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










Base de datos
Intervalo de año de publicación
1.
Cells ; 12(23)2023 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-38067166

RESUMEN

Human embryonic stem cells (hESCs) differentiate into specialized cells, including midbrain dopaminergic neurons (DANs), and Non-human primates (NHPs) injected with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine develop some alterations observed in Parkinson's disease (PD) patients. Here, we obtained well-characterized DANs from hESCs and transplanted them into two parkinsonian monkeys to assess their behavioral and imaging changes. DANs from hESCs expressed dopaminergic markers, generated action potentials, and released dopamine (DA) in vitro. These neurons were transplanted bilaterally into the putamen of parkinsonian NHPs, and using magnetic resonance imaging techniques, we calculated the fractional anisotropy (FA) and mean diffusivity (MD), both employed for the first time for these purposes, to detect in vivo axonal and cellular density changes in the brain. Likewise, positron-emission tomography scans were performed to evaluate grafted DANs. Histological analyses identified grafted DANs, which were quantified stereologically. After grafting, animals showed signs of partially improved motor behavior in some of the HALLWAY motor tasks. Improvement in motor evaluations was inversely correlated with increases in bilateral FA. MD did not correlate with behavior but presented a negative correlation with FA. We also found higher 11C-DTBZ binding in positron-emission tomography scans associated with grafts. Higher DA levels measured by microdialysis after stimulation with a high-potassium solution or amphetamine were present in grafted animals after ten months, which has not been previously reported. Postmortem analysis of NHP brains showed that transplanted DANs survived in the putamen long-term, without developing tumors, in immunosuppressed animals. Although these results need to be confirmed with larger groups of NHPs, our molecular, behavioral, biochemical, and imaging findings support the integration and survival of human DANs in this pre-clinical PD model.


Asunto(s)
Células Madre Embrionarias Humanas , Enfermedad de Parkinson , Animales , Humanos , Neuronas Dopaminérgicas/metabolismo , Células Madre Embrionarias Humanas/metabolismo , Haplorrinos/metabolismo , Mesencéfalo/metabolismo , Dopamina/metabolismo , Enfermedad de Parkinson/terapia , Enfermedad de Parkinson/metabolismo
2.
Mol Ther Nucleic Acids ; 33: 75-92, 2023 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-37416759

RESUMEN

Bernard-Soulier syndrome (BSS) is a rare congenital disease characterized by macrothrombocytopenia and frequent bleeding. It is caused by pathogenic variants in three genes (GP1BA, GP1BB, or GP9) that encode for the GPIbα, GPIbß, and GPIX subunits of the GPIb-V-IX complex, the main platelet surface receptor for von Willebrand factor, being essential for platelet adhesion and aggregation. According to the affected gene, we distinguish BSS type A1 (GP1BA), type B (GP1BB), or type C (GP9). Pathogenic variants in these genes cause absent, incomplete, or dysfunctional GPIb-V-IX receptor and, consequently, a hemorrhagic phenotype. Using gene-editing tools, we generated knockout (KO) human cellular models that helped us to better understand GPIb-V-IX complex assembly. Furthermore, we developed novel lentiviral vectors capable of correcting GPIX expression, localization, and functionality in human GP9-KO megakaryoblastic cell lines. Generated GP9-KO induced pluripotent stem cells produced platelets that recapitulated the BSS phenotype: absence of GPIX on the membrane surface and large size. Importantly, gene therapy tools reverted both characteristics. Finally, hematopoietic stem cells from two unrelated BSS type C patients were transduced with the gene therapy vectors and differentiated to produce GPIX-expressing megakaryocytes and platelets with a reduced size. These results demonstrate the potential of lentiviral-based gene therapy to rescue BSS type C.

3.
Cell Death Dis ; 14(6): 357, 2023 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-37301844

RESUMEN

Pediatric Acute Myeloid Leukemia (AML) is a rare and heterogeneous disease characterized by a high prevalence of gene fusions as driver mutations. Despite the improvement of survival in the last years, about 50% of patients still experience a relapse. It is not possible to improve prognosis only with further intensification of chemotherapy, as come with a severe cost to the health of patients, often resulting in treatment-related death or long-term sequels. To design more effective and less toxic therapies we need a better understanding of pediatric AML biology. The NUP98-KDM5A chimeric protein is exclusively found in a particular subgroup of young pediatric AML patients with complex karyotypes and poor prognosis. In this study, we investigated the impact of NUP98-KDM5A expression on cellular processes in human Pluripotent Stem Cell models and a patient-derived cell line. We found that NUP98-KDM5A generates genomic instability through two complementary mechanisms that involve accumulation of DNA damage and direct interference of RAE1 activity during mitosis. Overall, our data support that NUP98-KDM5A promotes genomic instability and likely contributes to malignant transformation.


Asunto(s)
Leucemia Mieloide Aguda , Proteínas de Fusión Oncogénica , Humanos , Niño , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Complejo Poro Nuclear/metabolismo , Proteínas Oncogénicas/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Inestabilidad Genómica , Proteína 2 de Unión a Retinoblastoma/metabolismo
4.
Biomed Pharmacother ; 162: 114627, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37018985

RESUMEN

Osteosarcomas are frequently associated to a poor prognosis and a modest response to current treatments. EC-8042 is a well-tolerated mithramycin analog that has demonstrated an efficient ability to eliminate tumor cells, including cancer stem cell subpopulations (CSC), in sarcomas. In transcriptomic and protein expression analyses, we identified NOTCH1 signaling as one of the main pro-stemness pathways repressed by EC-8042 in osteosarcomas. Overexpression of NOTCH-1 resulted in a reduced anti-tumor effect of EC-8042 in CSC-enriched 3D tumorspheres cultures. On the other hand, the depletion of the NOTCH-1 downstream target HES-1 was able to enhance the action of EC-8042 on CSCs. Moreover, HES1 depleted cells failed to recover after treatment withdrawal and showed reduced tumor growth potential in vivo. In contrast, mice xenografted with NOTCH1-overexpressing cells responded worse than parental cells to EC-8042. Finally, we found that active NOTCH1 levels in sarcoma patients was associated to advanced disease and lower survival. Overall, these data highlight the relevant role that NOTCH1 signaling plays in mediating stemness in osteosarcoma. Moreover, we demonstrate that EC-8042 is powerful inhibitor of NOTCH signaling and that the anti-CSC activity of this mithramycin analog highly rely on its ability to repress this pathway.


Asunto(s)
Neoplasias Óseas , Osteosarcoma , Animales , Ratones , Neoplasias Óseas/patología , Línea Celular Tumoral , Células Madre Neoplásicas/metabolismo , Osteosarcoma/patología , Plicamicina/farmacología , Receptor Notch1/metabolismo , Receptores Notch/metabolismo
5.
Front Cell Dev Biol ; 10: 846092, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35721502

RESUMEN

Pediatric acute myeloid leukemia (AML) is a rare and heterogeneous disease that remains the major cause of mortality in children with leukemia. To improve the outcome of pediatric AML we need to gain knowledge on the biological bases of this disease. NUP98-KDM5A (NK5A) fusion protein is present in a particular subgroup of young pediatric patients with poor outcome. We report the generation and characterization of human Embryonic Stem Cell (hESC) clonal lines with inducible expression of NK5A. Temporal control of NK5A expression during hematopoietic differentiation from hESC will be critical for elucidating its participation during the leukemogenic process.

6.
Front Immunol ; 13: 803995, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35493522

RESUMEN

In 1975 two independent groups noticed the presence of immune cells with a unique ability to recognize and eliminate transformed hematopoietic cells without any prior sensitization or expansion of specific clones. Since then, NK cells have been the axis of thousands of studies that have resulted until June 2021, in more than 70 000 publications indexed in PubMed. As result of this work, which include approaches in vitro, in vivo, and in natura, it has been possible to appreciate the role played by the NK cells, not only as effectors against specific pathogens, but also as regulators of the immune response. Recent advances have revealed previous unidentified attributes of NK cells including the ability to adapt to new conditions under the context of chronic infections, or their ability to develop some memory-like characteristics. In this review, we will discuss significant findings that have rule our understanding of the NK cell biology, the developing of these findings into new concepts in immunology, and how these conceptual platforms are being used in the design of strategies for cancer immunotherapy.


Asunto(s)
Neoplasias Hematológicas , Neoplasias , Neoplasias Hematológicas/terapia , Humanos , Inmunoterapia/métodos , Células Asesinas Naturales , Neoplasias/terapia
7.
Blood Adv ; 5(23): 4842-4854, 2021 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-34470043

RESUMEN

Acute myeloid leukemia (AML) is the most common acute leukemia in adults. Disease heterogeneity is well documented, and patient stratification determines treatment decisions. Patient-derived xenografts (PDXs) from risk-stratified AML are crucial for studying AML biology and testing novel therapeutics. Despite recent advances in PDX modeling of AML, reproducible engraftment of human AML is primarily limited to high-risk (HR) cases, with inconsistent or very protracted engraftment observed for favorable-risk (FR) and intermediate-risk (IR) patients. We used NSGS mice to characterize the engraftment robustness/kinetics of 28 AML patient samples grouped according to molecular/cytogenetic classification and assessed whether the orthotopic coadministration of patient-matched bone marrow mesenchymal stromal cells (BM MSCs) improves AML engraftment. PDX event-free survival correlated well with the predictable prognosis of risk-stratified AML patients. The majority (85-94%) of the mice were engrafted in bone marrow (BM) independently of the risk group, although HR AML patients showed engraftment levels that were significantly superior to those of FR or IR AML patients. Importantly, the engraftment levels observed in NSGS mice by week 6 remained stable over time. Serial transplantation and long-term culture-initiating cell (LTC-IC) assays revealed long-term engraftment limited to HR AML patients, fitter leukemia-initiating cells (LICs) in HR AML samples, and the presence of AML LICs in the CD34- leukemic fraction, regardless of the risk group. Finally, orthotopic coadministration of patient-matched BM MSCs and AML cells was dispensable for BM engraftment levels but favored peripheralization of engrafted AML cells. This comprehensive characterization of human AML engraftment in NSGS mice offers a valuable platform for in vivo testing of targeted therapies in risk-stratified AML patient samples.


Asunto(s)
Leucemia Mieloide Aguda , Animales , Antígenos CD34 , Médula Ósea , Humanos , Leucemia Mieloide Aguda/terapia , Ratones , Ratones Endogámicos NOD , Ratones SCID
8.
Life Sci Alliance ; 3(5)2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32284355

RESUMEN

Mammals optimize their physiology to the light-dark cycle by synchronization of the master circadian clock in the brain with peripheral clocks in the rest of the tissues of the body. Circadian oscillations rely on a negative feedback loop exerted by the molecular clock that is composed by transcriptional activators Bmal1 and Clock, and their negative regulators Period and Cryptochrome. Components of the molecular clock are expressed during early development, but onset of robust circadian oscillations is only detected later during embryogenesis. Here, we have used naïve pluripotent mouse embryonic stem cells (mESCs) to study the role of Bmal1 during early development. We found that, compared to wild-type cells, Bmal1-/- mESCs express higher levels of Nanog protein and altered expression of pluripotency-associated signalling pathways. Importantly, Bmal1-/- mESCs display deficient multi-lineage cell differentiation capacity during the formation of teratomas and gastrula-like organoids. Overall, we reveal that Bmal1 regulates pluripotent cell differentiation and propose that the molecular clock is an hitherto unrecognized regulator of mammalian development.


Asunto(s)
Factores de Transcripción ARNTL/metabolismo , Diferenciación Celular/fisiología , Células Madre Embrionarias de Ratones/metabolismo , Factores de Transcripción ARNTL/fisiología , Animales , Proteínas CLOCK/genética , Proteínas CLOCK/metabolismo , Relojes Circadianos/fisiología , Ritmo Circadiano/genética , Retroalimentación Fisiológica/fisiología , Expresión Génica/genética , Células Madre Pluripotentes Inducidas/citología , Ratones , Células Madre Embrionarias de Ratones/citología , Proteínas Circadianas Period/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Transcripción Genética
9.
Stem Cells Transl Med ; 9(5): 636-650, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32073751

RESUMEN

Multipotent mesenchymal stromal cells (MSCs) have emerged as a promising cell therapy in regenerative medicine and for autoimmune/inflammatory diseases. However, a main hurdle for MSCs-based therapies is the loss of their proliferative potential in vitro. Here we report that glycoprotein A repetitions predominant (GARP) is required for the proliferation and survival of adipose-derived MSCs (ASCs) via its regulation of transforming growth factor-ß (TGF-ß) activation. Silencing of GARP in human ASCs increased their activation of TGF-ß which augmented the levels of mitochondrial reactive oxygen species (mtROS), resulting in DNA damage, a block in proliferation and apoptosis. Inhibition of TGF-ß signaling reduced the levels of mtROS and DNA damage and restored the ability of GARP-/low ASCs to proliferate. In contrast, overexpression of GARP in ASCs increased their proliferative capacity and rendered them more resistant to etoposide-induced DNA damage and apoptosis, in a TGF-ß-dependent manner. In summary, our data show that the presence or absence of GARP on ASCs gives rise to distinct TGF-ß responses with diametrically opposing effects on ASC proliferation and survival.


Asunto(s)
Células Madre Mesenquimatosas/metabolismo , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Humanos
10.
Stem Cell Res ; 41: 101603, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31698193

RESUMEN

Familial Platelet Disorder with associated Myeloid Malignancy (FPDMM) is a rare platelet disorder caused by mutations in RUNX1. We generated an iPSC line (GENYOi005-A) from a FPDMM patient with a non-previously reported variant p.Thr196Ala. Non-integrative Sendai viruses expressing the Yamanaka reprogramming factors were used to reprogram peripheral blood mononuclear cells from this FPDMM patient. Characterization of GENYOi005-A included genetic analysis of RUNX1 locus, Short Tandem Repeats profiling, alkaline phosphatase enzymatic activity, expression of pluripotency-associated factors and differentiation studies in vitro and in vivo. This iPSC line will provide a powerful tool to study developmental alterations of FPDMM patients.


Asunto(s)
Trastornos de la Coagulación Sanguínea Heredados/genética , Trastornos de la Coagulación Sanguínea Heredados/patología , Trastornos de las Plaquetas Sanguíneas/genética , Trastornos de las Plaquetas Sanguíneas/patología , Diferenciación Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Células Madre Pluripotentes Inducidas/patología , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Leucocitos Mononucleares/patología , Mutación , Células Cultivadas , Reprogramación Celular , Femenino , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Leucocitos Mononucleares/metabolismo , Persona de Mediana Edad
11.
Stem Cell Res ; 37: 101446, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31035039

RESUMEN

ADNP syndrome is an intellectual disability associated with Autism spectrum disorder caused by mutations in ADNP. We generated an iPSC line from an ADNP syndrome pediatric patient harboring the mutation p.Trp719* (GENYOi004-A). Peripheral blood mononuclear cells were reprogrammed using a non-transmissible form of Sendai viruses expressing the four Yamanaka factors (Oct3/4, SOX2, KLF4 and c-MYC). Characterization of GENYOi004-A included mutation analysis of ADNP by allele-specific PCR, genetic identity by Short Tandem Repeats polymorphism profiling, alkaline phosphatase enzymatic activity, expression of pluripotency-associated factors and pluripotency studies in vivo. GENYOi004-A will be useful to evaluate ADNP syndrome alterations at early developmental stages.


Asunto(s)
Trastorno del Espectro Autista/genética , Diferenciación Celular , Proteínas de Homeodominio/genética , Células Madre Pluripotentes Inducidas/patología , Leucocitos Mononucleares/patología , Mutación , Proteínas del Tejido Nervioso/genética , Teratoma/etiología , Animales , Trastorno del Espectro Autista/patología , Células Cultivadas , Reprogramación Celular , Niño , Femenino , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Factor 4 Similar a Kruppel , Leucocitos Mononucleares/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Fenotipo , Teratoma/patología
12.
Stem Cell Res ; 25: 274-277, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29246573

RESUMEN

The Notch ligand DLL4 has key roles during embryonic development of different tissues, but most of the data comes from animal models. Here we describe the generation and characterization of 2 human Pluripotent Stem Cell (hPSC) lines that overexpress DLL4, as well as the two corresponding control hPSC lines. DLL4 expression can be detected at the mRNA and protein level, and does not affect the pluripotency of the cells. These hPSC lines can be used to study the role of DLL4 during human embryonic development.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/metabolismo , Células Madre Pluripotentes/citología , Proteínas Adaptadoras Transductoras de Señales , Adulto , Proteínas de Unión al Calcio , Diferenciación Celular , Línea Celular , Proliferación Celular , Células Cultivadas , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Células Madre Pluripotentes/metabolismo , Receptores Notch/genética , Receptores Notch/metabolismo
13.
Stem Cell Res ; 25: 286-290, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29246576

RESUMEN

HoxA9 is an evolutionarily conserved homeobox gene implicated in embryo development. To study the roles of Hoxa9 during human development we generated a transgenic H9 (hESC) line that overexpresses HoxA9 and the Enhanced Green Fluorescent Protein (EGFP), and a control H9 with a stable expression of the EGFP. The resulting H9-HoxA9-EGFP and H9-EGFP cell lines allow an efficient visualization of hESCs by fluorescent microscopy, quantification by flow cytometry and cell differentiation tracking. Both transgenic cell lines maintained the pluripotent phenotype, the ability to differentiate into all three germ layers and a normal karyotype.


Asunto(s)
Embrión de Mamíferos/metabolismo , Desarrollo Embrionario , Células Madre Embrionarias/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de Homeodominio/metabolismo , Diferenciación Celular , Células Cultivadas , Células Madre Embrionarias/citología , Femenino , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas de Homeodominio/genética , Humanos , Transfección
14.
Stem Cells ; 35(11): 2253-2266, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28869683

RESUMEN

Runt-related transcription factor 1 (Runx1) is a master hematopoietic transcription factor essential for hematopoietic stem cell (HSC) emergence. Runx1-deficient mice die during early embryogenesis due to the inability to establish definitive hematopoiesis. Here, we have used human pluripotent stem cells (hPSCs) as model to study the role of RUNX1 in human embryonic hematopoiesis. Although the three RUNX1 isoforms a, b, and c were induced in CD45+ hematopoietic cells, RUNX1c was the only isoform induced in hematoendothelial progenitors (HEPs)/hemogenic endothelium. Constitutive expression of RUNX1c in human embryonic stem cells enhanced the appearance of HEPs, including hemogenic (CD43+) HEPs and promoted subsequent differentiation into blood cells. Conversely, specific deletion of RUNX1c dramatically reduced the generation of hematopoietic cells from HEPs, indicating that RUNX1c is a master regulator of human hematopoietic development. Gene expression profiling of HEPs revealed a RUNX1c-induced proinflammatory molecular signature, supporting previous studies demonstrating proinflammatory signaling as a regulator of HSC emergence. Collectively, RUNX1c orchestrates hematopoietic specification of hPSCs, possibly in cooperation with proinflammatory signaling. Stem Cells 2017;35:2253-2266.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Perfilación de la Expresión Génica/métodos , Células Madre Pluripotentes/metabolismo , Animales , Diferenciación Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Humanos , Ratones , Transducción de Señal
15.
Stem Cell Res ; 19: 1-5, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28412998

RESUMEN

Pediatric Acute Megakaryoblastic Leukemia not associated to Down Syndrome (non-DS AMKL) is a rare disease with a dismal prognosis. Around 15% of patients carry the chromosomal translocation t(1;22) that originates the fusion oncogene RBM15-MKL1, which is linked to an earlier disease onset (median of 6months of age) and arises in utero. Here we report the generation of two hPSC cell lines constitutively expressing the oncogene RBM15-MKL1, resulting in an increased expression of known RBM15-MKL1 gene targets. These cell lines represent new disease models of pediatric AMKL to study the impact of the RBM15-MKL1 oncogene on human embryonic hematopoietic development.


Asunto(s)
Leucemia Megacarioblástica Aguda/patología , Proteínas de Fusión Oncogénica/metabolismo , Células Madre Pluripotentes/citología , Biomarcadores/metabolismo , Línea Celular , Niño , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Humanos , Leucemia Megacarioblástica Aguda/metabolismo , Modelos Biológicos , Proteínas de Fusión Oncogénica/genética , Células Madre Pluripotentes/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
16.
Stem Cell Res ; 20: 10-13, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28395735

RESUMEN

Bernard Soulier Syndrome (BSS) is a rare autosomal platelet disorder characterized by mutations in the von Willebrand factor platelet receptor complex GPIb-V-IX. In this work we have generated an induced pluripotent stem cell (BSS3-PBMC-iPS4F8) from peripheral blood mononuclear cells of a BSS patient with a p.Phe55Ser mutation in the GPIX gene. Characterization of BSS3-PBMC-iPS4F8 showed that these cells maintained the original mutation present in the BSS patient, expressed pluripotent stem cell markers and were able to differentiate into the three germline layers. This new iPSC line will contribute to better understand the biology of BSS disease.


Asunto(s)
Síndrome de Bernard-Soulier/patología , Células Madre Pluripotentes Inducidas/citología , Complejo GPIb-IX de Glicoproteína Plaquetaria/genética , Secuencia de Bases , Síndrome de Bernard-Soulier/genética , Síndrome de Bernard-Soulier/metabolismo , Diferenciación Celular , Línea Celular , Reprogramación Celular , Análisis Mutacional de ADN , Cuerpos Embrioides/metabolismo , Cuerpos Embrioides/patología , Femenino , Homocigoto , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Cariotipo , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/metabolismo , Polimorfismo de Nucleótido Simple , Secuencias Repetidas en Tándem/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
18.
Stem Cell Res ; 17(3): 603-606, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27934591

RESUMEN

Bernard Soulier Syndrome (BSS) is an inherited rare platelet disorder characterized by mutations in the platelet glycoprotein complex GPIb-IX-V. We generated an induced pluripotent stem cell (iPSC) line from a BSS patient with a mutation p.Asn45Ser in the GPIX locus (BSS2-PBMC-iPS4F24). Peripheral blood mononuclear cells were reprogrammed using non-integrative viral transduction. Characterization of BSS2-PBMC-iPS4F24 included mutational analysis of GPIX locus, analysis of conventional pluripotency-associated factors at mRNA and protein level and in vitro and in vivo differentiation studies. This iPSC line will provide a powerful tool to study the biology of BSS disease.


Asunto(s)
Síndrome de Bernard-Soulier/patología , Células Madre Pluripotentes Inducidas/citología , Complejo GPIb-IX de Glicoproteína Plaquetaria/genética , Animales , Secuencia de Bases , Síndrome de Bernard-Soulier/genética , Diferenciación Celular , Línea Celular , Reprogramación Celular , Análisis Mutacional de ADN , Cuerpos Embrioides/citología , Cuerpos Embrioides/metabolismo , Femenino , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/trasplante , Cariotipo , Leucocitos Mononucleares/citología , Ratones , Ratones SCID , Polimorfismo de Nucleótido Simple , Teratoma/patología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
20.
Stem Cell Reports ; 7(4): 602-618, 2016 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-27666791

RESUMEN

Induced pluripotent stem cells (iPSCs) are a powerful tool for disease modeling. They are routinely generated from healthy donors and patients from multiple cell types at different developmental stages. However, reprogramming leukemias is an extremely inefficient process. Few studies generated iPSCs from primary chronic myeloid leukemias, but iPSC generation from acute myeloid or lymphoid leukemias (ALL) has not been achieved. We attempted to generate iPSCs from different subtypes of B-ALL to address the developmental impact of leukemic fusion genes. OKSM(L)-expressing mono/polycistronic-, retroviral/lentiviral/episomal-, and Sendai virus vector-based reprogramming strategies failed to render iPSCs in vitro and in vivo. Addition of transcriptomic-epigenetic reprogramming "boosters" also failed to generate iPSCs from B cell blasts and B-ALL lines, and when iPSCs emerged they lacked leukemic fusion genes, demonstrating non-leukemic myeloid origin. Conversely, MLL-AF4-overexpressing hematopoietic stem cells/B progenitors were successfully reprogrammed, indicating that B cell origin and leukemic fusion gene were not reprogramming barriers. Global transcriptome/DNA methylome profiling suggested a developmental/differentiation refractoriness of MLL-rearranged B-ALL to reprogramming into pluripotency.


Asunto(s)
Transdiferenciación Celular/genética , Reprogramación Celular , Reordenamiento Génico , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Animales , Biomarcadores , Línea Celular Transformada , Línea Celular Tumoral , Análisis por Conglomerados , Metilación de ADN , Expresión Génica , Perfilación de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Xenoinjertos , Humanos , Ratones , Células Progenitoras Mieloides/metabolismo , Proteínas de Fusión Oncogénica/genética , Fenotipo , Células Precursoras de Linfocitos B/metabolismo , Transcriptoma , Translocación Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...