Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Cell ; 168(1-2): 59-72.e13, 2017 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-28065413

RESUMEN

Chromosomal translocations of the mixed-lineage leukemia (MLL) gene with various partner genes result in aggressive leukemia with dismal outcomes. Despite similar expression at the mRNA level from the wild-type and chimeric MLL alleles, the chimeric protein is more stable. We report that UBE2O functions in regulating the stability of wild-type MLL in response to interleukin-1 signaling. Targeting wild-type MLL degradation impedes MLL leukemia cell proliferation, and it downregulates a specific group of target genes of the MLL chimeras and their oncogenic cofactor, the super elongation complex. Pharmacologically inhibiting this pathway substantially delays progression, and it improves survival of murine leukemia through stabilizing wild-type MLL protein, which displaces the MLL chimera from some of its target genes and, therefore, relieves the cellular oncogenic addiction to MLL chimeras. Stabilization of MLL provides us with a paradigm in the development of therapies for aggressive MLL leukemia and perhaps for other cancers caused by translocations.


Asunto(s)
Leucemia Bifenotípica Aguda/tratamiento farmacológico , Leucemia Bifenotípica Aguda/metabolismo , Proteolisis/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Interleucina-1/metabolismo , Quinasas Asociadas a Receptores de Interleucina-1/antagonistas & inhibidores , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Enzimas Ubiquitina-Conjugadoras
2.
Cell ; 150(2): 351-65, 2012 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-22817897

RESUMEN

Wnt signaling is involved in self-renewal and maintenance of hematopoietic stem cells (HSCs); however, the particular role of noncanonical Wnt signaling in regulating HSCs in vivo is largely unknown. Here, we show Flamingo (Fmi) and Frizzled (Fz) 8, members of noncanonical Wnt signaling, both express in and functionally maintain quiescent long-term HSCs. Fmi regulates Fz8 distribution at the interface between HSCs and N-cadherin(+) osteoblasts (N-cad(+)OBs that enrich osteoprogenitors) in the niche. We further found that N-cad(+)OBs predominantly express noncanonical Wnt ligands and inhibitors of canonical Wnt signaling under homeostasis. Under stress, noncanonical Wnt signaling is attenuated and canonical Wnt signaling is enhanced in activation of HSCs. Mechanistically, noncanonical Wnt signaling mediated by Fz8 suppresses the Ca(2+)-NFAT- IFNγ pathway, directly or indirectly through the CDC42-CK1α complex and also antagonizes canonical Wnt signaling in HSCs. Taken together, our findings demonstrate that noncanonical Wnt signaling maintains quiescent long-term HSCs through Fmi and Fz8 interaction in the niche.


Asunto(s)
Cadherinas/metabolismo , Células Madre Hematopoyéticas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Nicho de Células Madre , Vía de Señalización Wnt , Animales , Cadherinas/genética , Femenino , Células Madre Hematopoyéticas/citología , Humanos , Interferón gamma/metabolismo , Ratones , Ratones Transgénicos , Factores de Transcripción NFATC/metabolismo , Receptores Acoplados a Proteínas G/genética
3.
Cell ; 136(3): 508-20, 2009 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-19203584

RESUMEN

Caspase-2 is unique among all the mammalian caspases in that it is the only caspase that is present constitutively in the cell nucleus, in addition to other cellular compartments. However, the functional significance of this nuclear localization is unknown. Here we show that DNA damage induced by gamma-radiation triggers the phosphorylation of nuclear caspase-2 at the S122 site within its prodomain, leading to its cleavage and activation. This phosphorylation is carried out by the nuclear serine/threonine protein kinase DNA-PKcs and promoted by the p53-inducible death-domain-containing protein PIDD within a large nuclear protein complex consisting of DNA-PKcs, PIDD, and caspase-2, which we have named the DNA-PKcs-PIDDosome. This phosphorylation and the catalytic activity of caspase-2 are involved in the maintenance of a G2/M DNA damage checkpoint and DNA repair mediated by the nonhomologous end-joining (NHEJ) pathway. The DNA-PKcs-PIDDosome thus represents a protein complex that impacts mammalian G2/M DNA damage checkpoint and NHEJ.


Asunto(s)
Proteínas Portadoras/metabolismo , Caspasa 2/metabolismo , Ciclo Celular , Cisteína Endopeptidasas/metabolismo , Proteína Quinasa Activada por ADN/metabolismo , Proteínas Nucleares/metabolismo , Secuencia de Aminoácidos , Animales , Caspasa 2/química , Línea Celular , Cisteína Endopeptidasas/química , Daño del ADN , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte , Fibroblastos/metabolismo , Rayos gamma , Humanos , Ratones , Mitosis , Datos de Secuencia Molecular , Alineación de Secuencia
4.
Mol Cell ; 60(3): 435-45, 2015 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-26527278

RESUMEN

Although it is established that some general transcription factors are inactivated at mitosis, many details of mitotic transcription inhibition (MTI) and its underlying mechanisms are largely unknown. We have identified mitotic transcriptional activation (MTA) as a key regulatory step to control transcription in mitosis for genes with transcriptionally engaged RNA polymerase II (Pol II) to activate and transcribe until the end of the gene to clear Pol II from mitotic chromatin, followed by global impairment of transcription reinitiation through MTI. Global nascent RNA sequencing and RNA fluorescence in situ hybridization demonstrate the existence of transcriptionally engaged Pol II in early mitosis. Both genetic and chemical inhibition of P-TEFb in mitosis lead to delays in the progression of cell division. Together, our study reveals a mechanism for MTA and MTI whereby transcriptionally engaged Pol II can progress into productive elongation and finish transcription to allow proper cellular division.


Asunto(s)
ADN Polimerasa II/metabolismo , Mitosis/fisiología , Factor B de Elongación Transcripcional Positiva/metabolismo , Elongación de la Transcripción Genética/fisiología , Activación Transcripcional/fisiología , Células HEK293 , Células HeLa , Humanos
5.
Nature ; 500(7462): 345-9, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23863936

RESUMEN

The epigenetic regulation of imprinted genes by monoallelic DNA methylation of either maternal or paternal alleles is critical for embryonic growth and development. Imprinted genes were recently shown to be expressed in mammalian adult stem cells to support self-renewal of neural and lung stem cells; however, a role for imprinting per se in adult stem cells remains elusive. Here we show upregulation of growth-restricting imprinted genes, including in the H19-Igf2 locus, in long-term haematopoietic stem cells and their downregulation upon haematopoietic stem cell activation and proliferation. A differentially methylated region upstream of H19 (H19-DMR), serving as the imprinting control region, determines the reciprocal expression of H19 from the maternal allele and Igf2 from the paternal allele. In addition, H19 serves as a source of miR-675, which restricts Igf1r expression. We demonstrate that conditional deletion of the maternal but not the paternal H19-DMR reduces adult haematopoietic stem cell quiescence, a state required for long-term maintenance of haematopoietic stem cells, and compromises haematopoietic stem cell function. Maternal-specific H19-DMR deletion results in activation of the Igf2-Igfr1 pathway, as shown by the translocation of phosphorylated FoxO3 (an inactive form) from nucleus to cytoplasm and the release of FoxO3-mediated cell cycle arrest, thus leading to increased activation, proliferation and eventual exhaustion of haematopoietic stem cells. Mechanistically, maternal-specific H19-DMR deletion leads to Igf2 upregulation and increased translation of Igf1r, which is normally suppressed by H19-derived miR-675. Similarly, genetic inactivation of Igf1r partly rescues the H19-DMR deletion phenotype. Our work establishes a new role for this unique form of epigenetic control at the H19-Igf2 locus in maintaining adult stem cells.


Asunto(s)
Células Madre Adultas/citología , Células Madre Adultas/fisiología , Impresión Genómica , Factor II del Crecimiento Similar a la Insulina/genética , Factor II del Crecimiento Similar a la Insulina/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Animales , Epigénesis Genética/genética , Regulación del Desarrollo de la Expresión Génica , Ratones , Receptor IGF Tipo 1/genética , Transducción de Señal , Activación Transcripcional
6.
Genes Dev ; 25(18): 1928-42, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21890648

RESUMEN

Although self-renewal is the central property of stem cells, the underlying mechanism remains inadequately defined. Using a hematopoietic stem and progenitor cell (HSPC)-specific conditional induction line, we generated a compound genetic model bearing both Pten deletion and ß-catenin activation. These double mutant mice exhibit a novel phenotype, including expansion of phenotypic long-term hematopoietic stem cells (LT-HSCs) without extensive differentiation. Unexpectedly, constitutive activation of ß-catenin alone results in apoptosis of HSCs. However, together, the Wnt/ß-catenin and PTEN/PI3k/Akt pathways interact to drive phenotypic LT-HSC expansion by inducing proliferation while simultaneously inhibiting apoptosis and blocking differentiation, demonstrating the necessity of complementary cooperation between the two pathways in promoting self-renewal. Mechanistically, ß-catenin activation reduces multiple differentiation-inducing transcription factors, blocking differentiation partially through up-regulation of Inhibitor of differentiation 2 (Id2). In double mutants, loss of Pten enhances the HSC anti-apoptotic factor Mcl-1. All of these contribute in a complementary way to HSC self-renewal and expansion. While permanent, genetic alteration of both pathways in double mutant mice leads to expansion of phenotypic HSCs, these HSCs cannot function due to blocked differentiation. We developed a pharmacological approach to expand normal, functional HSCs in culture using factors that reversibly activate both Wnt/ß-catenin and PI3K/Akt signaling simultaneously. We show for the first time that activation of either single pathway is insufficient to expand primitive HSCs, but in combination, both pathways drive self-renewal and expansion of HSCs with long-term functional capacity.


Asunto(s)
Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animales , Apoptosis , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Línea Celular , Proliferación Celular , Células Cultivadas , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/enzimología , Ratones , Mutación , Fosfohidrolasa PTEN/genética , Fenotipo , Fosfatidilinositol 3-Quinasas/genética , Proteínas Wnt/genética , beta Catenina/genética
7.
Development ; 141(5): 1095-103, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24550117

RESUMEN

Embryonic cells that migrate long distances must critically balance cell division in order to maintain stream dynamics and population of peripheral targets. Yet details of individual cell division events and how cell cycle is related to phases of migration remain unclear. Here, we examined these questions using the chick cranial neural crest (NC). In vivo time-lapse imaging revealed that a typical migrating NC cell division event lasted ~1 hour and included four stereotypical steps. Cell tracking showed that dividing NC cells maintained position relative to non-dividing neighbors. NC cell division orientation and the time and distance to first division after neural tube exit were stochastic. To address how cell cycle is related to phases of migration, we used FACs analysis to identify significant spatiotemporal differences in NC cell cycle profiles. Two-photon photoconversion of single and small numbers of mKikGR-labeled NC cells confirmed that lead NC cells exhibited a nearly fourfold faster doubling time after populating the branchial arches. By contrast, Ki-67 staining showed that one out of every five later emerging NC cells exited the cell cycle after reaching proximal head targets. The relatively quiescent mitotic activity during NC cell migration to the branchial arches was altered when premigratory cells were reduced in number by tissue ablation. Together, our results provide the first comprehensive details of the pattern and dynamics of cell division events during cranial NC cell migration.


Asunto(s)
Cresta Neural/citología , Animales , Ciclo Celular/genética , Ciclo Celular/fisiología , División Celular/genética , División Celular/fisiología , Movimiento Celular/genética , Movimiento Celular/fisiología , Proliferación Celular , Embrión de Pollo , Citometría de Flujo , Cresta Neural/metabolismo
8.
Gastroenterology ; 145(2): 383-95.e1-21, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23644405

RESUMEN

BACKGROUND & AIMS: Identification of intestinal stem cells (ISCs) has relied heavily on the use of transgenic reporters in mice, but this approach is limited by mosaic expression patterns and difficult to directly apply to human tissues. We sought to identify reliable surface markers of ISCs and establish a robust functional assay to characterize ISCs from mouse and human tissues. METHODS: We used immunohistochemistry, real-time reverse-transcription polymerase chain reaction, and fluorescence-activated cell sorting (FACS) to analyze intestinal epithelial cells isolated from mouse and human intestinal tissues. We compared different combinations of surface markers among ISCs isolated based on expression of Lgr5-green fluorescent protein. We developed a culture protocol to facilitate the identification of functional ISCs from mice and then tested the assay with human intestinal crypts and putative ISCs. RESULTS: CD44(+)CD24(lo)CD166(+) cells, isolated by FACS from mouse small intestine and colon, expressed high levels of stem cell-associated genes. Transit-amplifying cells and progenitor cells were then excluded based on expression of GRP78 or c-Kit. CD44(+)CD24(lo)CD166(+) GRP78(lo/-) putative stem cells from mouse small intestine included Lgr5-GFP(hi) and Lgr5-GFP(med/lo) cells. Incubation of these cells with the GSK inhibitor CHIR99021 and the E-cadherin stabilizer Thiazovivin resulted in colony formation by 25% to 30% of single-sorted ISCs. CONCLUSIONS: We developed a culture protocol to identify putative ISCs from mouse and human tissues based on cell surface markers. CD44(+)CD24(lo)CD166(+), GRP78(lo/-), and c-Kit(-) facilitated identification of putative stem cells from the mouse small intestine and colon, respectively. CD44(+)CD24(-/lo)CD166(+) also identified putative human ISCs. These findings will facilitate functional studies of mouse and human ISCs.


Asunto(s)
Células Madre Adultas/metabolismo , Antígenos de Superficie/metabolismo , Mucosa Intestinal/citología , Molécula de Adhesión Celular del Leucocito Activado/metabolismo , Animales , Antígeno CD24/metabolismo , Técnicas de Cultivo de Célula , Colon/citología , Ensayo de Unidades Formadoras de Colonias , Chaperón BiP del Retículo Endoplásmico , Citometría de Flujo , Proteínas de Choque Térmico/genética , Humanos , Receptores de Hialuranos/metabolismo , Intestino Delgado/citología , Ratones , Proteínas Proto-Oncogénicas c-kit/metabolismo
9.
Blood ; 120(9): 1831-42, 2012 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-22802336

RESUMEN

Previous studies have shown that fibroblast growth factor (FGF) signaling promotes hematopoietic stem and progenitor cell (HSPC) expansion in vitro. However, it is unknown whether FGF promotes HSPC expansion in vivo. Here we examined FGF receptor 1 (FGFR1) expression and investigated its in vivo function in HSPCs. Conditional knockout (CKO) of Fgfr1 did not affect phenotypical number of HSPCs and homeostatic hematopoiesis, but led to a reduced engraftment only in the secondary transplantation. When treated with 5-fluorouracil (5FU), the Fgfr1 CKO mice showed defects in both proliferation and subsequent mobilization of HSPCs. We identified megakaryocytes (Mks) as a major resource for FGF production, and further discovered a novel mechanism by which Mks underwent FGF-FGFR signaling dependent expansion to accelerate rapid FGF production under stress. Within HSPCs, we observed an up-regulation of nuclear factor κB and CXCR4, a receptor for the chemoattractant SDF-1, in response to bone marrow damage only in control but not in Fgfr1 CKO model, accounting for the corresponding defects in proliferation and migration of HSPCs. This study provides the first in vivo evidence that FGF signaling facilitates postinjury recovery of the mouse hematopoietic system by promoting proliferation and facilitating mobilization of HSPCs.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Sistema Hematopoyético/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal , Animales , Antimetabolitos Antineoplásicos/farmacología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Movimiento Celular/genética , Proliferación Celular , Células Cultivadas , Femenino , Citometría de Flujo , Fluorouracilo/farmacología , Expresión Génica/efectos de los fármacos , Sistema Hematopoyético/citología , Sistema Hematopoyético/efectos de los fármacos , Inmunohistoquímica , Masculino , Megacariocitos/efectos de los fármacos , Megacariocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , FN-kappa B/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptores CXCR4/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
Nature ; 441(7092): 518-22, 2006 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-16633340

RESUMEN

Haematopoietic stem cells (HSCs) must achieve a balance between quiescence and activation that fulfils immediate demands for haematopoiesis without compromising long-term stem cell maintenance, yet little is known about the molecular events governing this balance. Phosphatase and tensin homologue (PTEN) functions as a negative regulator of the phosphatidylinositol-3-OH kinase (PI(3)K)-Akt pathway, which has crucial roles in cell proliferation, survival, differentiation and migration. Here we show that inactivation of PTEN in bone marrow HSCs causes their short-term expansion, but long-term decline, primarily owing to an enhanced level of HSC activation. PTEN-deficient HSCs engraft normally in recipient mice, but have an impaired ability to sustain haematopoietic reconstitution, reflecting the dysregulation of their cell cycle and decreased retention in the bone marrow niche. Mice with PTEN-mutant bone marrow also have an increased representation of myeloid and T-lymphoid lineages and develop myeloproliferative disorder (MPD). Notably, the cell populations that expand in PTEN mutants match those that become dominant in the acute myeloid/lymphoid leukaemia that develops in the later stages of MPD. Thus, PTEN has essential roles in restricting the activation of HSCs, in lineage fate determination, and in the prevention of leukaemogenesis.


Asunto(s)
Linaje de la Célula , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Leucemia/patología , Leucemia/prevención & control , Fosfohidrolasa PTEN/metabolismo , Animales , Recuento de Células , Ciclo Celular , Proliferación Celular , Ciclina D1/metabolismo , Expresión Génica , Leucemia/metabolismo , Ratones , Mutación/genética , Fosfohidrolasa PTEN/deficiencia , Fosfohidrolasa PTEN/genética
11.
Mol Omics ; 17(1): 59-65, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-32924050

RESUMEN

Machine learning and topological analysis methods are becoming increasingly used on various large-scale omics datasets. Modern high dimensional flow cytometry data sets share many features with other omics datasets like genomics and proteomics. For example, genomics or proteomics datasets can be sparse and have high dimensionality, and flow cytometry datasets can also share these features. This makes flow cytometry data potentially a suitable candidate for employing machine learning and topological scoring strategies, for example, to gain novel insights into patterns within the data. We have previously developed a Topological Score (TopS) and implemented it for the analysis of quantitative protein interaction network datasets. Here we show that TopS approach for large scale data analysis is applicable to the analysis of a previously described flow cytometry sorted human hematopoietic stem cell dataset. We demonstrate that TopS is capable of effectively sorting this dataset into cell populations and identify rare cell populations. We demonstrate the utility of TopS when coupled with multiple approaches including topological data analysis, X-shift clustering, and t-Distributed Stochastic Neighbor Embedding (t-SNE). Our results suggest that TopS could be effectively used to analyze large scale flow cytometry datasets to find rare cell populations.


Asunto(s)
Citometría de Flujo/métodos , Aprendizaje Automático , Análisis de la Célula Individual/métodos , Células Madre/metabolismo , Algoritmos , Células Madre Hematopoyéticas , Humanos , Células Madre/citología
12.
Cell Rep ; 36(10): 109674, 2021 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-34496236

RESUMEN

Tumor-initiating stem cells (TSCs) are critical for drug resistance and immune escape. However, the mutual regulations between TSC and tumor microenvironment (TME) remain unclear. Using DNA-label retaining, single-cell RNA sequencing (scRNA-seq), and other approaches, we investigated intestinal adenoma in response to chemoradiotherapy (CRT), thus identifying therapy-resistant TSCs (TrTSCs). We find bidirectional crosstalk between TSCs and TME using CellPhoneDB analysis. An intriguing finding is that TSCs shape TME into a landscape that favors TSCs for immunosuppression and propagation. Using adenoma-organoid co-cultures, niche-cell depletion, and lineaging tracing, we characterize a functional role of cyclooxygenase-2 (Cox-2)-dependent signaling, predominantly occurring between tumor-associated monocytes and macrophages (TAMMs) and TrTSCs. We show that TAMMs promote TrTSC proliferation through prostaglandin E2 (PGE2)-PTGER4(EP4) signaling, which enhances ß-catenin activity via AKT phosphorylation. Thus, our study shows that the bidirectional crosstalk between TrTSC and TME results in a pro-tumorigenic and immunosuppressive contexture.


Asunto(s)
Carcinogénesis/patología , Forma de la Célula/fisiología , Células Madre Neoplásicas/patología , Microambiente Tumoral/fisiología , Animales , Ciclooxigenasa 2/metabolismo , Dinoprostona/metabolismo , Humanos , Intestinos/metabolismo , Ratones , Organoides/metabolismo
13.
Elife ; 82019 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-30681411

RESUMEN

Loss of sensory hair cells leads to deafness and balance deficiencies. In contrast to mammalian hair cells, zebrafish ear and lateral line hair cells regenerate from poorly characterized support cells. Equally ill-defined is the gene regulatory network underlying the progression of support cells to differentiated hair cells. scRNA-Seq of lateral line organs uncovered five different support cell types, including quiescent and activated stem cells. Ordering of support cells along a developmental trajectory identified self-renewing cells and genes required for hair cell differentiation. scRNA-Seq analyses of fgf3 mutants, in which hair cell regeneration is increased, demonstrates that Fgf and Notch signaling inhibit proliferation of support cells in parallel by inhibiting Wnt signaling. Our scRNA-Seq analyses set the foundation for mechanistic studies of sensory organ regeneration and is crucial for identifying factors to trigger hair cell production in mammals. The data is searchable and publicly accessible via a web-based interface.


Asunto(s)
Proliferación Celular , Factores de Crecimiento de Fibroblastos/metabolismo , Células Ciliadas Auditivas/citología , ARN Citoplasmático Pequeño/genética , Receptores Notch/metabolismo , Transducción de Señal , Células Madre/metabolismo , Animales , Pez Cebra
14.
Cell Res ; 28(9): 904-917, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30065315

RESUMEN

Transplantation of hematopoietic stem cells (HSCs) from human umbilical cord blood (hUCB) holds great promise for treating a broad spectrum of hematological disorders including cancer. However, the limited number of HSCs in a single hUCB unit restricts its widespread use. Although extensive efforts have led to multiple methods for ex vivo expansion of human HSCs by targeting single molecules or pathways, it remains unknown whether it is possible to simultaneously manipulate the large number of targets essential for stem cell self-renewal. Recent studies indicate that N6-methyladenosine (m6A) modulates the expression of a group of mRNAs critical for stem cell-fate determination by influencing their stability. Among several m6A readers, YTHDF2 is recognized as promoting targeted mRNA decay. However, the physiological functions of YTHDF2 in adult stem cells are unknown. Here we show that following the conditional knockout (KO) of mouse Ythdf2 the numbers of functional HSC were increased without skewing lineage differentiation or leading to hematopoietic malignancies. Furthermore, knockdown (KD) of human YTHDF2 led to more than a 10-fold increase in the ex vivo expansion of hUCB HSCs, a fivefold increase in colony-forming units (CFUs), and more than an eightfold increase in functional hUCB HSCs in the secondary serial of a limiting dilution transplantation assay. Mapping of m6A in RNAs from mouse hematopoietic stem and progenitor cells (HSPCs) as well as from hUCB HSCs revealed its enrichment in mRNAs encoding transcription factors critical for stem cell self-renewal. These m6A-marked mRNAs were recognized by Ythdf2 and underwent decay. In Ythdf2 KO HSPCs and YTHDF2 KD hUCB HSCs, these mRNAs were stabilized, facilitating HSC expansion. Knocking down one of YTHDF2's key targets, Tal1 mRNA, partially rescued the phenotype. Our study provides the first demonstration of the function of YTHDF2 in adult stem cell maintenance and identifies its important role in regulating HSC ex vivo expansion by regulating the stability of multiple mRNAs critical for HSC self-renewal, thus identifying potential for future clinical applications.


Asunto(s)
Adenosina/análogos & derivados , Autorrenovación de las Células , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Proteínas de Unión al ARN/antagonistas & inhibidores , Proteínas de Unión al ARN/metabolismo , Adenosina/metabolismo , Animales , Células Madre Hematopoyéticas/patología , Ratones , Ratones Noqueados
15.
Cell Res ; 28(10): 1042, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30150672

RESUMEN

In the initial published version of this article, there was an inadvertent omission from the Acknowledgements that this work was supported by Stowers Institute for Medical Research (SIMR-1004) and NIH National Cancer Institute grant to University of Kansas Cancer Center (P30 CA168524). This omission does not affect the description of the results or the conclusions of this work.

16.
Cell Stem Cell ; 22(5): 740-754.e7, 2018 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-29727682

RESUMEN

Hox genes modulate the properties of hematopoietic stem cells (HSCs) and reacquired Hox expression in progenitors contributes to leukemogenesis. Here, our transcriptome and DNA methylome analyses revealed that Hoxb cluster and retinoid signaling genes are predominantly enriched in LT-HSCs, and this coordinate regulation of Hoxb expression is mediated by a retinoid-dependent cis-regulatory element, distal element RARE (DERARE). Deletion of the DERARE reduced Hoxb expression, resulting in changes to many downstream signaling pathways (e.g., non-canonical Wnt signaling) and loss of HSC self-renewal and reconstitution capacity. DNA methyltransferases mediate DNA methylation on the DERARE, leading to reduced Hoxb cluster expression. Acute myeloid leukemia patients with DNMT3A mutations exhibit DERARE hypomethylation, elevated HOXB expression, and adverse outcomes. CRISPR-Cas9-mediated specific DNA methylation at DERARE attenuated HOXB expression and alleviated leukemogenesis. Collectively, these findings demonstrate pivotal roles for retinoid signaling and the DERARE in maintaining HSCs and preventing leukemogenesis by coordinate regulation of Hoxb genes.


Asunto(s)
Epigénesis Genética/efectos de los fármacos , Hematopoyesis/efectos de los fármacos , Proteínas de Homeodominio/antagonistas & inhibidores , Retinoides/farmacología , Animales , Elementos de Facilitación Genéticos/efectos de los fármacos , Elementos de Facilitación Genéticos/genética , Epigénesis Genética/genética , Células HEK293 , Hematopoyesis/genética , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Retinoides/química
18.
Cytometry B Clin Cytom ; 70(5): 344-54, 2006 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-16739216

RESUMEN

BACKGROUND: Applications of fluorescence-activated cell sorting (FACS) are ideally performed under aseptic conditions so that isolated cells can be successfully cultured, transplanted, or processed for the isolation of protein and nucleic acids. However, modern "off-the shelf" flow cytometers are suboptimally designed for these purposes because nonsterile instrument hardware components directly contact sample-harboring fluids, compromising their sterility. METHODS: We have described the design and modular modification of a cytometer with a sterile and disposable FACS fluid handling system that meets requirements of high-speed FACS and good manufacturing practice. This system was tested for functionality and its ability to maintain a clean and sterile fluid environment. RESULTS: Our data have shown that this new fluidic subsystem completely replicated the intended function of the manufacturer's standard fluid handling system, and isolates the fluid from contaminants such as bacteria and fungus, endotoxins, mycoplasma, and helicobacter. CONCLUSIONS: FACS has emerged as a powerful tool used to study and manipulate stem cells. However, if stem cell discoveries are to be fully utilized in clinical transplant medicine, aseptic instrument configurations must be developed. For this purpose, we have designed a disposable sterile fluid handling system.


Asunto(s)
Contaminación de Equipos , Citometría de Flujo/instrumentación , Citometría de Flujo/métodos , Endotoxinas , Helicobacter , Mycoplasma , Reacción en Cadena de la Polimerasa
19.
Cell Stem Cell ; 18(2): 214-28, 2016 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-26627594

RESUMEN

The mammalian imprinted Dlk1-Gtl2 locus produces multiple non-coding RNAs (ncRNAs) from the maternally inherited allele, including the largest miRNA cluster in the mammalian genome. This locus has characterized functions in some types of stem cell, but its role in hematopoietic stem cells (HSCs) is unknown. Here, we show that the Dlk1-Gtl2 locus plays a critical role in preserving long-term repopulating HSCs (LT-HSCs). Through transcriptome profiling in 17 hematopoietic cell types, we found that ncRNAs expressed from the Dlk1-Gtl2 locus are predominantly enriched in fetal liver HSCs and the adult LT-HSC population and sustain long-term HSC functionality. Mechanistically, the miRNA mega-cluster within the Dlk1-Gtl2 locus suppresses the entire PI3K-mTOR pathway. This regulation in turn inhibits mitochondrial biogenesis and metabolic activity and protects LT-HSCs from excessive reactive oxygen species (ROS) production. Our data therefore show that the imprinted Dlk1-Gtl2 locus preserves LT-HSC function by restricting mitochondrial metabolism.


Asunto(s)
Sitios Genéticos , Células Madre Hematopoyéticas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Mitocondrias/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , ARN Largo no Codificante/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Acetilcisteína/farmacología , Animales , Antígenos CD/metabolismo , Proteínas de Unión al Calcio , Feto/metabolismo , Impresión Genómica , Células HEK293 , Humanos , Hígado/citología , Hígado/embriología , Ratones Endogámicos C57BL , MicroARNs/genética , MicroARNs/metabolismo , Mitocondrias/ultraestructura , Mutación/genética , Biogénesis de Organelos , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Sirolimus/farmacología
20.
BMC Biol ; 2: 6, 2004 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-15132741

RESUMEN

BACKGROUND: Runx transcription factors are important regulators of metazoan development. The sea urchin Runx gene SpRunt was previously identified as a trans-activator of the CyIIIa actin gene, a differentiation marker of larval aboral ectoderm. Here we extend the functional analysis of SpRunt, using morpholino antisense oligonucleotides (morpholinos) to interfere with SpRunt expression in the embryo. RESULTS: The developmental effects of four different SpRunt-specific morpholinos were evaluated. The two morpholinos most effective at knocking down SpRunt produce an identical mitotic catastrophe phenotype at late cleavage stage that is an artifact of coincidental mis-targeting to histone mRNA, providing a cautionary example of the insufficiency of two different morpholinos as a control for specificity. The other two morpholinos produce gastrula stage proliferation and differentiation defects that are rescued by exogenous SpRunt mRNA. The expression of 22 genes involved in cell proliferation and differentiation was analyzed in the latter embryos by quantitative polymerase chain reaction. Knockdown of SpRunt was found to perturb the expression of differentiation markers in all of the major tissue territories as well as the expression of cell cycle control genes, including cyclin B and cyclin D. CONCLUSIONS: SpRunt is essential for embryonic development, and is required globally to coordinate cell proliferation and differentiation.


Asunto(s)
Embrión no Mamífero/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Oligonucleótidos Antisentido/farmacología , Erizos de Mar/embriología , Erizos de Mar/genética , Factores de Transcripción/genética , Animales , Embrión no Mamífero/embriología , Marcación de Gen , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/metabolismo , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA