Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Cell ; 172(1-2): 191-204.e10, 2018 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-29224778

RESUMEN

Hematopoietic stem cell transplantation is a potential curative therapy for malignant and nonmalignant diseases. Improving the efficiency of stem cell collection and the quality of the cells acquired can broaden the donor pool and improve patient outcomes. We developed a rapid stem cell mobilization regimen utilizing a unique CXCR2 agonist, GROß, and the CXCR4 antagonist AMD3100. A single injection of both agents resulted in stem cell mobilization peaking within 15 min that was equivalent in magnitude to a standard multi-day regimen of granulocyte colony-stimulating factor (G-CSF). Mechanistic studies determined that rapid mobilization results from synergistic signaling on neutrophils, resulting in enhanced MMP-9 release, and unexpectedly revealed genetic polymorphisms in MMP-9 that alter activity. This mobilization regimen results in preferential trafficking of stem cells that demonstrate a higher engraftment efficiency than those mobilized by G-CSF. Our studies suggest a potential new strategy for the rapid collection of an improved hematopoietic graft.


Asunto(s)
Movilización de Célula Madre Hematopoyética/métodos , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/inmunología , Adulto , Animales , Bencilaminas , Quimiocina CXCL2/farmacología , Ciclamas , Femenino , Células Madre Hematopoyéticas/efectos de los fármacos , Compuestos Heterocíclicos/farmacología , Humanos , Masculino , Metaloproteinasa 9 de la Matriz/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Endogámicos ICR , Polimorfismo Genético
2.
PLoS One ; 10(1): e0116933, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25635918

RESUMEN

Induced pluripotent stem (iPS) cells are at the forefront of research in regenerative medicine and are envisaged as a source for personalized tissue repair and cell replacement therapy. Here, we demonstrate for the first time that oligodendrocyte progenitors (OPs) can be derived from iPS cells generated using either an episomal, non-integrating plasmid approach or standard integrating retroviruses that survive and differentiate into mature oligodendrocytes after early transplantation into the injured spinal cord. The efficiency of OP differentiation in all 3 lines tested ranged from 40% to 60% of total cells, comparable to those derived from human embryonic stem cells. iPS cell lines derived using episomal vectors or retroviruses generated a similar number of early neural progenitors and glial progenitors while the episomal plasmid-derived iPS line generated more OPs expressing late markers O1 and RIP. Moreover, we discovered that iPS-derived OPs (iPS-OPs) engrafted 24 hours following a moderate contusive spinal cord injury (SCI) in rats survived for approximately two months and that more than 70% of the transplanted cells differentiated into mature oligodendrocytes that expressed myelin associated proteins. Transplanted OPs resulted in a significant increase in the number of myelinated axons in animals that received a transplantation 24 h after injury. In addition, nearly a 5-fold reduction in cavity size and reduced glial scarring was seen in iPS-treated groups compared to the control group, which was injected with heat-killed iPS-OPs. Although further investigation is needed to understand the mechanisms involved, these results provide evidence that patient-specific, iPS-derived OPs can survive for three months and improve behavioral assessment (BBB) after acute transplantation into SCI. This is significant as determining the time in which stem cells are injected after SCI may influence their survival and differentiation capacity.


Asunto(s)
Células Madre Pluripotentes Inducidas/trasplante , Células-Madre Neurales/trasplante , Traumatismos de la Médula Espinal/terapia , Animales , Axones/fisiología , Diferenciación Celular , Supervivencia Celular , Células Cultivadas , Intervención Médica Temprana , Femenino , Humanos , Actividad Motora , Vaina de Mielina/fisiología , Regeneración Nerviosa , Oligodendroglía/fisiología , Ratas Endogámicas Lew , Recuperación de la Función , Médula Espinal/patología , Médula Espinal/fisiopatología , Resultado del Tratamiento
3.
Stem Cells ; 32(2): 349-63, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23765875

RESUMEN

Reprogramming of somatic cells to induced pluripotent stem cells (iPSCs) shares much similarity to the cancer initiation process, and the molecular mechanisms underlying both processes remain to be elucidated. Here, we report that a tumor- or embryonic stem cell-specific Ras gene ERas, which encodes a constitutively active form of GTPase, and its downstream Phosphoinositide-3 kinase/Akt signaling pathway are important facilitators for the somatic reprogramming process. We found that overexpression of ERas retrovirally enhanced mouse iPSC induction while ERas knockdown repressed it. Modulation of Akt signaling by genetic or chemical means greatly impacted the reprogramming efficiency. Forced expression of a constitutively active Akt1 gene could rescue the reduced efficiency resulting from ERas knockdown, and point-mutation analyses further revealed that ERas is tightly coupled with Akt signaling to enhance reprogramming. Mechanistically, the forkhead transcription factor FoxO1 can function as a barrier to the iPSC induction, and the inactivation of FoxO1 by Akt-dependent phosphorylation largely accounts for the enhancing effect of ERas-Akt signaling on reprogramming. Collectively, these results unravel the significance of the ERas-Akt-FoxO1 signaling axis in iPSC generation, suggesting a possibly shared molecular basis for both somatic reprogramming and cancer initiation.


Asunto(s)
Células Madre Embrionarias/metabolismo , Factores de Transcripción Forkhead/genética , Proteína Oncogénica p21(ras)/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Diferenciación Celular/genética , Reprogramación Celular/genética , Proteína Forkhead Box O1 , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Neoplasias/genética , Proteínas Proto-Oncogénicas c-akt/genética , Transducción de Señal/genética
4.
Stem Cell Res ; 11(3): 1103-16, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23973800

RESUMEN

Large-scale production of human induced pluripotent stem cells (hiPSCs) by robust and economic methods has been one of the major challenges for translational realization of hiPSC technology. Here we demonstrate a scalable culture system for hiPSC expansion using the E8 chemically defined and xeno-free medium under either adherent or suspension conditions. To optimize suspension conditions guided by a computational simulation, we developed a method to efficiently expand hiPSCs as undifferentiated aggregates in spinner flasks. Serial passaging of two different hiPSC lines in the spinner flasks using the E8 medium preserved their normal karyotype and expression of undifferentiated state markers of TRA-1-60, SSEA4, OCT4, and NANOG. The hiPSCs cultured in spinner flasks for more than 10 passages not only could be remained pluripotent as indicated by in vitro and in vivo assays, but also could be efficiently induced toward mesodermal and hematopoietic differentiation. Furthermore, we established a xeno-free protocol of single-cell cryopreservation and recovery for the scalable production of hiPSCs in spinner flasks. This system is the first to enable an efficient scale-up bioprocess in completely xeno-free condition for the expansion and cryopreservation of hiPSCs with the quantity and quality compliant for clinical applications.


Asunto(s)
Técnicas de Cultivo de Célula , Medios de Cultivo/química , Células Madre Pluripotentes Inducidas/citología , Antígenos de Superficie/metabolismo , Adhesión Celular , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Proteínas de Homeodominio/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Cariotipificación , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Proteoglicanos/metabolismo , Antígenos Embrionarios Específico de Estadio/metabolismo
6.
Int J Hematol ; 95(6): 601-9, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22619021

RESUMEN

Postnatal hematopoietic stem cells (HSCs) from umbilical cord blood and adult marrow/blood have been successfully used for treating various human diseases in the past several decades. However, the availability of optimal numbers of HSCs from autologous patients or allogeneic donors with adequate match remains a great barrier to improve and extend HSC and marrow transplantation to more needing patients. In addition, the inability to expand functional human HSCs to sufficient quantity in the laboratory has hindered our research and understanding of human HSCs and hematopoiesis. Recent development in reprogramming technology has provided patient-specific pluripotent stem cells (iPSCs) as a powerful enabling tool for modeling disease and developing therapeutics. Studies have demonstrated the potential of human iPSCs, which can be expanded exponentially and amenable for genome engineering, for using in modeling both inherited and acquired blood diseases. Proof-of-principle studies have also shown the feasibility of iPSCs in gene and cell therapy. Here, we review the recent development in iPSC-based blood disease modeling, and discuss the unsolved issues and challenges in this new and promising field.


Asunto(s)
Enfermedades Hematológicas/terapia , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/trasplante , Animales , Diferenciación Celular , Modelos Animales de Enfermedad , Humanos , Células Madre Pluripotentes Inducidas/metabolismo
7.
Cell Stem Cell ; 10(3): 337-44, 2012 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-22385660

RESUMEN

The utility of induced pluripotent stem cells (iPSCs) as models to study diseases and as sources for cell therapy depends on the integrity of their genomes. Despite recent publications of DNA sequence variations in the iPSCs, the true scope of such changes for the entire genome is not clear. Here we report the whole-genome sequencing of three human iPSC lines derived from two cell types of an adult donor by episomal vectors. The vector sequence was undetectable in the deeply sequenced iPSC lines. We identified 1,058-1,808 heterozygous single-nucleotide variants (SNVs), but no copy-number variants, in each iPSC line. Six to twelve of these SNVs were within coding regions in each iPSC line, but ~50% of them are synonymous changes and the remaining are not selectively enriched for known genes associated with cancers. Our data thus suggest that episome-mediated reprogramming is not inherently mutagenic during integration-free iPSC induction.


Asunto(s)
Variación Genética , Células Madre Pluripotentes Inducidas , Plásmidos/genética , Línea Celular , Reprogramación Celular/genética , Vectores Genéticos/genética , Humanos , Leucocitos Mononucleares , Análisis por Micromatrices , Análisis de Secuencia de ADN
8.
Stem Cells Dev ; 21(12): 2298-311, 2012 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-22225458

RESUMEN

Derivation of pluripotent stem cells (iPSCs) induced from somatic cell types and the subsequent genetic modifications of disease-specific or patient-specific iPSCs are crucial steps in their applications for disease modeling as well as future cell and gene therapies. Conventional procedures of these processes require co-culture with primary mouse embryonic fibroblasts (MEFs) to support self-renewal and clonal growth of human iPSCs as well as embryonic stem cells (ESCs). However, the variability of MEF quality affects the efficiencies of all these steps. Furthermore, animal sourced feeders may hinder the clinical applications of human stem cells. In order to overcome these hurdles, we established immortalized human feeder cell lines by stably expressing human telomerase reverse transcriptase, Wnt3a, and drug resistance genes in adult mesenchymal stem cells. Here, we show that these immortalized human feeders support efficient derivation of virus-free, integration-free human iPSCs and long-term expansion of human iPSCs and ESCs. Moreover, the drug-resistance feature of these feeders also supports nonviral gene transfer and expression at a high efficiency, mediated by piggyBac DNA transposition. Importantly, these human feeders exhibit superior ability over MEFs in supporting homologous recombination-mediated gene targeting in human iPSCs, allowing us to efficiently target a transgene into the AAVS1 safe harbor locus in recently derived integration-free iPSCs. Our results have great implications in disease modeling and translational applications of human iPSCs, as these engineered human cell lines provide a more efficient tool for genetic modifications and a safer alternative for supporting self-renewal of human iPSCs and ESCs.


Asunto(s)
Técnicas de Cultivo de Célula , Células Nutrientes/fisiología , Ingeniería Genética , Células Madre Pluripotentes Inducidas/fisiología , Fosfatasa Alcalina/metabolismo , Animales , Antígenos de Diferenciación/metabolismo , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/fisiología , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Células Madre Embrionarias/metabolismo , Células Nutrientes/metabolismo , Femenino , Sitios Genéticos , Recombinación Homóloga , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/trasplante , Cariotipo , Masculino , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/fisiología , Ratones , Neoplasias Experimentales/patología , Telomerasa/genética , Teratoma/patología , Transducción Genética , Proteína Wnt3A/biosíntesis , Proteína Wnt3A/genética
9.
Blood ; 117(21): 5561-72, 2011 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-21411759

RESUMEN

We have developed induced pluripotent stem cells (iPSCs) from a patient with X-linked chronic granulomatous disease (X-CGD), a defect of neutrophil microbicidal reactive oxygen species (ROS) generation resulting from gp91(phox) deficiency. We demonstrated that mature neutrophils differentiated from X-CGD iPSCs lack ROS production, reproducing the pathognomonic CGD cellular phenotype. Targeted gene transfer into iPSCs, with subsequent selection and full characterization to ensure no off-target changes, holds promise for correction of monogenic diseases without the insertional mutagenesis caused by multisite integration of viral or plasmid vectors. Zinc finger nuclease-mediated gene targeting of a single-copy gp91(phox) therapeutic minigene into one allele of the "safe harbor" AAVS1 locus in X-CGD iPSCs without off-target inserts resulted in sustained expression of gp91(phox) and substantially restored neutrophil ROS production. Our findings demonstrate how precise gene targeting may be applied to correction of X-CGD using zinc finger nuclease and patient iPSCs.


Asunto(s)
Enfermedad Granulomatosa Crónica/terapia , Células Madre Pluripotentes Inducidas/metabolismo , Glicoproteínas de Membrana/fisiología , NADPH Oxidasas/deficiencia , Neutrófilos/enzimología , Adulto , Animales , Southern Blotting , Western Blotting , Médula Ósea/metabolismo , Diferenciación Celular , Dermatoglifia del ADN , Dependovirus/genética , Citometría de Flujo , Enfermedad Granulomatosa Crónica/genética , Enfermedad Granulomatosa Crónica/patología , Humanos , Cariotipificación , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones Desnudos , NADPH Oxidasa 2 , NADPH Oxidasas/genética , NADPH Oxidasas/fisiología , Fagocitosis , ARN Mensajero/genética , Especies Reactivas de Oxígeno/metabolismo , Recombinación Genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Dedos de Zinc
10.
Cell Res ; 21(3): 518-29, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21243013

RESUMEN

To identify accessible and permissive human cell types for efficient derivation of induced pluripotent stem cells (iPSCs), we investigated epigenetic and gene expression signatures of multiple postnatal cell types such as fibroblasts and blood cells. Our analysis suggested that newborn cord blood (CB) and adult peripheral blood (PB) mononuclear cells (MNCs) display unique signatures that are closer to iPSCs and human embryonic stem cells (ESCs) than age-matched fibroblasts to iPSCs/ESCs, thus making blood MNCs an attractive cell choice for the generation of integration-free iPSCs. Using an improved EBNA1/OriP plasmid expressing 5 reprogramming factors, we demonstrated highly efficient reprogramming of briefly cultured blood MNCs. Within 14 days of one-time transfection by one plasmid, up to 1000 iPSC-like colonies per 2 million transfected CB MNCs were generated. The efficiency of deriving iPSCs from adult PB MNCs was approximately 50-fold lower, but could be enhanced by inclusion of a second EBNA1/OriP plasmid for transient expression of additional genes such as SV40 T antigen. The duration of obtaining bona fide iPSC colonies from adult PB MNCs was reduced to half (∼14 days) as compared to adult fibroblastic cells (28-30 days). More than 9 human iPSC lines derived from PB or CB blood cells are extensively characterized, including those from PB MNCs of an adult patient with sickle cell disease. They lack V(D)J DNA rearrangements and vector DNA after expansion for 10-12 passages. This facile method of generating integration-free human iPSCs from blood MNCs will accelerate their use in both research and future clinical applications.


Asunto(s)
Células Sanguíneas/citología , Epigénesis Genética , Perfilación de la Expresión Génica , Células Madre Pluripotentes Inducidas/metabolismo , Plásmidos/metabolismo , Antígenos CD34/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Reprogramación Celular , Sangre Fetal/citología , Fibroblastos/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Plásmidos/genética , Proteínas de Unión al ARN
11.
Methods Mol Biol ; 636: 191-205, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20336524

RESUMEN

This chapter describes the methods we use to derive and characterize human induced pluripotent stem (iPS) cells. We describe in order, first our culture techniques for the starting fibroblast populations and methods for retrovirus preparation and concentration. Subsequently, a detailed iPS derivation protocol suitable for human fibroblast populations is discussed using standard retroviral vectors expressing the classic four or three reprogramming genes. Finally, we elaborate a robust technique for monitoring and identification of potential iPS cells through live staining of reprogrammed cells. We also outline steps for characterization of the resulting iPS cell lines.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Células Madre Pluripotentes Inducidas/fisiología , Animales , Técnicas de Cultivo de Célula/instrumentación , Separación Celular/métodos , Células Cultivadas , Reprogramación Celular , Fibroblastos/citología , Fibroblastos/fisiología , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Ratones , Retroviridae/genética , Retroviridae/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA