Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Acta Neuropathol Commun ; 8(1): 182, 2020 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-33153497

RESUMEN

Recent discoveries have provided valuable insight into the genomic landscape of pediatric low-grade gliomas (LGGs) at diagnosis, facilitating molecularly targeted treatment. However, little is known about their temporal and therapy-related genomic heterogeneity. An adequate understanding of the evolution of pediatric LGGs' genomic profiles over time is critically important in guiding decisions about targeted therapeutics and diagnostic biopsy at recurrence. Fluorescence in situ hybridization, mutation-specific immunohistochemistry, and/or targeted sequencing were performed on paired tumor samples from primary diagnostic and subsequent surgeries. Ninety-four tumor samples from 45 patients (41 with two specimens, four with three specimens) from three institutions underwent testing. Conservation of BRAF fusion, BRAFV600E mutation, and FGFR1 rearrangement status was observed in 100%, 98%, and 96% of paired specimens, respectively. No loss or gain of IDH1 mutations or NTRK2, MYB, or MYBL1 rearrangements were detected over time. Histologic diagnosis remained the same in all tumors, with no acquired H3K27M mutations or malignant transformation. Changes in CDKN2A deletion status at recurrence occurred in 11 patients (42%), with acquisition of hemizygous CDKN2A deletion in seven and loss in four. Shorter time to progression and shorter time to subsequent surgery were observed among patients with acquired CDKN2A deletions compared to patients without acquisition of this alteration [median time to progression: 5.5 versus 16.0 months (p = 0.048); median time to next surgery: 17.0 months versus 29.0 months (p = 0.031)]. Most targetable genetic aberrations in pediatric LGGs, including BRAF alterations, are conserved at recurrence and following chemotherapy or irradiation. However, changes in CDKN2A deletion status over time were demonstrated. Acquisition of CDKN2A deletion may define a higher risk subgroup of pediatric LGGs with a poorer prognosis. Given the potential for targeted therapies for tumors harboring CDKN2A deletions, biopsy at recurrence may be indicated in certain patients, especially those with rapid progression.


Asunto(s)
Neoplasias Encefálicas/genética , Glioma/genética , Recurrencia Local de Neoplasia/genética , Adolescente , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/terapia , Niño , Preescolar , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Progresión de la Enfermedad , Femenino , Eliminación de Gen , Genoma , Genómica , Glioma/patología , Glioma/terapia , Humanos , Lactante , Isocitrato Deshidrogenasa/genética , Masculino , Glicoproteínas de Membrana/genética , Clasificación del Tumor , Recurrencia Local de Neoplasia/terapia , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas c-myb/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor trkB/genética , Transactivadores/genética
3.
J Biol Chem ; 290(5): 2715-27, 2015 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-25505262

RESUMEN

Functional maintenance of hematopoietic stem cells (HSCs) is constantly challenged by stresses like DNA damage and oxidative stress. Here we show that the Fanconi anemia protein Fancd2 and stress transcriptional factor Foxo3a cooperate to prevent HSC exhaustion in mice. Deletion of both Fancd2 and Foxo3a led to an initial expansion followed by a progressive decline of bone marrow stem and progenitor cells. Limiting dilution transplantation and competitive repopulating experiments demonstrated a dramatic reduction of competitive repopulating units and progressive decline in hematopoietic repopulating ability of double-knockout (dKO) HSCs. Analysis of the transcriptome of dKO HSCs revealed perturbation of multiple pathways implicated in HSC exhaustion. Fancd2 deficiency strongly promoted cytoplasmic localization of Foxo3a in HSCs, and re-expression of Fancd2 completely restored nuclear Foxo3a localization. By co-expressing a constitutively active CA-FOXO3a and WT or a nonubiquitinated Fancd2 in dKO bone marrow stem/progenitor cells, we demonstrated that Fancd2 was required for nuclear retention of CA-FOXO3a and for maintaining hematopoietic repopulation of the HSCs. Collectively, these results implicate a functional interaction between the Fanconi anemia DNA repair and FOXO3a pathways in HSC maintenance.


Asunto(s)
Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Factores de Transcripción Forkhead/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Animales , Ciclo Celular/fisiología , Daño del ADN/fisiología , Reparación del ADN/fisiología , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Fémur/metabolismo , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/genética , Humanos , Ratones , Ratones Noqueados , Estrés Oxidativo/fisiología , Tibia/metabolismo
4.
Antioxid Redox Signal ; 21(12): 1675-92, 2014 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-24483844

RESUMEN

AIMS: This study seeks at investigating the cause of hydrocephalus, and at identifying therapeutic targets for the prevention of hydrocephalus. RESULTS: In this study, we show that inactivation of the Foxo3a gene in two mouse models of Fanconi anemia (FA) leads to the development of hydrocephalus in late embryonic stage and after birth. More than 50% of Foxo3a(-/-) Fancc(-/-) or Foxo3a(-/-) Fancd2(-/-) mice die during embryonic development or within 6 months of life as a result of hydrocephalus characterized by cranial distortion, dilation of the ventricular system, reduced thickness of the cerebral cortex, and disorganization of the ependymal cilia and subcommissural organ. Combined deficiency of Foxo3a and Fancc or Fancd2 not only impairs the self-renewal capacity but also markedly increases the apoptosis of neural stem and progenitor cells (NSPCs), leading to defective neurogenesis. Increased accumulation of reactive oxygen species (ROS) and subsequently de-regulated mitosis and ultimately apoptosis in the neural stem or progenitor cells is identified as one of the potential mechanisms of congenital obstructive hydrocephalus. INNOVATION: The work unravels a two-tier protective mechanism for preventing oxidative stress-induced hydrocephalus. CONCLUSION: The deletion of Foxo3a in FA mice increased the accumulation of ROS and subsequently de-regulated mitosis and ultimately apoptosis in the NSPCs, leading to hydrocephalus development.


Asunto(s)
Proteína del Grupo de Complementación C de la Anemia de Fanconi/metabolismo , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Factores de Transcripción Forkhead/metabolismo , Hidrocefalia/metabolismo , Estrés Oxidativo , Animales , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Proteína del Grupo de Complementación C de la Anemia de Fanconi/deficiencia , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/deficiencia , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/deficiencia , Factores de Transcripción Forkhead/genética , Hidrocefalia/genética , Ratones , Ratones Noqueados , Especies Reactivas de Oxígeno/metabolismo
5.
Blood ; 123(12): 1938-47, 2014 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-24501220

RESUMEN

Fanconi anemia (FA) is a genetic disorder associated with bone marrow (BM) failure and leukemia. Recent studies demonstrate variable immune defects in FA. However, the cause for FA immunodeficiency is unknown. Here we report that deletion of Fanca or Fancd2 dysregulates the suppressive activity of regulatory T cells (Tregs), shown functionally as exacerbation of graft-vs-host disease (GVHD) in mice. Recipient mice of Fanca(-/-) or Fancd2(-/-) BM chimeras exhibited severe acute GVHD after allogeneic BM transplantation (BMT). T cells from Fanca(-/-) or Fancd2(-/-) mice induced higher GVHD lethality than those from wild-type (WT) littermates. FA Tregs possessed lower proliferative suppression potential compared with WT Tregs, as demonstrated by in vitro proliferation assay and BMT. Analysis of CD25(+)Foxp3(+) Tregs indicated that loss of Fanca or Fancd2 dysregulated Foxp3 target gene expression. Additionally, CD25(+)Foxp3(+) Tregs of Fanca(-/-) or Fancd2(-/-) mice were less efficient in suppressing the production of GVHD-associated inflammatory cytokines. Consistently, aberrant NF-κB activity was observed in infiltrated T cells from FA GVHD mice. Conditional deletion of p65 in FA Tregs decreased GVHD mortality. Our study uncovers an essential role for FA proteins in maintaining Treg homeostasis, possibly explaining, at least in part, the immune deficiency reported in some FA patients.


Asunto(s)
Proteína del Grupo de Complementación A de la Anemia de Fanconi/deficiencia , Proteína del Grupo de Complementación A de la Anemia de Fanconi/inmunología , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/deficiencia , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/inmunología , Linfocitos T Reguladores/inmunología , Animales , Trasplante de Médula Ósea , Citocinas/biosíntesis , Anemia de Fanconi/genética , Anemia de Fanconi/inmunología , Anemia de Fanconi/metabolismo , Proteína del Grupo de Complementación A de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Factores de Transcripción Forkhead/metabolismo , Expresión Génica , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/patología , Humanos , Tolerancia Inmunológica , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Linfocitos T Reguladores/metabolismo , Quimera por Trasplante
6.
J Immunol ; 191(5): 2806-17, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23926327

RESUMEN

Hematopoietic stem cells (HSCs) can either self-renew or differentiate into various types of cells of the blood lineage. Signaling pathways that regulate this choice of self-renewal versus differentiation are currently under extensive investigation. In this study, we report that deregulation of Notch signaling skews HSC differentiation in mouse models of Fanconi anemia (FA), a genetic disorder associated with bone marrow failure and progression to leukemia and other cancers. In mice expressing a transgenic Notch reporter, deletion of the Fanca or Fancc gene enhances Notch signaling in multipotential progenitors (MPPs), which is correlated with decreased phenotypic long-term HSCs and increased formation of MPP1 progenitors. Furthermore, we found an inverse correlation between Notch signaling and self-renewal capacity in FA hematopoietic stem and progenitor cells. Significantly, FA deficiency in MPPs deregulates a complex network of genes in the Notch and canonical NF-κB pathways. Genetic ablation or pharmacologic inhibition of NF-κB reduces Notch signaling in FA MPPs to near wild type level, and blocking either NF-κB or Notch signaling partially restores FA HSC quiescence and self-renewal capacity. These results suggest a functional crosstalk between Notch signaling and NF-κB pathway in regulation of HSC differentiation.


Asunto(s)
Diferenciación Celular/fisiología , Anemia de Fanconi/metabolismo , Células Madre Hematopoyéticas/metabolismo , FN-kappa B/metabolismo , Receptores Notch/metabolismo , Transducción de Señal/fisiología , Animales , Modelos Animales de Enfermedad , Citometría de Flujo , Immunoblotting , Inflamación/metabolismo , Ratones , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor Cross-Talk/fisiología
7.
Antioxid Redox Signal ; 17(8): 1083-98, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22482891

RESUMEN

AIMS: Insulin resistance is a hallmark of obesity and type 2 diabetes. Reactive oxygen species (ROS) have been proposed to play a causal role in insulin resistance. However, evidence linking ROS to insulin resistance in disease settings has been scant. Since both oxidative stress and diabetes have been observed in patients with the Fanconi anemia (FA), we sought to investigate the link between ROS and insulin resistance in this unique disease model. RESULTS: Mice deficient for the Fanconi anemia complementation group A (Fanca) or Fanconi anemia complementation group C (Fancc) gene seem to be diabetes-prone, as manifested by significant hyperglycemia and hyperinsulinemia, and rapid weight gain when fed with a high-fat diet. These phenotypic features of insulin resistance are characterized by two critical events in insulin signaling: a reduction in tyrosine phosphorylation of the insulin receptor (IR) and an increase in inhibitory serine phosphorylation of the IR substrate-1 in the liver, muscle, and fat tissues from the insulin-challenged FA mice. High levels of ROS, spontaneously accumulated or generated by tumor necrosis factor alpha in these insulin-sensitive tissues of FA mice, were shown to underlie the FA insulin resistance. Treatment of FA mice with the natural anti-oxidant Quercetin restores IR signaling and ameliorates the diabetes- and obesity-prone phenotypes. Finally, pairwise screen identifies protein-tyrosine phosphatase (PTP)-α and stress kinase double-stranded RNA-dependent protein kinase (PKR) that mediate the ROS effect on FA insulin resistance. INNOVATION: These findings establish a pathogenic and mechanistic link between ROS and insulin resistance in a unique human disease setting. CONCLUSION: ROS accumulation contributes to the insulin resistance in FA deficiency by targeting both PTP-α and PKR.


Asunto(s)
Anemia de Fanconi/metabolismo , Resistencia a la Insulina , Obesidad/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Células 3T3-L1 , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Proteína del Grupo de Complementación A de la Anemia de Fanconi/deficiencia , Proteína del Grupo de Complementación A de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación A de la Anemia de Fanconi/metabolismo , Proteína del Grupo de Complementación C de la Anemia de Fanconi/deficiencia , Proteína del Grupo de Complementación C de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación C de la Anemia de Fanconi/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/genética , Estrés Oxidativo , Quercetina/administración & dosificación , Quercetina/farmacología , Receptor de Insulina/metabolismo , Transducción de Señal/efectos de los fármacos
8.
Blood ; 119(18): 4142-51, 2012 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-22408259

RESUMEN

Oxidative stress has been implicated in the pathogenesis of many human diseases including Fanconi anemia (FA), a genetic disorder associated with BM failure and cancer. Here we show that major antioxidant defense genes are down-regulated in FA patients, and that gene down-regulation is selectively associated with increased oxidative DNA damage in the promoters of the antioxidant defense genes. Assessment of promoter activity and DNA damage repair kinetics shows that increased initial damage, rather than a reduced repair rate, contributes to the augmented oxidative DNA damage. Mechanistically, FA proteins act in concert with the chromatin-remodeling factor BRG1 to protect the promoters of antioxidant defense genes from oxidative damage. Specifically, BRG1 binds to the promoters of the antioxidant defense genes at steady state. On challenge with oxidative stress, FA proteins are recruited to promoter DNA, which correlates with significant increase in the binding of BRG1 within promoter regions. In addition, oxidative stress-induced FANCD2 ubiquitination is required for the formation of a FA-BRG1-promoter complex. Taken together, these data identify a role for the FA pathway in cellular antioxidant defense.


Asunto(s)
Proteínas del Grupo de Complementación de la Anemia de Fanconi/fisiología , Anemia de Fanconi/metabolismo , Regulación de la Expresión Génica , Estrés Oxidativo/genética , Regiones Promotoras Genéticas/genética , 8-Hidroxi-2'-Desoxicoguanosina , Antioxidantes , Células de la Médula Ósea/metabolismo , Células Cultivadas/efectos de los fármacos , Daño del ADN , ADN Helicasas/metabolismo , Reparación del ADN , Desoxiguanosina/análogos & derivados , Desoxiguanosina/análisis , Regulación hacia Abajo , Anemia de Fanconi/genética , Anemia de Fanconi/patología , Proteínas del Grupo de Complementación de la Anemia de Fanconi/deficiencia , Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética , Humanos , Peróxido de Hidrógeno/toxicidad , Complejos Multiproteicos , Proteínas Nucleares/metabolismo , Oxidación-Reducción , Unión Proteica , Procesamiento Proteico-Postraduccional , Factores de Transcripción/metabolismo , Ubiquitinación
9.
Blood ; 119(18): 4162-73, 2012 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-22427203

RESUMEN

Salidroside is a phenylpropanoid glycoside isolated from the medicinal plant Rhodiola rosea, which has potent antioxidant properties. Here we show that salidroside prevented the loss of hematopoietic stem cells (HSCs) in mice under oxidative stress. Quiescent HSCs were recruited into cell cycling on in vivo challenge with oxidative stress, which was blocked by salidroside. Surprisingly, salidroside does not prevent the production of reactive oxygen species but reduces hydrogen peroxide-induced DNA-strand breaks in bone marrow cells enriched for HSCs. We tested whether salidroside enhances oxidative DNA damage repair in mice deficient for 5 DNA repair pathways known to be involved in oxidative DNA damage repair; we found that salidroside activated poly(ADP-ribose)polymerase-1 (PARP-1), a component of the base excision repair pathway, in mouse bone marrow HSCs as well as primary fibroblasts and human lymphoblasts. PARP-1 activation by salidroside protects quiescent HSCs from oxidative stress-induced cycling in native animals and self-renewal defect in transplanted recipients, which was abrogated by genetic ablation or pharmacologic inhibition of PARP-1. Together, these findings suggest that activation of PARP-1 by salidroside could affect the homeostasis and function of HSCs and contribute to the antioxidant effects of salidroside.


Asunto(s)
Antioxidantes/farmacología , Reparación del ADN/efectos de los fármacos , Glucósidos/farmacología , Células Madre Hematopoyéticas/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Fenoles/farmacología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Animales , Ciclo Celular/efectos de los fármacos , ADN/efectos de los fármacos , Roturas del ADN de Doble Cadena , Evaluación Preclínica de Medicamentos , Proteína del Grupo de Complementación A de la Anemia de Fanconi/deficiencia , Proteína del Grupo de Complementación C de la Anemia de Fanconi/deficiencia , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Peróxido de Hidrógeno/toxicidad , Ratones , Ratones Endogámicos C57BL , Oxidantes/toxicidad , Plantas Medicinales/química , Poli(ADP-Ribosa) Polimerasa-1 , Especies Reactivas de Oxígeno , Rhodiola/química
10.
Blood ; 117(16): 4243-52, 2011 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-21330473

RESUMEN

Patients with Fanconi anemia (FA) have a high risk of developing acute myeloid leukemia (AML). In this study, we attempted to identify cell-surface markers for leukemia-initiating cells in FA-AML patients. We found that the IL-3 receptor-α (IL-3Rα) is a promising candidate as an leukemia-initiating cell-specific antigen for FA-AML. Whereas IL-3Rα expression is undetectable on normal CD34(+)CD38(-) HSCs, it is overexpressed on CD34(+)CD38(-) cells from FA patients with AML. We examined the leukemia-initiating cell activity of IL-3Rα-positive FA-AML cells in a "humanized" FA xenotransplant model in which we separated AML cells into IL-3Rα-positive and IL-3Rα-negative CD34 fractions and transplanted them into irradiated recipient mice. In all 3 FA-AML samples, only IL-3Rα-positive cells showed significant levels of engraftment and developed leukemia in the recipient mice. The FA CD34(+)IL-3Rα(+) blasts isolated from leukemic mice exhibited hypersensitivity to IL-3 deprivation and JAK2-STAT5 overactivation after IL-3 treatment. Finally, treatment of FA CD34(+)IL-3Rα(+) blasts with an IL-3Rα-neutralizing antibody inhibited IL-3-mediated proliferation and STAT5 activation. These results demonstrate that IL-3Rα is a cell-surface marker present on FA-AML leukemia-initiating cells and may be a valuable therapeutic target.


Asunto(s)
ADP-Ribosil Ciclasa 1/inmunología , Antígenos CD34/inmunología , Anemia de Fanconi/complicaciones , Subunidad alfa del Receptor de Interleucina-3/genética , Leucemia Mieloide Aguda/inmunología , Células Madre/inmunología , Regulación hacia Arriba , Animales , Anemia de Fanconi/inmunología , Humanos , Subunidad alfa del Receptor de Interleucina-3/antagonistas & inhibidores , Subunidad alfa del Receptor de Interleucina-3/inmunología , Janus Quinasa 2/inmunología , Leucemia Mieloide Aguda/etiología , Ratones , Ratones Endogámicos NOD , Factor de Transcripción STAT5/inmunología , Trasplante Heterólogo
11.
J Biol Chem ; 285(48): 37436-44, 2010 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-20864535

RESUMEN

Eight of the Fanconi anemia (FA) proteins form a core complex that activates the FA pathway. Some core complex components also form subcomplexes for yet-to-be-elucidated functions. Here, we have analyzed the interaction between a cytoplasmic FA subcomplex and the leukemic nucleophosmin (NPMc). Exogenous NPMc was degraded rapidly in FA acute myeloid leukemia bone marrow cells. Knockdown of FANCA or FANCC in leukemic OCI/AML3 cells induced rapid degradation of endogenous NPMc. NPMc degradation was mediated by the ubiquitin-proteasome pathway involving the IBR-type RING-finger E3 ubiquitin ligase IBRDC2, and genetic correction of FA-A or FA-C lymphoblasts prevented NPMc ubiquitination. Moreover, cytoplasmic FANCA and FANCC formed a cytoplasmic complex and interacted with NPMc. Using a patient-derived FANCC mutant and a nuclearized FANCC, we demonstrated that the cytoplasmic FANCA-FANCC complex was essential for NPMc stability. Finally, depletion of FANCA and FANCC in NPMc-positive leukemic cells significantly increased inflammation and chemoresistance through NF-κB activation. Our findings not only reveal the molecular mechanism involving cytoplasmic retention of NPMc but also suggest cytoplasmic function of FANCA and FANCC in NPMc-related leukemogenesis.


Asunto(s)
Citoplasma/metabolismo , Proteína del Grupo de Complementación A de la Anemia de Fanconi/metabolismo , Proteína del Grupo de Complementación C de la Anemia de Fanconi/metabolismo , Leucemia/metabolismo , Secuencia de Bases , Línea Celular Tumoral , Citoplasma/química , Citoplasma/genética , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Proteína del Grupo de Complementación A de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación C de la Anemia de Fanconi/genética , Humanos , Leucemia/genética , Datos de Secuencia Molecular , Proteínas Nucleares , Nucleofosmina , Unión Proteica , Estabilidad Proteica , Transporte de Proteínas , Ubiquitinación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...