Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Chem Biol ; 30(7): 780-794.e8, 2023 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-37379846

RESUMEN

Overlapping principles of embryonic and tumor biology have been described, with recent multi-omics campaigns uncovering shared molecular profiles between human pluripotent stem cells (hPSCs) and adult tumors. Here, using a chemical genomic approach, we provide biological evidence that early germ layer fate decisions of hPSCs reveal targets of human cancers. Single-cell deconstruction of hPSCs-defined subsets that share transcriptional patterns with transformed adult tissues. Chemical screening using a unique germ layer specification assay for hPSCs identified drugs that enriched for compounds that selectively suppressed the growth of patient-derived tumors corresponding exclusively to their germ layer origin. Transcriptional response of hPSCs to germ layer inducing drugs could be used to identify targets capable of regulating hPSC specification as well as inhibiting adult tumors. Our study demonstrates properties of adult tumors converge with hPSCs drug induced differentiation in a germ layer specific manner, thereby expanding our understanding of cancer stemness and pluripotency.


Asunto(s)
Neoplasias , Células Madre Pluripotentes , Humanos , Diferenciación Celular/fisiología , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Genómica
2.
Stem Cells Transl Med ; 12(6): 334-354, 2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-37226319

RESUMEN

Screening of primary patient acute myeloid leukemia (AML) cells is challenging based on intrinsic characteristics of human AML disease and patient-specific conditions required to sustain AML cells in culture. This is further complicated by inter- and intra-patient heterogeneity, and "contaminating" normal cells devoid of molecular AML mutations. Derivation of induced pluripotent stem cells (iPSCs) from human somatic cells has provided approaches for the development of patient-specific models of disease biology and has recently included AML. Although reprogramming patient-derived cancer cells to pluripotency allows for aspects of disease modeling, the major limitation preventing applications and deeper insights using AML-iPSCs is the rarity of success and limited subtypes of AML disease that can be captured by reprogramming to date. Here, we tested and refined methods including de novo, xenografting, naïve versus prime states and prospective isolation for reprogramming AML cells using a total of 22 AML patient samples representing the wide variety of cytogenetic abnormalities. These efforts allowed us to derive genetically matched healthy control (isogenic) lines and capture clones found originally in patients with AML. Using fluorescently activated cell sorting, we revealed that AML reprogramming is linked to the differentiation state of diseased tissue, where use of myeloid marker CD33 compared to the stem cell marker, CD34, reduces reprogramming capture of AML+ clones. Our efforts provide a platform for further optimization of AML-iPSC generation, and a unique library of iPSC derived from patients with AML for detailed cellular and molecular study.


Asunto(s)
Células Madre Pluripotentes Inducidas , Leucemia Mieloide Aguda , Humanos , Reprogramación Celular/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/terapia , Diferenciación Celular/genética , Mutación
3.
Cells ; 11(12)2022 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-35741044

RESUMEN

The generation of human hematopoietic stem cells (HSCs) from human pluripotent stem cells (hPSCs) represents a major goal in regenerative medicine and is believed would follow principles of early development. HSCs arise from a type of endothelial cell called a "hemogenic endothelium" (HE), and human HSCs are experimentally detected by transplantation into SCID or other immune-deficient mouse recipients, termed SCID-Repopulating Cells (SRC). Recently, SRCs were detected by forced expression of seven transcription factors (TF) (ERG, HOXA5, HOXA9, HOXA10, LCOR, RUNX1, and SPI1) in hPSC-derived HE, suggesting these factors are deficient in hPSC differentiation to HEs required to generate HSCs. Here we derived PECAM-1-, Flk-1-, and VE-cadherin-positive endothelial cells that also lack CD45 expression (PFVCD45-) which are solely responsible for hematopoietic output from iPSC lines reprogrammed from AML patients. Using HEs derived from AML patient iPSCs devoid of somatic leukemic aberrations, we sought to generate putative SRCs by the forced expression of 7TFs to model autologous HSC transplantation. The expression of 7TFs in hPSC-derived HE cells from an enhanced hematopoietic progenitor capacity was present in vitro, but failed to acquire SRC activity in vivo. Our findings emphasize the benefits of forced TF expression, along with the continued challenges in developing HSCs for autologous-based therapies from hPSC sources.


Asunto(s)
Hemangioblastos , Células Madre Pluripotentes Inducidas , Leucemia Mieloide Aguda , Animales , Hemangioblastos/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Leucemia Mieloide Aguda/metabolismo , Ratones , Ratones SCID , Factores de Transcripción/metabolismo
4.
Cell Chem Biol ; 28(10): 1394-1406.e10, 2021 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-33979648

RESUMEN

Natural products (NPs) encompass a rich source of bioactive chemical entities. Here, we used human cancer stem cells (CSCs) in a chemical genomics campaign with NP chemical space to interrogate extracts from diverse strains of actinomycete for anti-cancer properties. We identified a compound (McM25044) capable of selectively inhibiting human CSC function versus normal stem cell counterparts. Biochemical and molecular studies revealed that McM025044 exerts inhibition on human CSCs through the small ubiquitin-like modifier (SUMO) cascade, found to be hyperactive in a variety of human cancers. McM025044 impedes the SUMOylation pathway via direct targeting of the SAE1/2 complex. Treatment of patient-derived CSCs resulted in reduced levels of SUMOylated proteins and suppression of progenitor and stem cell capacity measured in vitro and in vivo. Our study overcomes a barrier in chemically inhibiting oncogenic SUMOylation activity and uncovers a unique role for SAE2 in the biology of human cancers.


Asunto(s)
Células Madre Neoplásicas/metabolismo , Enzimas Activadoras de Ubiquitina/metabolismo , Animales , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Sitios de Unión , Productos Biológicos/química , Productos Biológicos/metabolismo , Productos Biológicos/farmacología , Productos Biológicos/uso terapéutico , Línea Celular Tumoral , Autorrenovación de las Células , Supervivencia Celular/efectos de los fármacos , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/patología , Ratones , Simulación del Acoplamiento Molecular , Células Madre Neoplásicas/citología , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Sumoilación/efectos de los fármacos , Enzimas Activadoras de Ubiquitina/química , Enzimas Activadoras de Ubiquitina/genética
5.
Cell Rep ; 34(10): 108818, 2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33691101

RESUMEN

Histone variants (HVs) are a subfamily of epigenetic regulators implicated in embryonic development, but their role in human stem cell fate remains unclear. Here, we reveal that the phosphorylation state of the HV H2A.X (γH2A.X) regulates self-renewal and differentiation of human pluripotent stem cells (hPSCs) and leukemic progenitors. As demonstrated by CRISPR-Cas deletion, H2A.X is essential in maintaining normal hPSC behavior. However, reduced levels of γH2A.X enhances hPSC differentiation toward the hematopoietic lineage with concomitant inhibition of neural development. In contrast, activation and sustained levels of phosphorylated H2A.X enhance hPSC neural fate while suppressing hematopoiesis. This controlled lineage bias correlates to occupancy of γH2A.X at genomic loci associated with ectoderm versus mesoderm specification. Finally, drug modulation of H2A.X phosphorylation overcomes differentiation block of patient-derived leukemic progenitors. Our study demonstrates HVs may serve to regulate pluripotent cell fate and that this biology could be extended to somatic cancer stem cell control.


Asunto(s)
Autorrenovación de las Células/fisiología , Histonas/metabolismo , Células Madre Neoplásicas/citología , Células Madre Pluripotentes/citología , Sistemas CRISPR-Cas/genética , Diferenciación Celular , Linaje de la Célula , Ectodermo/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Histonas/deficiencia , Histonas/genética , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Mesodermo/metabolismo , Células Madre Neoplásicas/metabolismo , Neuronas/citología , Neuronas/metabolismo , Nucleosomas/metabolismo , Fosforilación , Células Madre Pluripotentes/metabolismo
6.
Cancer Cell ; 34(3): 483-498.e5, 2018 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-30205048

RESUMEN

Despite successful remission induction, recurrence of acute myeloid leukemia (AML) remains a clinical obstacle thought to be caused by the retention of dormant leukemic stem cells (LSCs). Using chemotherapy-treated AML xenografts and patient samples, we have modeled patient remission and relapse kinetics to reveal that LSCs are effectively depleted via cell-cycle recruitment, leaving the origins of relapse unclear. Post-chemotherapy, in vivo characterization at the onset of disease relapse revealed a unique molecular state of leukemic-regenerating cells (LRCs) responsible for disease re-growth. LRCs are transient, can only be detected in vivo, and are molecularly distinct from therapy-naive LSCs. We demonstrate that LRC features can be used as markers of relapse and are therapeutically targetable to prevent disease recurrence.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Leucemia Mieloide Aguda/tratamiento farmacológico , Células Progenitoras Mieloides/efectos de los fármacos , Recurrencia Local de Neoplasia/prevención & control , Regeneración/efectos de los fármacos , Adulto , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Ciclo Celular/efectos de los fármacos , Femenino , Humanos , Leucemia Mieloide Aguda/patología , Masculino , Ratones , Células Progenitoras Mieloides/patología , Recurrencia Local de Neoplasia/diagnóstico , Cultivo Primario de Células , Pronóstico , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Stem Cell Reports ; 10(5): 1625-1641, 2018 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-29742393

RESUMEN

Human pluripotent stem cells (hPSCs) generate hematopoietic progenitor cells (HPCs) but fail to engraft xenograft models used to detect adult/somatic hematopoietic stem cells (HSCs) from donors. Recent progress to derive hPSC-derived HSCs has relied on cell-autonomous forced expression of transcription factors; however, the relationship of bone marrow to transplanted cells remains unknown. Here, we quantified a failure of hPSC-HPCs to survive even 24 hr post transplantation. Across several hPSC-HPC differentiation methodologies, we identified the lack of CXCR4 expression and function. Ectopic CXCR4 conferred CXCL12 ligand-dependent signaling of hPSC-HPCs in biochemical assays and increased migration/chemotaxis, hematopoietic progenitor capacity, and survival and proliferation following in vivo transplantation. This was accompanied by a transcriptional shift of hPSC-HPCs toward somatic/adult sources, but this approach failed to produce long-term HSC xenograft reconstitution. Our results reveal that networks involving CXCR4 should be targeted to generate putative HSCs with in vivo function from hPSCs.


Asunto(s)
Quimiocina CXCL12/metabolismo , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Células Madre Pluripotentes/metabolismo , Receptores CXCR4/metabolismo , Transducción de Señal , Animales , Células de la Médula Ósea/metabolismo , Humanos , Cinética , Ratones
8.
Stem Cells ; 35(9): 2095-2102, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28758276

RESUMEN

Induced pluripotent stem cell reprogramming has provided critical insights into disease processes by modeling the genetics and related clinical pathophysiology. Human cancer represents highly diverse genetics, as well as inter- and intra-patient heterogeneity, where cellular model systems capable of capturing this disease complexity would be invaluable. Acute myeloid leukemia (AML) represents one of most heterogeneous cancers and has been divided into genetic subtypes correlated with unique risk stratification over the decades. Here, we report our efforts to induce pluripotency from the heterogeneous population of human patients that represents this disease in the clinic. Using robust optimized reprogramming methods, we demonstrate that reprogramming of AML cells harboring leukemic genomic aberrations is a rare event with the exception of those with de novo mixed-lineage leukemia (MLL) mutations that can be reprogrammed and model drug responses in vitro. Our findings indicate that unlike hematopoietic cells devoid of genomic aberrations, AML cells harboring driver mutations are refractory to reprogramming. Expression of MLL fusion proteins in AML cells did not contribute to induced reprogramming success, which continued to select for patient derived cells devoid of AML patient-specific aberrations. Our study reveals that unanticipated blockades to achieving pluripotency reside within the majority of transformed AML patient cells. Stem Cells 2017;35:2095-2102.


Asunto(s)
Reprogramación Celular , Hematopoyesis , Células Madre Pluripotentes Inducidas/patología , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Mutación/genética , Células de la Médula Ósea/patología , Humanos , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Proteínas de Fusión Oncogénica/metabolismo
9.
Cell Chem Biol ; 24(7): 833-844.e9, 2017 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-28648376

RESUMEN

Targeting of human cancer stem cells (CSCs) requires the identification of vulnerabilities unique to CSCs versus healthy resident stem cells (SCs). Unfortunately, dysregulated pathways that support transformed CSCs, such as Wnt/ß-catenin signaling, are also critical regulators of healthy SCs. Using the ICG-001 and CWP family of small molecules, we reveal Sam68 as a previously unappreciated modulator of Wnt/ß-catenin signaling within CSCs. Disruption of CBP-ß-catenin interaction via ICG-001/CWP induces the formation of a Sam68-CBP complex in CSCs that alters Wnt signaling toward apoptosis and differentiation induction. Our study identifies Sam68 as a regulator of human CSC vulnerability.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Unión al ADN/metabolismo , Células Madre Neoplásicas/metabolismo , Fragmentos de Péptidos/metabolismo , Proteínas de Unión al ARN/metabolismo , Sialoglicoproteínas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/genética , Adulto , Anciano , Animales , Apoptosis/efectos de los fármacos , Compuestos de Azabiciclo/farmacología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Femenino , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Masculino , Ratones , Ratones Endogámicos NOD , Persona de Mediana Edad , Células Madre Neoplásicas/citología , Células Madre Neoplásicas/trasplante , Organofosfatos/farmacología , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Pirimidinonas/farmacología , Interferencia de ARN , Proteínas de Unión al ARN/antagonistas & inhibidores , Proteínas de Unión al ARN/genética , Sialoglicoproteínas/antagonistas & inhibidores , Sialoglicoproteínas/genética , Sumoilación/efectos de los fármacos , Transcriptoma/efectos de los fármacos , Vía de Señalización Wnt/efectos de los fármacos , beta Catenina/metabolismo
10.
Cell Rep ; 19(1): 20-35, 2017 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-28380358

RESUMEN

Human pluripotent stem cells (hPSCs) have been reported in naive and primed states. However, the ability to generate mature cell types remains the imperative property for utility of hPSCs. Here, we reveal that the naive state enhances self-renewal while restricting lineage differentiation in vitro to neural default fate. Molecular analyses indicate expression of multiple lineage-associated transcripts in naive hPSCs that failed to predict biased functional differentiation capacity. Naive hPSCs can be converted to primed state over long-term serial passage that permits recovery of multi-germ layer differentiation. Suppression of OCT4 but not NANOG allows immediate recovery directly from naive state. To this end, we identified chemical inhibitors of OCT4 that restore naive hPSC differentiation. Our study reveals unique cell-fate restrictions in human pluripotent states and provides an approach to overcome these barriers that harness both efficient naive hPSC growth while maintaining in vitro differentiation essential for hPSC applications.


Asunto(s)
Diferenciación Celular/genética , Linaje de la Célula/genética , Reprogramación Celular/genética , Estratos Germinativos/citología , Células Madre Pluripotentes/citología , Animales , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Hepatocitos/metabolismo , Humanos , Ratones , Miocitos Cardíacos/metabolismo , Proteína Homeótica Nanog/metabolismo , Nistatina/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , ARN/genética , Teratoma/metabolismo
11.
Cancer Cell ; 29(1): 61-74, 2016 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-26766591

RESUMEN

Initial pathway alternations required for pathogenesis of human acute myeloid leukemia (AML) are poorly understood. Here we reveal that removal of glycogen synthase kinase-3α (GSK-3α) and GSK-3ß dependency leads to aggressive AML. Although GSK-3α deletion alone has no effect, GSK-3ß deletion in hematopoietic stem cells (HSCs) resulted in a pre-neoplastic state consistent with human myelodysplastic syndromes (MDSs). Transcriptome and functional studies reveal that each GSK-3ß and GSK-3α uniquely contributes to AML by affecting Wnt/Akt/mTOR signaling and metabolism, respectively. The molecular signature of HSCs deleted for GSK-3ß provided a prognostic tool for disease progression and survival of MDS patients. Our study reveals that GSK-3α- and GSK-3ß-regulated pathways can be responsible for stepwise transition to MDS and subsequent AML, thereby providing potential therapeutic targets of disease evolution.


Asunto(s)
Glucógeno Sintasa Quinasa 3/metabolismo , Células Madre Hematopoyéticas/enzimología , Leucemia Mieloide Aguda/enzimología , Animales , Modelos Animales de Enfermedad , Glucógeno Sintasa Quinasa 3/deficiencia , Glucógeno Sintasa Quinasa 3 beta , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/terapia , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/fisiología
12.
Cell Rep ; 11(9): 1367-76, 2015 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-26004181

RESUMEN

The clinical applicability of direct cell fate conversion depends on obtaining tissue from patients that is easy to harvest, store, and manipulate for reprogramming. Here, we generate induced neural progenitor cells (iNPCs) from neonatal and adult peripheral blood using single-factor OCT4 reprogramming. Unlike fibroblasts that share molecular hallmarks of neural crest, OCT4 reprogramming of blood was facilitated by SMAD+GSK-3 inhibition to overcome restrictions on neural fate conversion. Blood-derived (BD) iNPCs differentiate in vivo and respond to guided differentiation in vitro, producing glia (astrocytes and oligodendrocytes) and multiple neuronal subtypes, including dopaminergic (CNS related) and nociceptive neurons (peripheral nervous system [PNS]). Furthermore, nociceptive neurons phenocopy chemotherapy-induced neurotoxicity in a system suitable for high-throughput drug screening. Our findings provide an easily accessible approach for generating human NPCs that harbor extensive developmental potential, enabling the study of clinically relevant neural diseases directly from patient cohorts.


Asunto(s)
Técnicas de Reprogramación Celular/métodos , Células-Madre Neurales/citología , Diferenciación Celular/fisiología , Humanos , Factor 3 de Transcripción de Unión a Octámeros/genética
13.
PLoS One ; 8(7): e67296, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23874411

RESUMEN

Hepatocytes play a central and crucial role in cholesterol and lipid homeostasis, and their proper function is of key importance for cardiovascular health. In particular, hepatocytes (especially periportal hepatocytes) endogenously synthesize large amounts of cholesterol and secrete it into circulating blood via apolipoprotein particles. Cholesterol-secreting hepatocytes are also the clinically-relevant cells targeted by statin treatment in vivo. The study of cholesterol homeostasis is largely restricted to the use of animal models and immortalized cell lines that do not recapitulate those key aspects of normal human hepatocyte function that result from genetic variation of individuals within a population. Hepatocyte-like cells (HLCs) derived from human embryonic and induced pluripotent stem cells can provide a cell culture model for the study of cholesterol homeostasis, dyslipidemias, the action of statins and other pharmaceuticals important for cardiovascular health. We have analyzed expression of core components for cholesterol homeostasis in untreated human iPS cells and in response to pravastatin. Here we show the production of differentiated cells resembling periportal hepatocytes from human pluripotent stem cells. These cells express a broad range of apolipoproteins required for secretion and elimination of serum cholesterol, actively secrete cholesterol into the medium, and respond functionally to statin treatment by reduced cholesterol secretion. Our research shows that HLCs derived from human pluripotent cells provide a robust cell culture system for the investigation of the hepatic contribution to human cholesterol homeostasis at both cellular and molecular levels. Importantly, it permits for the first time to also functionally assess the impact of genetic polymorphisms on cholesterol homeostasis. Finally, the system will also be useful for mechanistic studies of heritable dyslipidemias, drug discovery, and investigation of modes of action of cholesterol-modulatory drugs.


Asunto(s)
Fenómenos Fisiológicos Cardiovasculares , Colesterol/metabolismo , Células Madre Embrionarias/fisiología , Hepatocitos/fisiología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Células Madre Pluripotentes Inducidas/fisiología , Adulto , Apolipoproteínas/metabolismo , Sistema Cardiovascular/metabolismo , Sistema Cardiovascular/fisiopatología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Células Cultivadas , Preescolar , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/fisiología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Homeostasis/efectos de los fármacos , Homeostasis/fisiología , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/fisiología
14.
Cell Reprogram ; 15(2): 151-8, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23550731

RESUMEN

Multiple methods exist that can reprogram differentiated cells to a pluripotent state similar to that of embryonic stem cells (ESCs). These include somatic cell nuclear transfer (SCNT), fusion-mediated reprogramming (FMR) of somatic cells with ESCs, and the production of induced pluripotent stem cells (iPSCs). All of these methods yield cells in which the endogenous Oct4 gene is reactivated. We were interested in comparing the activity of the Oct4 promoter in three different classes of pluripotent cells, including normal ESCs, FMR cells (FMRCs), and iPSCs. We prepared cells of all three types that harbor a transgene composed of the mouse Oct4 promoter driving green fluorescent protein (Oct4-GFP). All cell derivations started with a characterized transgenic Oct4-GFP mouse, and from this we derived ESCs, FMRCs, and iPSCs with the Oct4-GFP transgene present in an identical genomic integration site in all three cell types. Using flow cytometry we assessed Oct4 promoter expression, cell cycle behavior, and differentiation kinetics. We found similar levels of GFP expression in all three cell types and no significant alterations in pluripotency or differentiation. Our results suggest that the pluripotent condition is a potent "local attractor" state, because it can be achieved through three vastly different avenues.


Asunto(s)
Transdiferenciación Celular , Células Madre Embrionarias/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/biosíntesis , Células Madre Pluripotentes/metabolismo , Regiones Promotoras Genéticas , Animales , Línea Celular , Células Madre Embrionarias/citología , Ratones , Ratones Transgénicos , Factor 3 de Transcripción de Unión a Octámeros/genética , Células Madre Pluripotentes/citología , Transgenes
15.
PLoS One ; 6(6): e21512, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21738686

RESUMEN

Macrohistones (mH2As) are unusual histone variants found exclusively in vertebrate chromatin. In mice, the H2afy gene encodes two splice variants, mH2A1.1 and mH2A1.2 and a second gene, H2afy2, encodes an additional mH2A2 protein. Both mH2A isoforms have been found enriched on the inactive X chromosome (Xi) in differentiated mammalian female cells, and are incorporated into the chromatin of developmentally-regulated genes. To investigate the functional significance of mH2A isoforms for X chromosome inactivation (XCI), we produced male and female embryonic stem cell (ESC) lines with stably-integrated shRNA constructs that simultaneously target both mH2A1 and mH2A2. Surprisingly, we find that female ESCs deficient for both mH2A1 and mH2A2 readily execute and maintain XCI upon differentiation. Furthermore, male and female mH2A-deficient ESCs proliferate normally under pluripotency culture conditions, and respond to several standard differentiation procedures efficiently. Our results show that XCI can readily proceed with substantially reduced total mH2A content.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Histonas/deficiencia , Inactivación del Cromosoma X/fisiología , Animales , Western Blotting , Diferenciación Celular/genética , Femenino , Técnica del Anticuerpo Fluorescente , Histonas/genética , Masculino , Ratones , Placa Neural/citología , Placa Neural/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Teratoma/genética , Teratoma/metabolismo , Inactivación del Cromosoma X/genética
16.
Int J Dev Biol ; 54(11-12): 1545-64, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21404178

RESUMEN

Pluripotent cells of the blastocyst inner cell mass (ICM) and their in vitro derivatives, embryonic stem (ES) cells, contain genomes in an epigenetic state that are poised for subsequent differentiation. Their chromatin is hyperdynamic in nature and relatively uncondensed. In addition, a large number of genes are expressed at low levels in both ICM and ES cells. Also, the chromatin of naturally pluripotent cells contains specialized histone modification patterns such as bivalent domains, which mark genes destined for later developmentally-regulated expression states. Female pluripotent cells contain X chromosomes that have yet to undergo the process of X chromosome inactivation. Collectively, these features of very early embyronic chromatin are required for the successful specification and production of differentiated cell lineages. Artificial reprogramming methods such as somatic nuclear transfer (SCNT), ES cell fusion-mediated reprogramming (FMR), and induced pluripotency (iPS) yield pluripotent cells that recapitulate many features of naturally pluripotent cells, including many of their epigenetic features. However, the route to pluripotent epigenomic states in artificial pluripotent cells differs drastically from that of their natural counterparts. Here, we compare and contrast the differing routes to pluripotency under natural and artificial conditions. In addition, we discuss the intrinsically metastable nature of the pluripotent epigenome and consider epigenetic aspects of reprogramming that may lead to incomplete or inaccurate reprogrammed states. Artificial methods of reprogramming hold immense promise for the development of autologous cell graft sources and for the development of cell culture models for human genetic disorders. However, the utility of artificially reprogrammed cells is highly dependent on the fidelity of the reprogramming process and it is therefore critically important to assess the epigenetic similarities between embryonic and induced pluripotent stem cells.


Asunto(s)
Reprogramación Celular/genética , Células Madre Embrionarias , Células Madre Pluripotentes , Animales , Diferenciación Celular/genética , Células Madre Embrionarias/citología , Epigénesis Genética , Regulación del Desarrollo de la Expresión Génica/genética , Humanos , Células Madre Pluripotentes/citología
17.
Birth Defects Res C Embryo Today ; 87(4): 297-313, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19960551

RESUMEN

Following fertilization, the newly formed zygote faces several critical decisions regarding cell fate and lineage commitment. First, the parental genomes must be reprogrammed and reset for the zygotic genome to assume responsibility for gene expression. Second, blastomeres must be committed to form either the inner cell mass or trophectoderm before implantation. A variety of epigenetic mechanisms underlies each of these steps, allowing for proper activation of transcriptional circuits which function to specify a cell's identity and maintain or adjust that state as developmental and environmental conditions dictate. These epigenetic mechanisms encompass DNA methylation, post-translational histone modification, chromatin remodeling, and alterations in nuclear architecture. In recent years, stem cells derived from the inner cell mass have been used to examine the epigenetic pathways that regulate pluripotency, differentiation, and lineage commitment. From a technical standpoint, embryonic stem cells provide an easier system to work with compared to preimplantation embryos; however, it is currently unknown how closely the epigenetic mechanisms of cultured stem cells resemble their counterparts in the intact embryo. Furthermore, it remains unclear how similar the reprogramming pathways in artificially created systems, such as nuclear transfer-derived embryos and induced pluripotent stem cells, are to those in naturally created embryos. In this review, we summarize the current knowledge of epigenetic influences during preimplantation development and shed light on the extent to which these pathways are conserved in cultured pluripotent cells in vitro. In doing so, we demonstrate the critical role that epigenetic mechanisms play in the establishment of cell fate during the earliest stages of mammalian development.


Asunto(s)
Desarrollo Embrionario/genética , Epigénesis Genética , Animales , Ensamble y Desensamble de Cromatina , Metilación de ADN , Células Madre Embrionarias/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Histonas/metabolismo , Humanos , Modelos Genéticos , Oocitos/metabolismo , Células Madre Pluripotentes/metabolismo , Embarazo , ARN no Traducido/genética , Inactivación del Cromosoma X , Cigoto/metabolismo
18.
Epigenetics ; 4(5): 330-8, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19571681

RESUMEN

Human embryonic stem cells (hESCs) can be maintained in culture over a large number of passages while maintaining apparently normal colony morphology. However, recent reports describe variability in epigenetic states in comparisons among different human ES cell lines. These epigenetic differences include changes in CpG methylation, expression of imprinted genes, and the status of X chromosome inactivation (XCI). We report here that the status of XCI in the female hESC line H9 (WA09) is hypervariable. We find that XIST expression can differ between individual culture isolates of H9. In addition, we find that XIST expression status can vary even between different colonies present within the same H9 culture, effectively rendering the culture mosaic. H9 cultures that lack XIST expression, but have cytological evidence of completed XCI, can also exhibit altered response to BMP4, a growth factor known to induce differentiation of hESCs to a trophectodermal lineage. In the same cultures we find biallelic expression of X-linked genes suggesting that these lines consist of mixtures of cells that retain inactivation of one of two X chromosomes following random choice. Prolonged culture of the XIST-negative isolates to high passage numbers did not result in changes in global epiproteomic signatures, demonstrating rather stable levels of post-translational nucleosome modifications within the culture-adapted hESC lines. The results show that epigenetic variants arise within human ES cell cultures after cell line derivation. In addition, the results indicate that apparently normal cultures of hESCs may contain mixtures of cells with differing epigenetic states. Assays of epigenetic integrity are warranted as quality control measures for the culture of hESCs.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Epigénesis Genética , Animales , Secuencia de Bases , Proteína Morfogenética Ósea 4/farmacología , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Análisis Citogenético , Células Madre Embrionarias/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Genes Ligados a X , Histonas/metabolismo , Humanos , Ratones , Datos de Secuencia Molecular , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Polimorfismo de Nucleótido Simple/genética , Procesamiento Proteico-Postraduccional/efectos de los fármacos , ARN Largo no Codificante , ARN no Traducido/genética , ARN no Traducido/metabolismo
19.
Stem Cells ; 25(5): 1104-13, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17272499

RESUMEN

Recent experiments demonstrate that somatic nuclei can be reprogrammed to a pluripotent state when fused to ESCs. The resulting hybrids are pluripotent as judged by developmental assays, but detailed analyses of the underlying molecular-genetic control of reprogrammed transcription in such hybrids are required to better understand fusion-mediated reprogramming. We produced hybrids of mouse ESCs and fibroblasts that, although nearly tetraploid, exhibit characteristics of normal ESCs, including apparent immortality in culture, ESC-like colony morphology, and pluripotency. Comprehensive analysis of the mouse embryonic fibroblast/ESC hybrid transcriptome revealed global patterns of gene expression reminiscent of ESCs. However, combined analysis of variance and hierarchical clustering analyses revealed at least seven distinct classes of differentially regulated genes in comparisons of hybrids, ESCs, and somatic cells. The largest class includes somatic genes that are silenced in hybrids and ESCs, but a smaller class includes genes that are expressed at nearly equivalent levels in hybrids and ESCs that contain many genes implicated in pluripotency and chromatin function. Reprogrammed genes are distributed throughout the genome. Reprogramming events include both transcriptional silencing and activation of genes residing on chromosomes of somatic origin. Somatic/ESC hybrid cell lines resemble their pre-fusion ESC partners in terms of behavior in culture and pluripotency. However, they contain unique expression profiles that are similar but not identical to normal ESCs. ESC fusion-mediated reprogramming provides a tractable system for the investigation of mechanisms of reprogramming. Disclosure of potential conflicts of interest is found at the end of this article.


Asunto(s)
Reprogramación Celular/genética , Quimera/genética , Células Madre Embrionarias/metabolismo , Genoma , Alelos , Animales , Secuencia de Bases , Línea Celular , Cromosomas de los Mamíferos/genética , Análisis por Conglomerados , Análisis Mutacional de ADN , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Cariotipificación , Ratones , Datos de Secuencia Molecular , Células Madre Pluripotentes/metabolismo , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...