Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Genes Dev ; 31(1): 12-17, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-28115466

RESUMEN

Global DNA demethylation is a hallmark of embryonic epigenetic reprogramming. However, embryos engage noncanonical DNA methylation maintenance mechanisms to ensure inheritance of exceptional epigenetic germline features to the soma. Besides the paradigmatic genomic imprints, these exceptions remain ill-defined, and the mechanisms ensuring demethylation resistance in the light of global reprogramming remain poorly understood. Here we show that the Y-linked gene Rbmy1a1 is highly methylated in mature sperm and resists DNA demethylation post-fertilization. Aberrant hypomethylation of the Rbmy1a1 promoter results in its ectopic activation, causing male-specific peri-implantation lethality. Rbmy1a1 is a novel target of the TRIM28 complex, which is required to protect its repressive epigenetic state during embryonic epigenetic reprogramming.


Asunto(s)
Metilación de ADN/genética , Desarrollo Embrionario/genética , Epigénesis Genética/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas Represoras/genética , Animales , Células Cultivadas , Reprogramación Celular/genética , Implantación del Embrión/genética , Embrión de Mamíferos , Femenino , Regulación del Desarrollo de la Expresión Génica , Impresión Genómica/genética , Masculino , Mutación , Regiones Promotoras Genéticas/genética , Proteínas de Unión al ARN/genética , Espermatozoides/metabolismo , Proteína 28 que Contiene Motivos Tripartito
2.
Stem Cells ; 40(12): 1078-1093, 2022 12 31.
Artículo en Inglés | MEDLINE | ID: mdl-36124999

RESUMEN

Myeloid differentiation blockage at immature and self-renewing stages is a common hallmark across all subtypes of acute myeloid leukemia (AML), despite their genetic heterogeneity. Metabolic state is an important regulator of hematopoietic stem cell (HSC) self-renewal and lineage-specific differentiation as well as several aggressive cancers. However, how O-GlcNAcylation, a nutrient-sensitive posttranslational modification of proteins, contributes to both normal myelopoiesis and AML pathogenesis remains largely unknown. Using small molecule inhibitors and the CRISPR/Cas9 system, we reveal for the first time that inhibition of either OGA or OGT, which subsequently caused an increase or decrease in cellular O-GlcNAcylation, inhibits the self-renewal and maintenance of CD34+ hematopoietic stem/progenitor cells (HSPCs) and leukemic stem/progenitor cells and drives normal and malignant myeloid differentiation. We further unveiled the distinct roles of OGA and OGT inhibition in lineage-specific differentiation. While OGT inhibition induces macrophage differentiation, OGA inhibition promotes the differentiation of both CD34+ HSPCs and AML cells into dendritic cells (DCs), in agreement with an upregulation of a multitude of genes involved in DC development and function and their ability to induce T-cell proliferation, via STAT3/5 signaling. Our novel findings provide significant basic knowledge that could be important in understanding AML pathogenesis and overcoming differentiation blockage-agnostic to the genetic background of AML. Additionally, the parallel findings in normal HSPCs may lay the groundwork for future cellular therapy as a means to improve the ex vivo differentiation of normal DCs and macrophages.


Asunto(s)
Autorrenovación de las Células , Leucemia Mieloide Aguda , Humanos , Antígenos CD34/metabolismo , Diferenciación Celular , Células Madre Hematopoyéticas/metabolismo , Leucemia Mieloide Aguda/genética , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT5
3.
Int J Mol Sci ; 23(23)2022 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-36498961

RESUMEN

Yes-associated protein (YAP) and WW domain-containing transcription regulator protein 1 (WWTR1, also known as TAZ) are transcriptional coactivators in the Hippo signaling pathway. Both are well-known regulators of cell proliferation and organ size control, and they have significant roles in promoting cell proliferation and differentiation. The roles of YAP and TAZ in stem cell pluripotency and differentiation have been extensively studied. However, the upstream mediators of YAP and TAZ are not well understood. Recently, a novel role of YAP in mechanosensing and mechanotransduction has been reported. The present review updates information on the regulation of YAP by mechanical cues such as extracellular matrix stiffness, fluid shear stress, and actin cytoskeleton tension in stem cell behaviors and differentiation. The review explores mesenchymal stem cell fate decisions, pluripotent stem cells (PSCs), self-renewal, pluripotency, and differentiation to blood products. Understanding how cells sense their microenvironment or niche and mimic those microenvironments in vitro could improve the efficiency of producing stem cell products and the efficacy of the products.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Células Madre Pluripotentes , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Mecanotransducción Celular/fisiología , Proteínas Señalizadoras YAP , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Células Madre Pluripotentes/metabolismo
4.
Genes Dev ; 27(13): 1441-6, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23824537

RESUMEN

Cellular localization of the Yes-associated protein (YAP) is dependent on large tumor suppressor (LATS) kinase activity and initiates lineage specification in the preimplantation embryo. We temporally reduced LATS activity to disrupt this early event, allowing its reactivation at later stages. This interference resulted in an irreversible lineage misspecification and aberrant polarization of the inner cell mass (ICM). Complementation experiments revealed that neither epiblast nor primitive endoderm can be established from these ICMs. We therefore conclude that precisely timed YAP localization in early morulae is essential to prevent trophectoderm marker expression in, and lineage specification of, the ICM.


Asunto(s)
Masa Celular Interna del Blastocisto/citología , Blastocisto/citología , Diferenciación Celular , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteínas de Ciclo Celular , Linaje de la Célula , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Prueba de Complementación Genética , Vía de Señalización Hippo , Ratones , Fosfoproteínas/metabolismo , Transducción de Señal , Factores de Tiempo , Proteínas Señalizadoras YAP
5.
Genes Dev ; 27(12): 1378-90, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23788624

RESUMEN

Although it is known that OCT4-NANOG are required for maintenance of pluripotent cells in vitro, the upstream signals that regulate this circuit during early development in vivo have not been identified. Here we demonstrate, for the first time, signal transducers and activators of transcription 3 (STAT3)-dependent regulation of the OCT4-NANOG circuitry necessary to maintain the pluripotent inner cell mass (ICM), the source of in vitro-derived embryonic stem cells (ESCs). We show that STAT3 is highly expressed in mouse oocytes and becomes phosphorylated and translocates to the nucleus in the four-cell and later stage embryos. Using leukemia inhibitory factor (Lif)-null embryos, we found that STAT3 phosphorylation is dependent on LIF in four-cell stage embryos. In blastocysts, interleukin 6 (IL-6) acts in an autocrine fashion to ensure STAT3 phosphorylation, mediated by janus kinase 1 (JAK1), a LIF- and IL-6-dependent kinase. Using genetically engineered mouse strains to eliminate Stat3 in oocytes and embryos, we firmly establish that STAT3 is essential for maintenance of ICM lineages but not for ICM and trophectoderm formation. Indeed, STAT3 directly binds to the Oct4 and Nanog distal enhancers, modulating their expression to maintain pluripotency of mouse embryonic and induced pluripotent stem cells. These results provide a novel genetic model of cell fate determination operating through STAT3 in the preimplantation embryo and pluripotent stem cells in vivo.


Asunto(s)
Masa Celular Interna del Blastocisto , Linaje de la Célula , Células Madre Embrionarias/fisiología , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio , Factor 3 de Transcripción de Unión a Octámeros , Factor de Transcripción STAT3 , Animales , Masa Celular Interna del Blastocisto/citología , Masa Celular Interna del Blastocisto/metabolismo , Células Cultivadas , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Femenino , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Janus Quinasa 1/genética , Janus Quinasa 1/metabolismo , Factor Inhibidor de Leucemia/genética , Factor Inhibidor de Leucemia/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Fosforilación , Células Madre Pluripotentes/fisiología , Unión Proteica , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo
6.
Development ; 143(11): 1993-9, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27246714

RESUMEN

ß-catenin (CTNNB1) is integral to cell adhesion and to the canonical Wnt signaling pathway. The effects of maternal and zygotic CTNNB1 on embryogenesis have each been separately assessed, whereas the effect of its total absence has not. As the 'traditional' conditional Ctnnb1 knockout alleles give rise to truncated CTNNB1 fragments, we designed a new knockout allele incapable of CTNNB1 production. Mouse embryos lacking intact maternal/zygotic CTNNB1 from two knockout strains were examined in detail. Preimplantation embryos are formed, yet abnormalities in their size and shape were found throughout pre- and early postimplantation development. In the absence of the zona pellucida, embryos lacking CTNNB1 undergo fission and these separated blastomeres can become small trophoblastic vesicles, which in turn induce decidual reactions. Comparing the severity of this defective adhesion phenotype in embryos bearing the null allele with those carrying the 'traditional' knockout allele suggests a hypomorphic effect of the truncated CTNNB1 protein fragment, an important observation with possible impact on previous and future studies.


Asunto(s)
Desarrollo Embrionario , beta Catenina/metabolismo , Alelos , Animales , Blastocisto/citología , Blastocisto/metabolismo , Adhesión Celular , Eliminación de Gen , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Mutantes/metabolismo , Cigoto/metabolismo , beta Catenina/deficiencia
7.
Development ; 140(18): 3819-25, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23924633

RESUMEN

Mobilization of endogenous retrotransposons can destabilize the genome, an imminent danger during epigenetic reprogramming of cells in the germline. The P-element-induced wimpy testis (PIWI)-interacting RNA (piRNA) pathway is known to silence retrotransposons in the mouse testes. Several piRNA pathway components localize to the unique, germline structure known as the nuage. In this study, we surveyed mouse ovaries and found, for the first time, transient appearance of nuage-like structures in oocytes of primordial follicles. Mouse vasa homolog (MVH), Piwi-like 2 (PIWIL2/MILI) and tudor domain-containing 9 (TDRD9) are present in these structures, whereas aggregates of germ cell protein with ankyrin repeats, sterile alpha motif and leucine zipper (GASZ) localize separately in the cytoplasm. Retrotransposons are silenced in primordial ovarian follicles, and de-repressed upon reduction of piRNA expression in Mvh, Mili or Gasz mutants. However, these null-mutant females, unlike their male counterparts, are fertile, uncoupling retrotransposon activation from sterility.


Asunto(s)
Estructuras Celulares/metabolismo , Silenciador del Gen , Folículo Ovárico/metabolismo , Retroelementos/genética , Animales , Estructuras Celulares/ultraestructura , Femenino , Regulación de la Expresión Génica , Células Germinativas/metabolismo , Infertilidad Femenina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación/genética , Oogénesis , Folículo Ovárico/ultraestructura , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo
8.
Ann Hematol ; 95(10): 1617-25, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27465155

RESUMEN

Paroxysmal nocturnal hemoglobinuria (PNH) is an acquired hemolytic anemia caused by lack of CD55 and CD59 on blood cell membrane leading to increased sensitivity of blood cells to complement. Hematopoietic stem cell transplantation (HSCT) is the only curative therapy for PNH, however, lack of HLA-matched donors and post-transplant complications are major concerns. Induced pluripotent stem cells (iPSCs) derived from patients are an attractive source for generating autologous HSCs to avoid adverse effects resulting from allogeneic HSCT. The disease involves only HSCs and their progeny; therefore, other tissues are not affected by the mutation and may be used to produce disease-free autologous HSCs. This study aimed to derive PNH patient-specific iPSCs from human dermal fibroblasts (HDFs), characterize and differentiate to hematopoietic cells using a feeder-free protocol. Analysis of CD55 and CD59 expression was performed before and after reprogramming, and hematopoietic differentiation. Patients' dermal fibroblasts expressed CD55 and CD59 at normal levels and the normal expression remained after reprogramming. The iPSCs derived from PNH patients had typical pluripotent properties and differentiation capacities with normal karyotype. After hematopoietic differentiation, the differentiated cells expressed early hematopoietic markers (CD34 and CD43) with normal CD59 expression. The iPSCs derived from HDFs of PNH patients have normal levels of CD55 and CD59 expression and hold promise as a potential source of HSCs for autologous transplantation to cure PNH patients.


Asunto(s)
Técnicas de Reprogramación Celular , Trasplante de Células Madre Hematopoyéticas , Hemoglobinuria Paroxística/terapia , Células Madre Pluripotentes Inducidas/citología , Animales , Antígenos CD34/biosíntesis , Antígenos CD55/biosíntesis , Antígenos CD59/biosíntesis , Cuerpos Embrioides , Femenino , Fibroblastos/citología , Historia del Siglo XVI , Historia del Siglo XVII , Humanos , Células Madre Pluripotentes Inducidas/trasplante , Leucosialina/biosíntesis , Ratones Endogámicos BALB C , Ratones Desnudos , Piel/citología , Teratoma/patología , Trasplante Autólogo
9.
Development ; 139(20): 3722-31, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22991438

RESUMEN

The inside-outside model has been invoked to explain cell-fate specification of the pre-implantation mammalian embryo. Here, we investigate whether cell-cell interaction can influence the fate specification of embryonic blastomeres by sequentially separating the blastomeres in two-cell stage mouse embryos and continuing separation after each cell division throughout pre-implantation development. This procedure eliminates information provided by cell-cell interaction and cell positioning. Gene expression profiles, polarity protein localization and functional tests of these separated blastomeres reveal that cell interactions, through cell position, influence the fate of the blastomere. Blastomeres, in the absence of cell contact and inner-outer positional information, have a unique pattern of gene expression that is characteristic of neither inner cell mass nor trophectoderm, but overall they have a tendency towards a 'trophectoderm-like' gene expression pattern and preferentially contribute to the trophectoderm lineage.


Asunto(s)
Blastómeros/citología , Blastómeros/fisiología , Comunicación Celular , Embrión de Mamíferos/citología , Desarrollo Embrionario , Animales , Diferenciación Celular , Linaje de la Célula , Embrión de Mamíferos/metabolismo , Femenino , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Ratones , Embarazo
10.
Biol Reprod ; 92(6): 143, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25947059

RESUMEN

In preimplantation mouse embryos, the first lineage differentiation takes place in the 8- to 16-cell-stage embryo and results in formation of the trophectoderm (TE) and inner cell mass (ICM), which will give rise to the trophoblast of the placenta and the embryo proper, respectively. Although, it is widely accepted that positioning of a cell within the embryo influences lineage differentiation, the mechanism underlying differential lineage differentiation and how it involves cell position are largely unknown. Interestingly, novel cues from the Hippo pathway have been recently demonstrated in the preimplantation mouse embryo. Unlike the mechanisms reported from epithelium-cultured cells, the Hippo pathway was found to be responsible for translating positional information to lineage specification through a position-sensing mechanism. Disruption of Hippo pathway-component genes in early embryos results in failure of lineage specification and failure of postimplantation development. In this review, we discuss the unique role of the Hippo signaling pathway in early embryo development and its role in lineage specification. Understanding the activity and regulation of the Hippo pathway may offer new insights into other areas of developmental biology that evolve from understanding of this cell-fate specification in the early embryonic cell.


Asunto(s)
Blastocisto/metabolismo , Implantación del Embrión/fisiología , Embrión de Mamíferos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/fisiología , Animales , Femenino , Regulación del Desarrollo de la Expresión Génica , Vía de Señalización Hippo , Ratones , Embarazo
11.
Sci Rep ; 14(1): 3993, 2024 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-38368446

RESUMEN

Chronic myelogenous leukemia (CML) is a clonal hematologic malignancy of the myeloid lineage caused by the oncogenic BCR/ABL fusion protein that promotes CML cell proliferation and protects them against drug-induced apoptosis. In this study, we determine LATS1 and LATS2 expression in CML cells derived from patients who are resistant to imatinib (IM) treatment. Significant upregulation of LATS1 and LATS2 was found in these CML patients compared to healthy donors. To further explore whether the expression of LATS1/2 contributes to the IM-resistant phenotype, IM-resistant CML cell lines generated by culturing CML-derived erythroblastic K562 cells in increasing concentrations of IM were used as in vitro models. Up-regulation of LATS1 and LATS2 was observed in IM-resistant K562 cells. Reduction of LATS using either Lats-IN-1 (TRULI), a specific LATS inhibitor, or shRNA targeting LATS1/2 significantly reduced clonogenicity, increased apoptosis and induced differentiation of K562 cells to late-stage erythroid cells. Furthermore, depletion of LATS1 and LATS2 also increased the sensitivity of K562 cells to IM. Taken together, our results suggest that LATS could be one of the key factors contributing to the rapid proliferation, reduced apoptosis, and IM resistance of CML cells. Targeting LATS could be a promising treatment to enhance the therapeutic effect of a conventional BCR/ABL tyrosine kinase inhibitor such as IM.


Asunto(s)
Resistencia a Antineoplásicos , Leucemia Mielógena Crónica BCR-ABL Positiva , Humanos , Mesilato de Imatinib/farmacología , Mesilato de Imatinib/uso terapéutico , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Proteínas de Fusión bcr-abl/genética , Proteínas Serina-Treonina Quinasas , Células K562 , Apoptosis , Proteínas Supresoras de Tumor
12.
Stem Cell Res ; 78: 103466, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38852425

RESUMEN

The Rh-negative type O blood group (O Rh-) is considered a universal donor for emergency blood transfusions. Due to the constant shortage of this rare blood group, the production of blood cells from iPSCs derived from the O Rh- donor could potentially serve as a limitless blood source for transfusions. In this report, we establish a MUSIi017-A iPSC line from peripheral blood mononuclear cells of a healthy donor with the O Rh- blood group. The established iPSC line exhibited a normal karyotype, showed identical STR compared to donor peripheral blood mononuclear cells, and could differentiate to all three germ layers.


Asunto(s)
Células Madre Pluripotentes Inducidas , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Línea Celular , Sistema del Grupo Sanguíneo ABO , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/metabolismo , Diferenciación Celular , Donantes de Sangre
13.
Front Cell Dev Biol ; 12: 1361943, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38752196

RESUMEN

Hematopoiesis continues throughout life to produce all types of blood cells from hematopoietic stem cells (HSCs). Metabolic state is a known regulator of HSC self-renewal and differentiation, but whether and how metabolic sensor O-GlcNAcylation, which can be modulated via an inhibition of its cycling enzymes O-GlcNAcase (OGA) and O-GlcNAc transferase (OGT), contributes to hematopoiesis remains largely unknown. Herein, isogenic, single-cell clones of OGA-depleted (OGAi) and OGT-depleted (OGTi) human induced pluripotent stem cells (hiPSCs) were successfully generated from the master hiPSC line MUSIi012-A, which were reprogrammed from CD34+ hematopoietic stem/progenitor cells (HSPCs) containing epigenetic memory. The established OGAi and OGTi hiPSCs exhibiting an increase or decrease in cellular O-GlcNAcylation concomitant with their loss of OGA and OGT, respectively, appeared normal in phenotype and karyotype, and retained pluripotency, although they may favor differentiation toward certain germ lineages. Upon hematopoietic differentiation through mesoderm induction and endothelial-to-hematopoietic transition, we found that OGA inhibition accelerates hiPSC commitment toward HSPCs and that disruption of O-GlcNAc homeostasis affects their commitment toward erythroid lineage. The differentiated HSPCs from all groups were capable of giving rise to all hematopoietic progenitors, thus confirming their functional characteristics. Altogether, the established single-cell clones of OGTi and OGAi hiPSCs represent a valuable platform for further dissecting the roles of O-GlcNAcylation in blood cell development at various stages and lineages of blood cells. The incomplete knockout of OGA and OGT in these hiPSCs makes them susceptible to additional manipulation, i.e., by small molecules, allowing the molecular dynamics studies of O-GlcNAcylation.

14.
Sci Rep ; 14(1): 15732, 2024 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-38977826

RESUMEN

YAP plays a vital role in controlling growth and differentiation in various cell lineages. Although the expression of YAP in mice testicular and spermatogenic cells suggests its role in mammalian spermatogenesis, the role of YAP in the development of human male germ cells has not yet been determined. Using an in vitro model and a gene editing approach, we generated human spermatogonia stem cell-like cells (hSSLCs) from human embryonic stem cells (hESCs) and investigated the role of YAP in human spermatogenesis. The results showed that reducing YAP expression during the early stage of spermatogenic differentiation increased the number of PLZF+ hSSLCs and haploid spermatid-like cells. We also demonstrated that the up-regulation of YAP is essential for maintaining spermatogenic cell survival during the later stages of spermatogenic differentiation. The expression of YAP that deviates from this pattern results in a lower number of hSSLCs and an increased level of spermatogenic cell death. Taken together, our result demonstrates that the dynamic expression pattern of YAP is essential for human spermatogenesis. Modulating the level of YAP during human spermatogenesis could improve the production yield of male germ cells derived from hESCs, which could provide the optimization method for in vitro gametogenesis and gain insight into the application in the treatment of male infertility.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Diferenciación Celular , Células Madre Embrionarias Humanas , Espermatogénesis , Factores de Transcripción , Proteínas Señalizadoras YAP , Masculino , Humanos , Células Madre Embrionarias Humanas/metabolismo , Células Madre Embrionarias Humanas/citología , Proteínas Señalizadoras YAP/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Espermatogonias/metabolismo , Espermatogonias/citología , Proteína de la Leucemia Promielocítica con Dedos de Zinc/metabolismo , Proteína de la Leucemia Promielocítica con Dedos de Zinc/genética
15.
Stem Cell Res ; 66: 103014, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36603353

RESUMEN

MUSIi016-A, a human induced pluripotent stem cell (iPSC), generated from peripheral blood mononuclear cells of a healthy blood group O Rh positive donor was reprogrammed using Sendai viral vectors containing Yamanaka's factors. MUSIi016-A iPSC showed pluripotent stem cell characteristics, highly expressed pluripotent markers, and a capacity to differentiate into all three embryonic cell lineages. This iPSC can be used as a model for the generation of blood cells in vitro.


Asunto(s)
Células Madre Pluripotentes Inducidas , Células Madre Pluripotentes , Humanos , Leucocitos Mononucleares , Linaje de la Célula , Diferenciación Celular
16.
Gels ; 9(3)2023 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-36975637

RESUMEN

Levan is a biopolymer composed of fructose chains covalently linked by ß-2,6 glycosidic linkages. This polymer self-assembles into a nanoparticle of uniform size, making it useful for a wide range of applications. Also, levan exhibits various biological activities such as antioxidants, anti-inflammatory, and anti-tumor, that make this polymer very attractive for biomedical application. In this study, levan synthesized from Erwinia tasmaniensis was chemically modified by glycidyl trimethylammonium chloride (GTMAC) to produce cationized nanolevan (QA-levan). The structure of the obtained GTMAC-modified levan was determined by FT-IR, 1H-NMR and elemental (CHN) analyzer. The size of the nanoparticle was calculated using the dynamic light scattering method (DLS). The formation of DNA/QA-levan polyplex was then investigated by gel electrophoresis. The modified levan was able to increase the solubility of quercetin and curcumin by 11-folds and 205-folds, respectively, compared to free compounds. Cytotoxicity of levan and QA-levan was also investigated in HEK293 cells. This finding suggests that GTMAC-modified levan should have a potential application for drug and nucleic acid delivery.

17.
Stem Cell Res Ther ; 14(1): 279, 2023 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-37775798

RESUMEN

BACKGROUND: In vitro production of hematopoietic stem/progenitor cells (HSPCs) from human-induced pluripotent stem cells (hiPSCs) provides opportunities for fundamental research, disease modeling, and large-scale production of HLA-matched HSPCs for therapeutic applications. However, a comprehensive understanding of the signaling mechanisms that regulate human hematopoiesis is needed to develop a more effective procedure for deriving HSPCs from hiPSCs. METHODS: In this study, we investigate the role of YAP during the hematopoietic differentiation of hiPSCs to HSPCs and erythrocytes using the isogenic YAP-overexpressing (YAP-S5A) and YAP-depleting (YAP-KD) hiPSCs to eliminate the effects of a genetic background variation. RESULTS: Although YAP is dispensable for maintaining the self-renewal and pluripotency of these hiPSCs, it affects the early cell-fate determination and hematopoietic differentiation of hiPSCs. Depleting YAP enhances the derivation efficiency of HSPCs from hiPSCs by inducing the mesodermal lineage commitment, promoting hematopoietic differentiation, and preventing the differentiation toward endothelial lineage. On the contrary, the overexpression of YAP reduced HSPCs yield by inducing the endodermal lineage commitment, suppressing hematopoietic differentiation, and promoting the differentiation toward endothelial lineage. CONCLUSIONS: Expression of YAP is crucial for the differentiation of hiPSC-derived HSPCs toward mature erythrocytes. We believe that by manipulating YAP activity using small molecules, the efficiency of the large-scale in vitro production system for generating hematopoietic stem/progenitor cells for future therapeutic use could be improved.


Asunto(s)
Células Madre Pluripotentes Inducidas , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Linaje de la Célula/genética , Diferenciación Celular/genética , Células Madre Hematopoyéticas/metabolismo , Hematopoyesis
18.
Stem Cell Res ; 67: 103035, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36753833

RESUMEN

Runt-Related Transcription Factor 1c (RUNX1c) plays an important role in regulating the development of hematopoietic stem cells (HSC). Using CRISPR/Cas9 gene editing technology, we established a RUNX1c-eGFP reporter cell line from the MUSIi012-A cell line. The MUSIi012-A-4 cell line has normal stem cell morphology and karyotype, expresses pluripotency markers, and can be differentiated into all three germ layers in vitro and in vivo. This cell line serves as a valuable model to observe the expression of RUNX1c via eGFP tracking during human hematopoietic development.


Asunto(s)
Células Madre Pluripotentes Inducidas , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Sistemas CRISPR-Cas , Línea Celular , Edición Génica , Diferenciación Celular
19.
Stem Cell Res ; 66: 102990, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36528979

RESUMEN

Yes-associated protein (YAP), an important effector protein of the Hippo signaling pathway, acts as a molecular switch in controlling cell proliferation and apoptosis. In this study, a YAP-targeted isogenic sub-clone of the MUSIe002-A was generated, designated as MUSIe002-A-1. The MUSIe002-1 cell line had normal pluripotent stem cell characteristics and karyotype. Its ability to differentiate into three germ layers was confirmed. As reduction of YAP does not disturb the pluripotency of hESCs, this cell line serves as a valuable model to extrapolate the functional role of YAP in stem cell biology and its applications.


Asunto(s)
Células Madre Embrionarias Humanas , Factores de Transcripción , Humanos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Transducción de Señal/fisiología , Células Madre Embrionarias Humanas/metabolismo , Sistemas CRISPR-Cas/genética , Proteínas Señalizadoras YAP , Línea Celular
20.
Biomedicines ; 10(1)2022 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-35052826

RESUMEN

The hippo signaling pathway plays an essential role in controlling organ size and balancing tissue homeostasis. Its two main effectors, yes-associated protein (YAP) and WW domain-containing transcription regulator 1, WWTR1 or TAZ, have also been shown to regulate endothelial cell functions and angiogenesis. In this study, the functions of YAP and TAZ in human endothelial progenitor cells (EPCs) were investigated by a loss-of-function study using CRISPR/Cas9-mediated gene knockdown (KD). Depletion of either YAP or TAZ reduced EPC survival and impaired many of their critical functions, including migration, invasion, vessel-formation, and expression of pro-angiogenic genes. Notably, TAZ-KD EPCs exhibited more severe phenotypes in comparison to YAP-KD EPCs. Moreover, the conditioned medium derived from TAZ-KD EPCs reduced the survivability of human lung cancer cells and increased their sensitivity to chemotherapeutic agents. The overexpression of either wild-type or constitutively active TAZ rescued the impaired phenotypes of TAZ-KD EPCs and restored the expression of pro-angiogenic genes in those EPCs. In summary, we demonstrate the crucial role of Hippo signaling components, YAP and TAZ, in controlling several aspects of EPC functions that can potentially be used as a drug target to enhance EPC functions in patients.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA