración Retiniana/genética , Trasplante de Células Madre , Alelos , Animales , Línea Celular , Orden Génico , Sitios Genéticos , Terapia Genética , Vectores Genéticos/genética , Recombinación Homóloga , Humanos , Células Madre Pluripotentes Inducidas/citología , Intrones , Mutación , Proteínas Serina-Treonina Quinasas/genética , ARN Guía de Kinetoplastida , Degeneración Retiniana/terapia , Trasplante de Células Madre/métodos , Trasplante Autólogo
7.
Blood ; 125(9): 1418-26, 2015 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-25587036

RESUMEN

Hematopoietic and vascular development share many common features, including cell surface markers and sites of origin. Recent lineage-tracing studies have established that definitive hematopoietic stem and progenitor cells arise from vascular endothelial-cadherin(+) hemogenic endothelial cells of the aorta-gonad-mesonephros region, but the genetic programs underlying the specification of hemogenic endothelial cells remain poorly defined. Here, we discovered that Notch induction enhances hematopoietic potential and promotes the specification of hemogenic endothelium in differentiating cultures of mouse embryonic stem cells, and we identified Foxc2 as a highly upregulated transcript in the hemogenic endothelial population. Studies in zebrafish and mouse embryos revealed that Foxc2 and its orthologs are required for the proper development of definitive hematopoiesis and function downstream of Notch signaling in the hemogenic endothelium. These data establish a pathway linking Notch signaling to Foxc2 in hemogenic endothelial cells to promote definitive hematopoiesis.


Asunto(s)
Células Madre Embrionarias/citología , Endotelio Vascular/citología , Factores de Transcripción Forkhead/metabolismo , Regulación del Desarrollo de la Expresión Génica , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Receptor Notch1/metabolismo , Animales , Apoptosis , Western Blotting , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Células Cultivadas , Células Madre Embrionarias/metabolismo , Endotelio Vascular/metabolismo , Factores de Transcripción Forkhead/genética , Células Madre Hematopoyéticas/metabolismo , Ratones , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor Notch1/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/metabolismo
8.
Proc Natl Acad Sci U S A ; 111(28): 10299-304, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24982195

RESUMEN

Lung cancer is notorious for its ability to metastasize, but the pathways regulating lung cancer metastasis are largely unknown. An in vitro system designed to discover factors critical for lung cancer cell migration identified brain-derived neurotrophic factor, which stimulates cell migration through activation of tropomyosin-related kinase B (TrkB; also called NTRK2). Knockdown of TrkB in human lung cancer cell lines significantly decreased their migratory and metastatic ability in vitro and in vivo. In an autochthonous lung adenocarcinoma model driven by activated oncogenic Kras and p53 loss, TrkB deficiency significantly reduced metastasis. Hypoxia-inducible factor-1 directly regulated TrkB expression, and, in turn, TrkB activated Akt signaling in metastatic lung cancer cells. Finally, TrkB expression was correlated with metastasis in patient samples, and TrkB was detected more often in tumors that did not have Kras or epidermal growth factor receptor mutations. These studies demonstrate that TrkB is an important therapeutic target in metastatic lung adenocarcinoma.


Asunto(s)
Adenocarcinoma/enzimología , Movimiento Celular , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/enzimología , Glicoproteínas de Membrana/biosíntesis , Proteínas Tirosina Quinasas/biosíntesis , Receptor trkB/biosíntesis , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/patología , Animales , Línea Celular Tumoral , Técnicas de Silenciamiento del Gen , Humanos , Factor 1 Inducible por Hipoxia/genética , Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Glicoproteínas de Membrana/genética , Ratones Mutantes , Metástasis de la Neoplasia/genética , Metástasis de la Neoplasia/patología , Proteínas Tirosina Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor trkB/genética , Transducción de Señal/genética
9.
J Biol Chem ; 289(11): 7835-43, 2014 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-24509859

RESUMEN

Mitochondrial iron is essential for the biosynthesis of heme and iron-sulfur ([Fe-S]) clusters in mammalian cells. In developing erythrocytes, iron is imported into the mitochondria by MFRN1 (mitoferrin-1, SLC25A37). Although loss of MFRN1 in zebrafish and mice leads to profound anemia, mutant animals showed no overt signs of porphyria, suggesting that mitochondrial iron deficiency does not result in an accumulation of protoporphyrins. Here, we developed a gene trap model to provide in vitro and in vivo evidence that iron regulatory protein-1 (IRP1) inhibits protoporphyrin accumulation. Mfrn1(+/gt);Irp1(-/-) erythroid cells exhibit a significant increase in protoporphyrin levels. IRP1 attenuates protoporphyrin biosynthesis by binding to the 5'-iron response element (IRE) of alas2 mRNA, inhibiting its translation. Ectopic expression of alas2 harboring a mutant IRE, preventing IRP1 binding, in Mfrn1(gt/gt) cells mimics Irp1 deficiency. Together, our data support a model whereby impaired mitochondrial [Fe-S] cluster biogenesis in Mfrn1(gt/gt) cells results in elevated IRP1 RNA-binding that attenuates ALAS2 mRNA translation and protoporphyrin accumulation.


Asunto(s)
5-Aminolevulinato Sintetasa/metabolismo , Regulación de la Expresión Génica , Proteína 1 Reguladora de Hierro/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Porfirias/metabolismo , Animales , Blastocisto/citología , Diferenciación Celular , Línea Celular Tumoral , Femenino , Genotipo , Células HEK293 , Hemo/química , Humanos , Hierro/química , Proteínas Hierro-Azufre/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Biosíntesis de Proteínas , Protoporfirinas/metabolismo , Pez Cebra
10.
NPJ Syst Biol Appl ; 10(1): 3, 2024 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-38184707

RESUMEN

Experimental studies have shown that chromatin modifiers have a critical effect on cellular reprogramming, i.e., the conversion of differentiated cells to pluripotent stem cells. Here, we develop a model of the OCT4 gene regulatory network that includes genes expressing chromatin modifiers TET1 and JMJD2, and the chromatin modification circuit on which these modifiers act. We employ this model to compare three reprogramming approaches that have been considered in the literature with respect to reprogramming efficiency and latency variability. These approaches are overexpression of OCT4 alone, overexpression of OCT4 with TET1, and overexpression of OCT4 with JMJD2. Our results show more efficient and less variable reprogramming when also JMJD2 and TET1 are overexpressed, consistent with previous experimental data. Nevertheless, TET1 overexpression can lead to more efficient reprogramming compared to JMJD2 overexpression. This is the case when the recruitment of DNA methylation by H3K9me3 is weak and the methyl-CpG-binding domain (MBD) proteins are sufficiently scarce such that they do not hamper TET1 binding to methylated DNA. The model that we developed provides a mechanistic understanding of existing experimental results and is also a tool for designing optimized reprogramming approaches that combine overexpression of cell-fate specific transcription factors (TFs) with targeted recruitment of epigenetic modifiers.


Asunto(s)
Reprogramación Celular , Redes Reguladoras de Genes , Reprogramación Celular/genética , Diferenciación Celular/genética , Redes Reguladoras de Genes/genética , Cromatina , Epigénesis Genética/genética
11.
Epigenetics ; 19(1): 2346694, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38739481

RESUMEN

The transgenerational effects of exposing male mice to chronic social instability (CSI) stress are associated with decreased sperm levels of multiple members of the miR-34/449 family that persist after their mating through preimplantation embryo (PIE) development. Here we demonstrate the importance of these miRNA changes by showing that restoring miR-34c levels in PIEs derived from CSI stressed males prevents elevated anxiety and defective sociability normally found specifically in their adult female offspring. It also restores, at least partially, levels of sperm miR-34/449 normally reduced in their male offspring who transmit these sex-specific traits to their offspring. Strikingly, these experiments also revealed that inducing miR-34c levels in PIEs enhances the expression of its own gene and that of miR-449 in these cells. The same induction of embryo miR-34/449 gene expression likely occurs after sperm-derived miR-34c is introduced into oocytes upon fertilization. Thus, suppression of this miRNA amplification system when sperm miR-34c levels are reduced in CSI stressed mice can explain how a comparable fold-suppression of miR-34/449 levels can be found in PIEs derived from them, despite sperm containing ~50-fold lower levels of these miRNAs than those already present in PIEs. We previously found that men exposed to early life trauma also display reduced sperm levels of miR-34/449. And here we show that miR-34c can also increase the expression of its own gene, and that of miR-449 in human embryonic stem cells, suggesting that human PIEs derived from men with low sperm miR-34/449 levels may also contain this potentially harmful defect.


Asunto(s)
Blastocisto , Epigénesis Genética , MicroARNs , Espermatozoides , Estrés Psicológico , MicroARNs/genética , MicroARNs/metabolismo , Masculino , Animales , Espermatozoides/metabolismo , Femenino , Ratones , Blastocisto/metabolismo , Estrés Psicológico/metabolismo , Estrés Psicológico/genética , Humanos , Ratones Endogámicos C57BL
12.
Nat Commun ; 15(1): 1274, 2024 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-38341433

RESUMEN

Although emerging evidence indicates that alterations in proteins within nuclear compartments elicit changes in chromosomal architecture and differentiation, the underlying mechanisms are not well understood. Here we investigate the direct role of the abundant nuclear complex protein Matrin3 (Matr3) in chromatin architecture and development in the context of myogenesis. Using an acute targeted protein degradation platform (dTAG-Matr3), we reveal the dynamics of development-related chromatin reorganization. High-throughput chromosome conformation capture (Hi-C) experiments revealed substantial chromatin loop rearrangements soon after Matr3 depletion. Notably, YY1 binding was detected, accompanied by the emergence of novel YY1-mediated enhancer-promoter loops, which occurred concurrently with changes in histone modifications and chromatin-level binding patterns. Changes in chromatin occupancy by Matr3 also correlated with these alterations. Overall, our results suggest that Matr3 mediates differentiation through stabilizing chromatin accessibility and chromatin loop-domain interactions, and highlight a conserved and direct role for Matr3 in maintenance of chromosomal architecture.


Asunto(s)
Cromatina , Elementos de Facilitación Genéticos , Proteínas Asociadas a Matriz Nuclear , Proteínas de Unión al ARN , Núcleo Celular , Cromatina/química , Cromatina/genética , Cromatina/metabolismo , Cromosomas , Regiones Promotoras Genéticas/genética , Humanos , Proteínas de Unión al ARN/metabolismo , Proteínas Asociadas a Matriz Nuclear/metabolismo
13.
bioRxiv ; 2023 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-37786715

RESUMEN

Chronically stressing male mice can alter the behavior of their offspring across generations. This effect is thought to be mediated by stress-induced changes in the content of specific sperm miRNAs that modify embryo development after their delivery to oocytes at fertilization. A major problem with this hypothesis is that the levels of mouse sperm miRNAs are much lower than those present in preimplantation embryos. This makes it unclear how embryos could be significantly impacted without an amplification system to magnify changes in sperm miRNA content, like those present in lower organisms where transgenerational epigenetic inheritance is well established. Here, we describe such a system for Chronic Social Instability (CSI) stress that can explain how it reduces the levels of the miR-34b,c/449a,b family of miRNAs not only in sperm of exposed males but also in preimplantation embryos ( PIEs ) derived from their mating, as well as in sperm of male offspring. Sperm-derived miR-34c normally positively regulates expression of its own gene and that of miR-449 in PIEs. This feed forward, auto-amplification process is suppressed when CSI stress reduces sperm miR-34c levels. Its suppression is important for the transmission of traits to offspring because restoring miR-34c levels in PIEs from CSI stressed males, which also restores levels of miR-449 in them, suppresses elements of elevated anxiety and defective sociability normally found specifically in their female offspring, as well as reduced sperm miR-34 and miR-449 levels normally found in male offspring, who pass on these traits to their offspring. We previously published that the content of sperm miR-34/449 is also reduced in men raised in highly abusive and/or dysfunctional families. We show here that a similar miRNA auto-amplification system functions in human embryonic stem cells. This raises the possibility that PIEs in offspring of these men also display reduced levels of miR-34/449, enhancing the potential translational significance of these studies.

14.
bioRxiv ; 2023 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-36747813

RESUMEN

Reprogramming human fibroblasts to induced pluripotent stem cells (iPSCs) is inefficient, with heterogeneity among transcription factor (TF) trajectories driving divergent cell states. Nevertheless, the impact of TF dynamics on reprogramming efficiency remains uncharted. Here, we identify the successful reprogramming trajectories of the core pluripotency TF, OCT4, and design a genetic controller that enforces such trajectories with high precision. By combining a genetic circuit that generates a wide range of OCT4 trajectories with live-cell imaging, we track OCT4 trajectories with clonal resolution and find that a distinct constant OCT4 trajectory is required for colony formation. We then develop a synthetic genetic circuit that yields a tight OCT4 distribution around the identified trajectory and outperforms in terms of reprogramming efficiency other circuits that less accurately regulate OCT4. Our synthetic biology approach is generalizable for identifying and enforcing TF dynamics for cell fate programming applications.

15.
Theranostics ; 13(11): 3707-3724, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37441602

RESUMEN

Background: Extracellular vesicles (EVs) carry bioactive molecules associated with various biological processes, including miRNAs. In both Huntington's disease (HD) models and human samples, altered expression of miRNAs involved in synapse regulation was reported. Recently, the use of EV cargo to reverse phenotypic alterations in disease models with synaptopathy as the end result of the pathophysiological cascade has become an interesting possibility. Methods: Here, we assessed the contribution of EVs to GABAergic synaptic alterations using a human HD model and studied the miRNA content of isolated EVs. Results: After differentiating human induced pluripotent stem cells into electrophysiologically active striatal-like GABAergic neurons, we found that HD-derived neurons displayed reduced density of inhibitory synapse markers and GABA receptor-mediated ionotropic signaling. Treatment with EVs secreted by control (CTR) fibroblasts reversed the deficits in GABAergic synaptic transmission and increased the density of inhibitory synapses in HD-derived neuron cultures, while EVs from HD-derived fibroblasts had the opposite effects on CTR-derived neurons. Moreover, analysis of miRNAs from purified EVs identified a set of differentially expressed miRNAs between manifest HD, premanifest, and CTR lines with predicted synaptic targets. Conclusion: The EV-mediated reversal of the abnormal GABAergic phenotype in HD-derived neurons reinforces the potential role of EV-miRNAs on synapse regulation.


Asunto(s)
Vesículas Extracelulares , Enfermedad de Huntington , Células Madre Pluripotentes Inducidas , MicroARNs , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , MicroARNs/metabolismo , Neuronas GABAérgicas/metabolismo , Vesículas Extracelulares/metabolismo
16.
Sci Adv ; 9(48): eadg8495, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-38019912

RESUMEN

Reprogramming human fibroblasts to induced pluripotent stem cells (iPSCs) is inefficient, with heterogeneity among transcription factor (TF) trajectories driving divergent cell states. Nevertheless, the impact of TF dynamics on reprogramming efficiency remains uncharted. We develop a system that accurately reports OCT4 protein levels in live cells and use it to reveal the trajectories of OCT4 in successful reprogramming. Our system comprises a synthetic genetic circuit that leverages noise to generate a wide range of OCT4 trajectories and a microRNA targeting endogenous OCT4 to set total cellular OCT4 protein levels. By fusing OCT4 to a fluorescent protein, we are able to track OCT4 trajectories with clonal resolution via live-cell imaging. We discover that a supraphysiological, stable OCT4 level is required, but not sufficient, for efficient iPSC colony formation. Our synthetic genetic circuit design and high-throughput live-imaging pipeline are generalizable for investigating TF dynamics for other cell fate programming applications.


Asunto(s)
Reprogramación Celular , Células Madre Pluripotentes Inducidas , Humanos , Diferenciación Celular/genética , Células Cultivadas , Reprogramación Celular/genética , Fibroblastos/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
17.
Blood ; 115(17): 3453-62, 2010 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-20089964

RESUMEN

Fanconi anemia (FA) is a genetically heterogeneous, autosomal recessive disorder characterized by pediatric bone marrow failure and congenital anomalies. The effect of FA gene deficiency on hematopoietic development in utero remains poorly described as mouse models of FA do not develop hematopoietic failure and such studies cannot be performed on patients. We have created a human-specific in vitro system to study early hematopoietic development in FA using a lentiviral RNA interference (RNAi) strategy in human embryonic stem cells (hESCs). We show that knockdown of FANCA and FANCD2 in hESCs leads to a reduction in hematopoietic fates and progenitor numbers that can be rescued by FA gene complementation. Our data indicate that hematopoiesis is impaired in FA from the earliest stages of development, suggesting that deficiencies in embryonic hematopoiesis may underlie the progression to bone marrow failure in FA. This work illustrates how hESCs can provide unique insights into human development and further our understanding of genetic disease.


Asunto(s)
Células Madre Embrionarias/metabolismo , Proteína del Grupo de Complementación A de la Anemia de Fanconi , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi , Anemia de Fanconi/metabolismo , Técnicas de Silenciamiento del Gen , Médula Ósea/metabolismo , Médula Ósea/patología , Línea Celular , Células Madre Embrionarias/patología , Anemia de Fanconi/genética , Anemia de Fanconi/patología , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Humanos , Modelos Biológicos , Interferencia de ARN
18.
J Allergy Clin Immunol ; 127(6): 1400-7.e4, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21185069

RESUMEN

BACKGROUND: The novel ability to epigenetically reprogram somatic cells into induced pluripotent stem cells (iPSCs) through the exogenous expression of transcription promises to revolutionize the study of human diseases. OBJECTIVE: Here we report on the generation of 25 iPSC lines from 6 patients with various forms of primary immunodeficiencies (PIDs) affecting adaptive immunity, innate immunity, or both. METHODS: Patients' dermal fibroblasts were reprogrammed by expression of 4 transcription factors, octamer-binding transcription factor 4 (OCT4), sex determining region Y-box 2 (SOX2), Krueppel-like factor 4 (KLF4), and cellular myelomonocytosis proto-oncogene (cMYC), by using a single excisable polycistronic lentiviral vector. RESULTS: iPSCs derived from patients with PIDs show a stemness profile that is comparable with that observed in human embryonic stem cells. After in vitro differentiation into embryoid bodies, pluripotency of the patient-derived iPSC lines was demonstrated by expression of genes characteristic of each of the 3 embryonic layers. We have confirmed the patient-specific origin of the iPSC lines and ascertained maintenance of karyotypic integrity. CONCLUSION: By providing a limitless source of diseased stem cells that can be differentiated into various cell types in vitro, the repository of iPSC lines from patients with PIDs represents a unique resource to investigate the pathophysiology of hematopoietic and extrahematopoietic manifestations of these diseases and might assist in the development of novel therapeutic approaches based on gene correction.


Asunto(s)
Síndromes de Inmunodeficiencia/patología , Síndromes de Inmunodeficiencia/fisiopatología , Células Madre Pluripotentes Inducidas/patología , Células Madre Pluripotentes Inducidas/fisiología , Inmunidad Adaptativa , Desdiferenciación Celular , Diferenciación Celular , Línea Celular , Transdiferenciación Celular , ADN/genética , Expresión Génica , Genes myc , Humanos , Inmunidad Innata , Síndromes de Inmunodeficiencia/genética , Cariotipificación , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Factor 3 de Transcripción de Unión a Octámeros/genética , Proto-Oncogenes Mas , Factores de Transcripción SOXB1/genética
19.
Cell Stem Cell ; 29(8): 1181-1196.e6, 2022 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-35931029

RESUMEN

Human induced pluripotent stem cells (iPSCs) provide a potentially unlimited resource for cell therapies, but the derivation of mature cell types remains challenging. The histone methyltransferase EZH1 is a negative regulator of lymphoid potential during embryonic hematopoiesis. Here, we demonstrate that EZH1 repression facilitates in vitro differentiation and maturation of T cells from iPSCs. Coupling a stroma-free T cell differentiation system with EZH1-knockdown-mediated epigenetic reprogramming, we generated iPSC-derived T cells, termed EZ-T cells, which display a highly diverse T cell receptor (TCR) repertoire and mature molecular signatures similar to those of TCRαß T cells from peripheral blood. Upon activation, EZ-T cells give rise to effector and memory T cell subsets. When transduced with chimeric antigen receptors (CARs), EZ-T cells exhibit potent antitumor activities in vitro and in xenograft models. Epigenetic remodeling via EZH1 repression allows efficient production of developmentally mature T cells from iPSCs for applications in adoptive cell therapy.


Asunto(s)
Células Madre Pluripotentes Inducidas , Receptores Quiméricos de Antígenos , Diferenciación Celular , Humanos , Inmunoterapia Adoptiva , Células Madre Pluripotentes Inducidas/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Receptores Quiméricos de Antígenos/metabolismo , Linfocitos T
20.
Nat Commun ; 12(1): 3626, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34131117

RESUMEN

Platelet aggregation at the site of atherosclerotic vascular injury is the underlying pathophysiology of myocardial infarction and stroke. To build upon prior GWAS, here we report on 16 loci identified through a whole genome sequencing (WGS) approach in 3,855 NHLBI Trans-Omics for Precision Medicine (TOPMed) participants deeply phenotyped for platelet aggregation. We identify the RGS18 locus, which encodes a myeloerythroid lineage-specific regulator of G-protein signaling that co-localizes with expression quantitative trait loci (eQTL) signatures for RGS18 expression in platelets. Gene-based approaches implicate the SVEP1 gene, a known contributor of coronary artery disease risk. Sentinel variants at RGS18 and PEAR1 are associated with thrombosis risk and increased gastrointestinal bleeding risk, respectively. Our WGS findings add to previously identified GWAS loci, provide insights regarding the mechanism(s) by which genetics may influence cardiovascular disease risk, and underscore the importance of rare variant and regulatory approaches to identifying loci contributing to complex phenotypes.


Asunto(s)
Plaquetas/metabolismo , Mapeo Cromosómico , Secuenciación Completa del Genoma , Secuencia de Bases , Proteínas de Unión al GTP , Estudio de Asociación del Genoma Completo , Células HEK293 , Humanos , Células K562 , Fenotipo , Agregación Plaquetaria , Pruebas de Función Plaquetaria , Polimorfismo de Nucleótido Simple , Sitios de Carácter Cuantitativo , Proteínas RGS/genética , Proteínas RGS/metabolismo , Receptores de Superficie Celular/genética , Trombosis/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA