Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Chem Biol ; 30(7): 780-794.e8, 2023 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-37379846

RESUMEN

Overlapping principles of embryonic and tumor biology have been described, with recent multi-omics campaigns uncovering shared molecular profiles between human pluripotent stem cells (hPSCs) and adult tumors. Here, using a chemical genomic approach, we provide biological evidence that early germ layer fate decisions of hPSCs reveal targets of human cancers. Single-cell deconstruction of hPSCs-defined subsets that share transcriptional patterns with transformed adult tissues. Chemical screening using a unique germ layer specification assay for hPSCs identified drugs that enriched for compounds that selectively suppressed the growth of patient-derived tumors corresponding exclusively to their germ layer origin. Transcriptional response of hPSCs to germ layer inducing drugs could be used to identify targets capable of regulating hPSC specification as well as inhibiting adult tumors. Our study demonstrates properties of adult tumors converge with hPSCs drug induced differentiation in a germ layer specific manner, thereby expanding our understanding of cancer stemness and pluripotency.


Asunto(s)
Neoplasias , Células Madre Pluripotentes , Humanos , Diferenciación Celular/fisiología , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Genómica
2.
Drug Discov Today ; 27(12): 103407, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36243303

RESUMEN

The discovery and development of effective drugs for cancer patients has seen limited success in the clinic from phase I trials onward. The high attrition rate of current drug development approaches requires careful evaluation to provide a better understanding of the factors that correlate with or predict positive clinical outcomes. Here, we examine pre-clinical drug development approaches and conduct a meta-analysis of 2918 clinical studies involving 466 unique drugs tested in clinical trials for acute myeloid leukemia (AML). Our goal was to determine whether there are key shared pre-clinical characteristics that ultimately relate to successful or unsuccessful drugs in patients. We provide an evidence-based recommendation for the use of phenotypic drug discovery rather than other methods during pre-clinical development. Although our analysis was limited to AML, similar analyses are likely to be informative for other tumor-specific drug discovery campaigns, informing and improving the foundational discovery screens and platforms for other cancers.


Asunto(s)
Leucemia Mieloide Aguda , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Descubrimiento de Drogas
3.
F1000Res ; 10: 964, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34909193

RESUMEN

Data from the Istituto Superiore di Sanità (ISS) emphasized by the media indicate that COVID-19 vaccination reduces related infections, hospitalizations and deaths. However, a comparison showed significantly more hospitalizations and intensive care unit accesses in the corresponding months and days in 2021 versus 2020 and no significant differences in deaths. The combination of non-alternative hypotheses may help explain the discrepancy between the results in the entire population and the vaccination's success claimed by the ISS in reducing infections, serious cases and deaths: a bias: counting as unvaccinated also "those vaccinated with 1 dose in the two weeks following the inoculation", and as incompletely vaccinated also "those vaccinated with 2 doses within two weeks of the 2nd inoculation".a systematic error: counting as unvaccinated also "vaccinated with 1 dose in the two weeks following the inoculation", and as incompletely vaccinated also "vaccinated with 2 doses within two weeks of the 2nd inoculation". Many reports show an increase in COVID-19 cases in these time-windows, and related data should be separated levels of protective effectiveness in vaccinated people, often considered stable, actually show signs of progressive reduction over time, which could contribute to reducing the overall population resultunvaccinated people show more severe disease than in 2020, supporting also in humans the theory of imperfect vaccines, which offer less resistance to the entry of germs than the resistance later encountered inside the human body. This favors the selection of more resistant and virulent mutants, that can be spread by vaccinated people. This damages first the unvaccinated people, but ultimately the whole community. An open scientific debate is needed to discuss these hypotheses, following the available evidence (as well as to discuss the inconsistent theory of unvaccinated young people as reservoirs of viruses/mutants), to assess the long-term and community impact of different vaccination strategies.


Asunto(s)
COVID-19 , Adolescente , Vacunas contra la COVID-19 , Hospitalización , Humanos , SARS-CoV-2 , Vacunación
4.
Cell Chem Biol ; 28(10): 1394-1406.e10, 2021 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-33979648

RESUMEN

Natural products (NPs) encompass a rich source of bioactive chemical entities. Here, we used human cancer stem cells (CSCs) in a chemical genomics campaign with NP chemical space to interrogate extracts from diverse strains of actinomycete for anti-cancer properties. We identified a compound (McM25044) capable of selectively inhibiting human CSC function versus normal stem cell counterparts. Biochemical and molecular studies revealed that McM025044 exerts inhibition on human CSCs through the small ubiquitin-like modifier (SUMO) cascade, found to be hyperactive in a variety of human cancers. McM025044 impedes the SUMOylation pathway via direct targeting of the SAE1/2 complex. Treatment of patient-derived CSCs resulted in reduced levels of SUMOylated proteins and suppression of progenitor and stem cell capacity measured in vitro and in vivo. Our study overcomes a barrier in chemically inhibiting oncogenic SUMOylation activity and uncovers a unique role for SAE2 in the biology of human cancers.


Asunto(s)
Células Madre Neoplásicas/metabolismo , Enzimas Activadoras de Ubiquitina/metabolismo , Animales , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Sitios de Unión , Productos Biológicos/química , Productos Biológicos/metabolismo , Productos Biológicos/farmacología , Productos Biológicos/uso terapéutico , Línea Celular Tumoral , Autorrenovación de las Células , Supervivencia Celular/efectos de los fármacos , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/patología , Ratones , Simulación del Acoplamiento Molecular , Células Madre Neoplásicas/citología , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Sumoilación/efectos de los fármacos , Enzimas Activadoras de Ubiquitina/química , Enzimas Activadoras de Ubiquitina/genética
5.
Cell Rep ; 34(10): 108818, 2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33691101

RESUMEN

Histone variants (HVs) are a subfamily of epigenetic regulators implicated in embryonic development, but their role in human stem cell fate remains unclear. Here, we reveal that the phosphorylation state of the HV H2A.X (γH2A.X) regulates self-renewal and differentiation of human pluripotent stem cells (hPSCs) and leukemic progenitors. As demonstrated by CRISPR-Cas deletion, H2A.X is essential in maintaining normal hPSC behavior. However, reduced levels of γH2A.X enhances hPSC differentiation toward the hematopoietic lineage with concomitant inhibition of neural development. In contrast, activation and sustained levels of phosphorylated H2A.X enhance hPSC neural fate while suppressing hematopoiesis. This controlled lineage bias correlates to occupancy of γH2A.X at genomic loci associated with ectoderm versus mesoderm specification. Finally, drug modulation of H2A.X phosphorylation overcomes differentiation block of patient-derived leukemic progenitors. Our study demonstrates HVs may serve to regulate pluripotent cell fate and that this biology could be extended to somatic cancer stem cell control.


Asunto(s)
Autorrenovación de las Células/fisiología , Histonas/metabolismo , Células Madre Neoplásicas/citología , Células Madre Pluripotentes/citología , Sistemas CRISPR-Cas/genética , Diferenciación Celular , Linaje de la Célula , Ectodermo/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Histonas/deficiencia , Histonas/genética , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Mesodermo/metabolismo , Células Madre Neoplásicas/metabolismo , Neuronas/citología , Neuronas/metabolismo , Nucleosomas/metabolismo , Fosforilación , Células Madre Pluripotentes/metabolismo
6.
Cell Rep ; 34(11): 108845, 2021 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-33730576

RESUMEN

Identifying precise targets of individual cancers remains challenging. Chronic lymphocytic leukemia (CLL) represents the most common adult hematologic malignancy, and trisomy 12 (tri12) represents a quarter of CLL patients. We report that tri12 human pluripotent stem cells (hPSCs) allow for the identification of gene networks and targets specific to tri12, which are controlled by comparative normal PSCs. Identified targets are upregulated in tri12 leukemic cells from a cohort of 159 patients with monoclonal B cell lymphocytosis and CLL. tri12 signaling patterns significantly influence progression-free survival. Actionable targets are identified using high-content drug testing and functionally validated in an additional 44 CLL patient samples. Using xenograft models, interleukin-1 receptor-associated kinase 4 (IRAK4) inhibitor is potent and selective against human tri12 CLL versus healthy patient-derived xenografts. Our study uses hPSCs to uncover targets from genetic aberrations and apply them to cancer. These findings provide immediate translational potential as biomarkers and targets for therapeutic intervention.


Asunto(s)
Leucemia Linfocítica Crónica de Células B/genética , Células Madre Pluripotentes/metabolismo , Trisomía/genética , Adulto , Anciano , Anciano de 80 o más Años , Animales , Línea Celular , Progresión de la Enfermedad , Femenino , Dosificación de Gen , Redes Reguladoras de Genes , Humanos , Leucemia Linfocítica Crónica de Células B/patología , Masculino , Ratones Endogámicos NOD , Persona de Mediana Edad , Modelos Genéticos , Reproducibilidad de los Resultados , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Cell ; 177(4): 910-924.e22, 2019 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-30982595

RESUMEN

The assembly of organized colonies is the earliest manifestation in the derivation or induction of pluripotency in vitro. However, the necessity and origin of this assemblance is unknown. Here, we identify human pluripotent founder cells (hPFCs) that initiate, as well as preserve and establish, pluripotent stem cell (PSC) cultures. PFCs are marked by N-cadherin expression (NCAD+) and reside exclusively at the colony boundary of primate PSCs. As demonstrated by functional analysis, hPFCs harbor the clonogenic capacity of PSC cultures and emerge prior to commitment events or phenotypes associated with pluripotent reprogramming. Comparative single-cell analysis with pre- and post-implantation primate embryos revealed hPFCs share hallmark properties with primitive endoderm (PrE) and can be regulated by non-canonical Wnt signaling. Uniquely informed by primate embryo organization in vivo, our study defines a subset of founder cells critical to the establishment pluripotent state.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Diferenciación Celular , Linaje de la Célula , Desarrollo Embrionario , Células Madre Embrionarias/metabolismo , Endodermo/metabolismo , Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica/genética , Humanos , Análisis de la Célula Individual , Vía de Señalización Wnt
8.
Int J Mol Sci ; 18(9)2017 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-28902128

RESUMEN

Evaluation of the extent and nature of induced pluripotent stem cell (iPSC) genetic instability is important for both basic research and future clinical use. As previously demonstrated regarding embryonic stem cells, such DNA aberrations might affect the differentiation capacity of the cells and increase their tumorigenicity. Here, we first focus on the contribution of multiple DNA damage response pathways during cellular reprogramming. We then discuss the origin and mechanisms responsible for the modification of genetic material in iPSCs (pre-existing variations in somatic cells, mutations induced by reprogramming factors, and mutations induced by culture expansion) and deepen the possible functional consequences of genetic variations in these cells. Lastly, we present some recent improvements of iPSC generation methods aimed at obtaining cells with fewer genetic variations.


Asunto(s)
Reprogramación Celular/fisiología , Inestabilidad Genómica , Células Madre Pluripotentes Inducidas/fisiología , Técnicas de Reprogramación Celular/métodos , Inestabilidad Cromosómica , Daño del ADN/fisiología , Reparación del ADN/fisiología , Dosificación de Gen , Variación Genética , Humanos , Mutación , Estrés Oxidativo/fisiología
9.
Stem Cells ; 35(9): 2095-2102, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28758276

RESUMEN

Induced pluripotent stem cell reprogramming has provided critical insights into disease processes by modeling the genetics and related clinical pathophysiology. Human cancer represents highly diverse genetics, as well as inter- and intra-patient heterogeneity, where cellular model systems capable of capturing this disease complexity would be invaluable. Acute myeloid leukemia (AML) represents one of most heterogeneous cancers and has been divided into genetic subtypes correlated with unique risk stratification over the decades. Here, we report our efforts to induce pluripotency from the heterogeneous population of human patients that represents this disease in the clinic. Using robust optimized reprogramming methods, we demonstrate that reprogramming of AML cells harboring leukemic genomic aberrations is a rare event with the exception of those with de novo mixed-lineage leukemia (MLL) mutations that can be reprogrammed and model drug responses in vitro. Our findings indicate that unlike hematopoietic cells devoid of genomic aberrations, AML cells harboring driver mutations are refractory to reprogramming. Expression of MLL fusion proteins in AML cells did not contribute to induced reprogramming success, which continued to select for patient derived cells devoid of AML patient-specific aberrations. Our study reveals that unanticipated blockades to achieving pluripotency reside within the majority of transformed AML patient cells. Stem Cells 2017;35:2095-2102.


Asunto(s)
Reprogramación Celular , Hematopoyesis , Células Madre Pluripotentes Inducidas/patología , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Mutación/genética , Células de la Médula Ósea/patología , Humanos , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Proteínas de Fusión Oncogénica/metabolismo
10.
Cell Chem Biol ; 24(7): 833-844.e9, 2017 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-28648376

RESUMEN

Targeting of human cancer stem cells (CSCs) requires the identification of vulnerabilities unique to CSCs versus healthy resident stem cells (SCs). Unfortunately, dysregulated pathways that support transformed CSCs, such as Wnt/ß-catenin signaling, are also critical regulators of healthy SCs. Using the ICG-001 and CWP family of small molecules, we reveal Sam68 as a previously unappreciated modulator of Wnt/ß-catenin signaling within CSCs. Disruption of CBP-ß-catenin interaction via ICG-001/CWP induces the formation of a Sam68-CBP complex in CSCs that alters Wnt signaling toward apoptosis and differentiation induction. Our study identifies Sam68 as a regulator of human CSC vulnerability.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Unión al ADN/metabolismo , Células Madre Neoplásicas/metabolismo , Fragmentos de Péptidos/metabolismo , Proteínas de Unión al ARN/metabolismo , Sialoglicoproteínas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/genética , Adulto , Anciano , Animales , Apoptosis/efectos de los fármacos , Compuestos de Azabiciclo/farmacología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Femenino , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Masculino , Ratones , Ratones Endogámicos NOD , Persona de Mediana Edad , Células Madre Neoplásicas/citología , Células Madre Neoplásicas/trasplante , Organofosfatos/farmacología , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Pirimidinonas/farmacología , Interferencia de ARN , Proteínas de Unión al ARN/antagonistas & inhibidores , Proteínas de Unión al ARN/genética , Sialoglicoproteínas/antagonistas & inhibidores , Sialoglicoproteínas/genética , Sumoilación/efectos de los fármacos , Transcriptoma/efectos de los fármacos , Vía de Señalización Wnt/efectos de los fármacos , beta Catenina/metabolismo
11.
Stem Cell Res ; 15(1): 240-2, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26141785

RESUMEN

The combination of OCT4 expression and short-term exposure to reprogramming media induces a state of transcriptional plasticity in human fibroblasts, capable of responding to changes in the extracellular environment. Here we provide characterization of iPSCs established through continued culture of OCT4-induced plastic human fibroblasts in pluripotent-supportive reprogramming media. Human iPSC(OCT4) are morphologically indistinguishable from conventionally derived iPSCs and express core proteins involved in maintenance of pluripotency. iPSC(OCT4) display bona fide functional pluripotency as measured by in vivo teratoma formation consisting of the three germ layers.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Factor 3 de Transcripción de Unión a Octámeros/farmacología , Adulto , Animales , Reprogramación Celular/efectos de los fármacos , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Ratones Endogámicos NOD , Ratones SCID
12.
Stem Cell Res ; 15(1): 221-30, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26117529

RESUMEN

The combination of OCT4 expression and short-term exposure to reprogramming media induces a state of transcriptional plasticity in human fibroblasts, capable of responding to changes in the extracellular environment that facilitate direct cell fate conversion toward lineage specific progenitors. Here we reveal that continued exposure of OCT4-induced plastic human fibroblasts to reprogramming media (RM) is sufficient to induce pluripotency. OCT4-derived induced pluripotent stem cell (iPSC(OCT4)) colonies emerged after prolonged culture in RM, and formed independently of lineage specific progenitors. Human iPSC(OCT4) are morphologically indistinguishable from conventionally derived iPSCs and express core proteins involved in maintenance of pluripotency. iPSC(OCT4) display in vivo functional pluripotency as measured by teratoma formation consisting of the three germ layers, and are capable of targeted in vitro differentiation. Our study indicates that acquisition of pluripotency is one of multiple cell fate choices that can be facilitated through environmental stimulation of OCT4-induced plasticity, and suggests the role of other reprogramming factors to induce pluripotency can be substituted by prolonged culture of plastic fibroblasts.


Asunto(s)
Fibroblastos/citología , Células Madre Pluripotentes Inducidas/citología , Factor 3 de Transcripción de Unión a Octámeros/farmacología , Adulto , Animales , Linaje de la Célula/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Humanos , Inmunofenotipificación , Ratones Endogámicos NOD , Ratones SCID , Modelos Biológicos
13.
Int J Mol Sci ; 14(2): 2617-36, 2013 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-23358251

RESUMEN

Recent studies have shown that mouse embryonic stem cells (mESCs) rely on a distinctive genome caretaking network. In this review, we will discuss how mESCs functionally respond to DNA damage and describe several modifications in mESC DNA damage response, which accommodate dynamic cycling and preservation of genetic information. Subsequently, we will discuss how the transition from mESCs to adult stem/progenitor cells can be involved in the decline of tissue integrity and function in the elderly.

14.
Small ; 8(20): 3192-200, 2012 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-22821625

RESUMEN

Highly bright and photostable cyanine dye-doped silica nanoparticles, IRIS Dots, are developed, which can efficiently label human mesenchymal stem cells (hMSCs). The application procedure used to label hMSCs is fast (2 h), the concentration of IRIS Dots for efficient labeling is low (20 µg mL(-1) ), and the labeled cells can be visualized by flow cytometry, confocal microscopy, and transmission electron microscopy. Labeled hMSCs are unaffected in their viability and proliferation, as well as stemness surface marker expression and differentiation capability into osteocytes. Moreover, this is the first report that shows nonfunctionalized IRIS Dots can discriminate between live and early-stage apoptotic stem cells (both mesenchymal and embryonic) through a distinct external cell surface distribution. On the basis of biocompatibility, efficient labeling, and apoptotic discrimination potential, it is suggested that IRIS Dots can serve as a promising stem cell tracking agent.


Asunto(s)
Nanopartículas/química , Imagen Óptica/métodos , Dióxido de Silicio/química , Células Madre , Apoptosis/fisiología , Células Cultivadas , Citometría de Flujo , Humanos , Microscopía Confocal
15.
Stem Cells ; 30(7): 1414-23, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22628289

RESUMEN

Phosphorylation of histone H2AX (γH2AX) is known to be the earliest indicator of DNA double-strand breaks. Recently, it has been shown that mouse embryonic stem cells (mESCs) have very high basal levels of γH2AX, even when they have not been exposed to genotoxic agents. As the specialized role of high basal γH2AX levels in pluripotent stem cells is still debated, we investigated whether H2AX phosphorylation is important in maintaining self-renewal of these cells. Here, we report that not only mESCs but also mouse-induced pluripotent stem cells (miPSCs), have high basal levels of γH2AX. We show that basal γH2AX levels decrease upon ESC and iPSC differentiation and increase when the cells are treated with self-renewal-enhancing small molecules. We observe that self-renewal activity is highly compromised in H2AX-/- cells and that it can be restored in these cells through reconstitution with a wild-type, but not a phospho-mutated, H2AX construct. Taken together, our findings suggest a novel function of H2AX that expands the knowledge of this histone variant beyond its role in DNA damage and into a new specialized biological function in mouse pluripotent stem cells.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Histonas/metabolismo , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Animales , Daño del ADN/genética , Citometría de Flujo , Histonas/genética , Ratones , Microscopía Confocal , Fosforilación
16.
PLoS One ; 7(1): e30234, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22291922

RESUMEN

The ability to reprogram somatic cells to induced pluripotent stem cells (iPSCs), exhibiting properties similar to those of embryonic stem cells (ESCs), has attracted much attention, with many studies focused on improving efficiency of derivation and unraveling the mechanisms of reprogramming. Despite this widespread interest, our knowledge of the molecular signaling pathways that are active in iPSCs and that play a role in controlling their fate have not been studied in detail. To address this shortfall, we have characterized the influence of different signals on the behavior of a model mouse iPSC line. We demonstrate significant responses of this iPSC line to the presence of serum, which leads to profoundly enhanced proliferation and, depending on the medium used, a reduction in the capacity of the iPSCs to self-renew. Surprisingly, this iPSC line was less sensitive to withdrawal of LIF compared to ESCs, exemplified by maintenance of expression of a Nanog-GFP reporter and enhanced self-renewal in the absence of LIF. While inhibition of phosphoinositide-3 kinase (PI3K) signaling decreased iPSC self-renewal, inhibition of Gsk-3 promoted it, even in the absence of LIF. High passages of this iPSC line displayed altered characteristics, including genetic instability and a reduced ability to self-renew. However, this second feature could be restored upon inhibition of Gsk-3. Collectively, our data suggest modulation of Gsk-3 activity plays a key role in the control of iPSC fate. We propose that more careful consideration should be given to characterization of the molecular pathways that control the fate of different iPSC lines, since perturbations from those observed in naïve pluripotent ESCs could render iPSCs and their derivatives susceptible to aberrant and potentially undesirable behaviors.


Asunto(s)
Proliferación Celular , Células Madre Pluripotentes Inducidas/fisiología , Transducción de Señal/fisiología , Animales , Técnicas de Cultivo de Célula , Proliferación Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Células Cultivadas , Reprogramación Celular/efectos de los fármacos , Reprogramación Celular/fisiología , Medios de Cultivo/farmacología , Activación Enzimática/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3/fisiología , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Factor Inhibidor de Leucemia/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factores de Tiempo
17.
DNA Repair (Amst) ; 10(10): 1044-50, 2011 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-21880556

RESUMEN

DNA double-strand breaks (DSB) are the most lethal form of ionizing radiation-induced DNA damage, and failure to repair them results in cell death. In order to see if any associations exist between DNA repair gene polymorphisms and phenotypic profiles of DSB repair (DSBR) we performed a genotype-phenotype correlation study in 118 young healthy subjects (mean age 25.8±6.7years). Subjects were genotyped for 768 single nucleotide polymorphisms (SNPs) with a custom Illumina Golden Gate Assay, and an H2AX histone phosphorylation assay was done to test DSBR capacity. We found that H2AX phosphorylation at 1h was significantly lower in subjects heterozygous (no variant homozygotes were observed) for the XPA gene SNP rs3176683 (p-value=0.005), while dephosphorylation was significantly higher in subjects carrying the variant allele in three MRE11A gene SNPs: rs1014666, rs476137 and rs2508784 (p-value=0.003, 0.003 and 0.008, respectively). An additive effect of low-activity DNA repair alleles was associated with altered DSBR activity, as demonstrated by both H2AX phosphorylation at 1 h (p-trend <0.0001) and γH2AX dephosphorylation at 3h (p-trend <0.0001). Our study revealed that in addition to SNPs of genes that are well-established players in DSBR, non-DSBR genes, such as the XPA gene that is mainly involved in the nucleotide excision repair pathway, can also influence DSBR in healthy subjects. This suggests that successful DSBR may require both DSBR and non-DSBR mechanisms.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN/genética , Proteínas de Unión al ADN/genética , Proteína de la Xerodermia Pigmentosa del Grupo A/genética , Adulto , Alelos , Proteínas de Unión al ADN/metabolismo , Femenino , Estudios de Asociación Genética , Haplotipos , Histonas/química , Humanos , Proteína Homóloga de MRE11 , Masculino , Persona de Mediana Edad , Fosforilación , Polimorfismo de Nucleótido Simple/genética , Radiación Ionizante
18.
Mol Immunol ; 48(12-13): 1369-76, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21481940

RESUMEN

Induction of the TCR signaling pathway terminates the expression of RAG genes, and a link between this pathway and their transcriptional control is evident from the recent demonstration of their re-expression if the TCR is subsequently lost or down-regulated. Since unstimulated T cells display a steady-state level of "tonic" TCR signaling, i.e. in the absence of any antigenic stimulus, it was uncertain whether this control was exerted through ligand-dependent or ligand-independent TCR signaling. Here we demonstrate for the first time that exogenous TCR α and ß chains transferred into the human immature RAG(+) T cell line Sup-T1 by lentiviral transduction inhibit RAG expression through tonic signaling, and that this inhibition could itself be reverted by pharmacological tonic pathway inhibitors. We also suggest that mature T cells already expressing an endogenous TCR on their surface maintain some levels of plasticity at the RAG locus when their basal TCR signaling is interfered with. Lastly, we show that the TCR constructs employed in TCR gene therapy do not possess the same basal signaling transduction capability, a feature that may have therapeutic implications.


Asunto(s)
Proteínas de Unión al ADN/genética , Genes RAG-1 , Proteínas Nucleares/genética , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Transducción de Señal , Linfocitos T/inmunología , Linfocitos T/metabolismo , Benzamidas , Línea Celular , Membrana Celular/inmunología , Membrana Celular/metabolismo , Niño , Proteínas de Unión al ADN/metabolismo , Dimetilsulfóxido/farmacología , Regulación hacia Abajo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Mesilato de Imatinib , Immunoblotting , Células Jurkat , Ligandos , Proteínas Nucleares/metabolismo , Piperazinas/farmacología , Pirimidinas/farmacología , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tacrolimus/farmacología , Transducción Genética
19.
DNA Repair (Amst) ; 9(11): 1200-8, 2010 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-20947454

RESUMEN

We have previously shown that whereas T-cells from normal individuals undergo accumulation of p53 and apoptosis when treated with the genotoxic agent Actinomycin D (ActD), those from Ataxia Telangiectasia (AT) and Nijmegen Breakage Syndrome (NBS) patients resist ActD-induced apoptosis [1]. We have now found similar resistance by the p53-null Jurkat T-cell line and by siRNA p53-knockdown normal T-cells. This evidence that ActD initiates a p53-dependent apoptotic responce prompted us to look for defective p53 accumulation by AT and NBS T-cells. Surprisingly the total p53 level was only slightly reduced compared to normal T cells but its intracellular localization was highly defective: p53 was poorly accumulated in the cytosol and nearly undetectable in mitochondria. In accordance with the dependence of ActD-induced apoptosis on a mitochondrial p53 function, in control T-cells specific inhibition of mitochondrial p53 translocation with µ pifithrin reduced apoptosis by 86%, whereas treatment with α pifithrin, which blocks p53-mediated transcription, had no effect. We also showed that nuclear export is not required for mitochondrial p53 translocation. Observation of an altered p53 ubiquitination pattern and Mdm2 accumulation in ActD-treated AT and NBS T-cells provided a mechanistic link to their defective extranuclear p53 localization. Our results disclose an undescribed defect in mitochondrial p53 accumulation in AT and NBS T-cells that makes them resistant to apoptosis following unrepairable DNA damage.


Asunto(s)
Apoptosis , Ataxia Telangiectasia/patología , Daño del ADN , Mitocondrias/metabolismo , Síndrome de Nijmegen/patología , Linfocitos T/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Apoptosis/efectos de los fármacos , Ataxia Telangiectasia/genética , Ataxia Telangiectasia/metabolismo , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Citoplasma/efectos de los fármacos , Citoplasma/metabolismo , Dactinomicina/toxicidad , Humanos , Células Jurkat , Mitocondrias/efectos de los fármacos , Mitocondrias/genética , Síndrome de Nijmegen/genética , Síndrome de Nijmegen/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Transducción de Señal/efectos de los fármacos , Linfocitos T/citología , Linfocitos T/efectos de los fármacos , Linfocitos T/patología , Ubiquitinación/efectos de los fármacos , Proteína X Asociada a bcl-2/metabolismo
20.
Immunol Lett ; 134(1): 17-25, 2010 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-20709103

RESUMEN

In T lymphocytes, the internalization of the R2 chain of the IFN-γ receptor (IFN-γR2) prevents the switching-on of pro-apoptotic and anti-proliferative genes induced by the IFN-γ/STAT1 pathway. In fibroblasts, a critical role of controlling the IFN-γR2 internalization is played by the LI(255-256) intracellular motif. Here we show that, in human malignant T cells, the expression of a mutated IFN-γR2 chain in which the LI(255-256) internalization motif is replaced by two alanines (LI(255-256)AA) induces cell surface accumulation of the receptor and reinstates the cell sensitivity to IFN-γ. In comparison with T cells that expressed wild-type IFN-γR2, cells that expressed the mutated receptor displayed, in response to IFN-γ a sustained activation of STAT1. The activation of this signaling pathway leads to higher induction of MHC class I and FasL expression and triggered apoptosis. Malignant ST4 cells transduced with either wild-type or mutated receptor were able to grow in SCID mice, but only the proliferation of T cells expressing the mutated receptor was inhibited by IFN-γ. Finally, lentiviral-mediated transduction of the mutated receptor in T lymphoblasts from healthy donors reinstated their IFN-γ-dependent apoptosis. As a whole, these data indicate that perturbation of IFN-γR2 internalization by mutating the LI(255-256) motif induces a timely coordinated activation of IFN-γ/STAT1 signaling pathways that leads to the apoptosis of T cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Interferón gamma/farmacología , Receptores de Interferón/metabolismo , Linfocitos T/efectos de los fármacos , Secuencias de Aminoácidos/genética , Animales , Western Blotting , Línea Celular Tumoral , Dipéptidos/genética , Dipéptidos/metabolismo , Endocitosis/efectos de los fármacos , Proteína Ligando Fas/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Interferón gamma/metabolismo , Células Jurkat , Leucina/genética , Leucina/metabolismo , Linfoma de Células T/metabolismo , Linfoma de Células T/patología , Linfoma de Células T/prevención & control , Ratones , Ratones SCID , Mutación , Fosforilación/efectos de los fármacos , Receptores de Interferón/genética , Factor de Transcripción STAT1/metabolismo , Transducción de Señal , Linfocitos T/metabolismo , Linfocitos T/patología , Ensayos Antitumor por Modelo de Xenoinjerto , Receptor fas/metabolismo , Receptor de Interferón gamma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...