Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-38507076

RESUMEN

PURPOSE: To describe the clinical, audiological, and psychometric features observed in patients with chronic tinnitus and rare variants in the ANK2 gene. METHODS: We report a case series of 12 patients with chronic tinnitus and heterozygous variants in the ANK2 gene. Tinnitus phenotyping included audiological (standard and high-frequency audiometry, Auditory Brainstem Responses (ABR) and Auditory Middle Latency Responses (AMLR)), psychoacoustic and psychometric assessment by a Visual Analog Scale (VAS) for tinnitus annoyance, the Tinnitus Handicap Inventory (THI), the test on Hypersensitivity to Sound (THS-GÜF), the Patient Health Questionnaire (PHQ-9), the Hospital Anxiety and Depression Scale (HADS) and the Montreal Cognitive Assessment (MoCA). RESULTS: All patients reported a persistent, unilateral noise-type tinnitus, mainly described as white noise or narrowband noise. Seven patients (58%) were considered to have extreme phenotype (THI score > 76), and all patients reported some degree of hyperacusis (THS-GÜF score > 18 in 75% of patients). Seven patients scored MoCA < 26, regardless of the age reported, suggesting a mild cognitive disorder. ABR showed no significant differences in latencies and amplitudes between ears with or without tinnitus. Similarly, the latencies of Pa, Pb waves, and NaPa complex in the AMLR did not differ based on the presence of tinnitus. However, there were statistical differences in the amplitudes of Pa waves in AMLR, with significantly greater amplitudes observed in ears with tinnitus. CONCLUSION: Patients with ANK2 variants and severe tinnitus exhibit an endophenotype featuring hyperacusis, persistent noise-like tinnitus, high-frequency hearing loss, and decreased amplitudes in AMLR. However, anxiety, depression, and cognitive symptoms vary among individuals.

2.
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
3.
Cells ; 12(7)2023 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-37048061

RESUMEN

Genetic sensorineural hearing loss and Meniere disease have been associated with rare variations in the coding and non-coding region of the human genome. Most of these variants were classified as likely pathogenic or variants of unknown significance and require functional validation in cellular or animal models. Given the difficulties to obtain human samples and the raising concerns about animal experimentation, human-induced pluripotent stem cells emerged as cellular models to investigate the interaction of genetic and environmental factors in the pathogenesis of inner ear disorders. The generation of human sensory epithelia and neuron-like cells carrying the variants of interest may facilitate a better understanding of their role during differentiation. These cellular models will allow us to explore new strategies for restoring hearing and vestibular sensory epithelia as well as neurons. This review summarized the use of human-induced pluripotent stem cells in sensorineural hearing loss and Meniere disease and proposed some strategies for its application in clinical practice.


Asunto(s)
Pérdida Auditiva Sensorineural , Células Madre Pluripotentes Inducidas , Enfermedad de Meniere , Animales , Humanos , Enfermedad de Meniere/genética , Pérdida Auditiva Sensorineural/genética , Pérdida Auditiva Sensorineural/complicaciones , Diferenciación Celular/genética
4.
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.

5.
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
6.
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
7.
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
8.
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
10.
Stem Cell Res ; 16(3): 692-5, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27346198

RESUMEN

We generated an induced pluripotent stem cell (iPSC) line from a Bernard-Soulier Syndrome (BSS) patient carrying the mutation p.Trp71Arg in the GPIX locus (BSS1-PBMC-iPS4F4). Peripheral blood mononuclear cells (PBMCs) were reprogrammed using heat sensitive non-integrative Sendai viruses containing the reprogramming factors Oct3/4, SOX2, KLF4 and c-MYC. Successful silencing of the exogenous reprogramming factors was checked by RT-PCR. Characterization of BSS1-PBMC-iPS4F4 included mutation analysis of GPIX locus, Short Tandem Repeats (STR) profiling, alkaline phosphatase enzymatic activity, analysis of conventional pluripotency-associated factors at mRNA and protein level and in vivo differentiation studies. BSS1-PBMC-iPS4F4 will provide a powerful tool to study BSS.


Asunto(s)
Síndrome de Bernard-Soulier/patología , Células Madre Pluripotentes Inducidas/citología , Complejo GPIb-IX de Glicoproteína Plaquetaria/genética , Animales , Síndrome de Bernard-Soulier/metabolismo , Diferenciación Celular , Células Cultivadas , Reprogramación Celular , Femenino , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/trasplante , Cariotipo , Factor 4 Similar a Kruppel , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mutación , Teratoma/metabolismo , Teratoma/patología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
11.
Mol Ther ; 23(1): 158-70, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25292191

RESUMEN

Human embryonic stem cells (hESCs) are a unique in vitro model for studying human developmental biology and represent a potential source for cell replacement strategies. Platelets can be generated from cord blood progenitors and hESCs; however, the molecular mechanisms and determinants controlling the in vitro megakaryocytic specification of hESCs remain elusive. We have recently shown that stem cell leukemia (SCL) overexpression accelerates the emergence of hemato-endothelial progenitors from hESCs and promotes their subsequent differentiation into blood cells with higher clonogenic potential. Given that SCL participates in megakaryocytic commitment, we hypothesized that it may potentiate megakaryopoiesis from hESCs. We show that ectopic SCL expression enhances the emergence of megakaryocytic precursors, mature megakaryocytes (MKs), and platelets in vitro. SCL-overexpressing MKs and platelets respond to different activating stimuli similar to their control counterparts. Gene expression profiling of megakaryocytic precursors shows that SCL overexpression renders a megakaryopoietic molecular signature. Connectivity Map analysis reveals that trichostatin A (TSA) and suberoylanilide hydroxamic acid (SAHA), both histone deacetylase (HDAC) inhibitors, functionally mimic SCL-induced effects. Finally, we confirm that both TSA and SAHA treatment promote the emergence of CD34(+) progenitors, whereas valproic acid, another HDAC inhibitor, potentiates MK and platelet production. We demonstrate that SCL and HDAC inhibitors are megakaryopoiesis regulators in hESCs.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Plaquetas/metabolismo , Células Madre Embrionarias/metabolismo , Redes Reguladoras de Genes , Megacariocitos/metabolismo , Proteínas Proto-Oncogénicas/genética , Trombopoyesis/genética , Antígenos CD34/genética , Antígenos CD34/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Plaquetas/citología , Plaquetas/efectos de los fármacos , Diferenciación Celular , Linaje de la Célula/efectos de los fármacos , Linaje de la Célula/genética , Células Cultivadas , Células Madre Embrionarias/citología , Células Madre Embrionarias/efectos de los fármacos , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Ácidos Hidroxámicos/farmacología , Megacariocitos/citología , Megacariocitos/efectos de los fármacos , Plásmidos/metabolismo , Mapeo de Interacción de Proteínas , Proteínas Proto-Oncogénicas/metabolismo , Proteína 1 de la Leucemia Linfocítica T Aguda , Trombopoyesis/efectos de los fármacos , Transcripción Genética , Ácido Valproico/farmacología , Vorinostat
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...