Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 141(15): 1846-1857, 2023 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-36508705

RESUMEN

NPM 1-mutated acute myeloid leukemia (AML) shows unique features. However, the characteristics of "therapy-related" NPM1-mutated AML (t-NPM1 AML) are poorly understood. We compared the genetics, transcriptional profile, and clinical outcomes of t-NPM1 AML, de novo NPM1-mutated AML (dn-NPM1 AML), and therapy-related AML (t-AML) with wild-type NPM1 (t-AML). Normal karyotype was more frequent in t-NPM1 AML (n = 78/96, 88%) and dn-NPM1 (n = 1986/2394, 88%) than in t-AML (n = 103/390, 28%; P < .001). DNMT3A and TET2 were mutated in 43% and 40% of t-NPM1 AML (n = 107), similar to dn-NPM1 (n = 88, 48% and 30%; P > 0.1), but more frequently than t-AML (n = 162; 14% and 10%; P < 0.001). Often mutated in t-AML, TP53 and PPM1D were wild-type in 97% and 96% of t-NPM1 AML, respectively. t-NPM1 and dn-NPM1 AML were transcriptionally similar, (including HOX genes upregulation). At 62 months of median follow-up, the 3-year overall survival (OS) for t-NPM1 AML (n = 96), dn-NPM1 AML (n = 2394), and t-AML (n = 390) were 54%, 60%, and 31%, respectively. In multivariable analysis, OS was similar for the NPM1-mutated groups (hazard ratio [HR] 0.9; 95% confidence interval [CI], 0.65-1.25; P = .45), but better in t-NPM1 AML than in t-AML (HR, 1.86; 95% CI, 1.30-2.68; P < .001). Relapse-free survival was similar between t-NPM1 and dn-NPM1 AML (HR, 1.02; 95% CI, 0.72-1.467; P = .90), but significantly higher in t-NPM1 AML versus t-AML (HR, 1.77; 95% CI, 1.19-2.64; P = .0045). t-NPM1 and dn-NPM1 AML have overlapping features, suggesting that they should be classified as a single disease entity.


Asunto(s)
Leucemia Mieloide Aguda , Proteínas Nucleares , Humanos , Proteínas Nucleares/genética , Nucleofosmina , Mutación , Pronóstico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/terapia
2.
EMBO J ; 38(12)2019 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-31126958

RESUMEN

Autophagy and energy metabolism are known to follow a circadian pattern. However, it is unclear whether autophagy and the circadian clock are coordinated by common control mechanisms. Here, we show that the oscillation of autophagy genes is dependent on the nutrient-sensitive activation of TFEB and TFE3, key regulators of autophagy, lysosomal biogenesis, and cell homeostasis. TFEB and TFE3 display a circadian activation over the 24-h cycle and are responsible for the rhythmic induction of genes involved in autophagy during the light phase. Genetic ablation of TFEB and TFE3 in mice results in deregulated autophagy over the diurnal cycle and altered gene expression causing abnormal circadian wheel-running behavior. In addition, TFEB and TFE3 directly regulate the expression of Rev-erbα (Nr1d1), a transcriptional repressor component of the core clock machinery also involved in the regulation of whole-body metabolism and autophagy. Comparative analysis of the cistromes of TFEB/TFE3 and REV-ERBα showed an extensive overlap of their binding sites, particularly in genes involved in autophagy and metabolic functions. These data reveal a direct link between nutrient and clock-dependent regulation of gene expression shedding a new light on the crosstalk between autophagy, metabolism, and circadian cycles.


Asunto(s)
Autofagia , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/fisiología , Relojes Circadianos , Metabolismo Energético , Nutrientes/fisiología , Animales , Autofagia/efectos de los fármacos , Autofagia/genética , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/efectos de los fármacos , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Sitios de Unión , Células Cultivadas , Relojes Circadianos/efectos de los fármacos , Relojes Circadianos/genética , Ritmo Circadiano/efectos de los fármacos , Ritmo Circadiano/fisiología , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/genética , Regulación de la Expresión Génica , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/fisiología , Nutrientes/farmacología , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/genética , Factores de Transcripción/fisiología
3.
Blood ; 137(5): 589-599, 2021 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-33171486

RESUMEN

Mutations of the nucleophosmin (NPM1) gene, encoding for a nucleolar multifunctional protein, occur in approximately one-third of adult acute myeloid leukemia (AML). NPM1-mutated AML exhibits unique molecular, pathological, and clinical features, which led to its recognition as distinct entity in the 2017 World Health Organization (WHO) classification of myeloid neoplasms. Although WHO criteria for the diagnosis of NPM1-mutated AML are well established, its distinction from other AML entities may be difficult. Moreover, the percentage of blasts required to diagnose NPM1-mutated AML remains controversial. According to the European LeukemiaNet (ELN), determining the mutational status of NPM1 (together with FLT3) is mandatory for accurate relapse-risk assessment. NPM1 mutations are ideal targets for measurable residual disease (MRD) monitoring, since they are AML specific, frequent, very stable at relapse, and do not drive clonal hematopoiesis of undetermined significance. MRD monitoring by quantitative polymerase chain reaction of NPM1-mutant transcripts, possibly combined with ELN genetic-based risk stratification, can guide therapeutic decisions after remission. Furthermore, immunohistochemistry can be very useful in selected situations, such as diagnosis of NPM1-mutated myeloid sarcoma. Herein, we present 4 illustrative cases of NPM1-mutated AML that address important issues surrounding the biology, diagnosis, and therapy of this common form of leukemia.


Asunto(s)
Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/terapia , Proteínas Nucleares/genética , Proteínas de Fusión Oncogénica/genética , Pautas de la Práctica en Medicina , Factores de Edad , Anciano , Algoritmos , Aloinjertos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Médula Ósea/patología , Compuestos Bicíclicos Heterocíclicos con Puentes/administración & dosificación , Linaje de la Célula , Ensayos Clínicos como Asunto , Evolución Clonal , Terapia Combinada , Diagnóstico Diferencial , Manejo de la Enfermedad , Femenino , Gemtuzumab/administración & dosificación , Trasplante de Células Madre Hematopoyéticas , Humanos , Leucemia Mieloide Aguda/clasificación , Leucemia Mieloide Aguda/genética , Masculino , Persona de Mediana Edad , Terapia Molecular Dirigida , Síndromes Mielodisplásicos/inducido químicamente , Síndromes Mielodisplásicos/diagnóstico , Células Madre Neoplásicas/patología , Proteínas Nucleares/antagonistas & inhibidores , Nucleofosmina , Proteínas de Fusión Oncogénica/antagonistas & inhibidores , Selección de Paciente , Inducción de Remisión , Medición de Riesgo , Terapia Recuperativa , Sulfonamidas/administración & dosificación , Tirosina Quinasa 3 Similar a fms/genética
4.
Blood ; 138(25): 2696-2701, 2021 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-34343258

RESUMEN

Nucleophosmin (NPM1) mutations in acute myeloid leukemia (AML) affect exon 12, but also sporadically affect exons 9 and 11, causing changes at the protein C-terminal end (tryptophan loss, nuclear export signal [NES] motif creation) that lead to aberrant cytoplasmic NPM1 (NPM1c+), detectable by immunohistochemistry. Combining immunohistochemistry and molecular analyses in 929 patients with AML, we found non-exon 12 NPM1 mutations in 5 (1.3%) of 387 NPM1c+ cases. Besides mutations in exons 9 (n = 1) and 11 (n = 1), novel exon 5 mutations were discovered (n = 3). Another exon 5 mutation was identified in an additional 141 patients with AML selected for wild-type NPM1 exon 12. Three NPM1 rearrangements (NPM1/RPP30, NPM1/SETBP1, NPM1/CCDC28A) were detected and characterized among 13 979 AML samples screened by cytogenetic/fluorescence in situ hybridization and RNA sequencing. Functional studies demonstrated that in AML cases, new NPM1 proteins harbored an efficient extra NES, either newly created or already present in the fusion partner, ensuring its cytoplasmic accumulation. Our findings support NPM1 cytoplasmic relocation as critical for leukemogenesis and reinforce the role of immunohistochemistry in predicting AML-associated NPM1 genetic lesions. This study highlights the need to develop new assays for molecular diagnosis and monitoring of NPM1-mutated AML.


Asunto(s)
Leucemia Mieloide Aguda/genética , Mutación , Nucleofosmina/genética , Adulto , Exones , Femenino , Fusión Génica , Reordenamiento Génico , Humanos , Masculino , Persona de Mediana Edad
5.
Blood ; 137(5): 624-636, 2021 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-32902645

RESUMEN

Immune checkpoint therapy has resulted in remarkable improvements in the outcome for certain cancers. To broaden the clinical impact of checkpoint targeting, we devised a strategy that couples targeting of the cytokine-inducible Src homology 2-containing (CIS) protein, a key negative regulator of interleukin 15 (IL-15) signaling, with fourth-generation "armored" chimeric antigen receptor (CAR) engineering of cord blood-derived natural killer (NK) cells. This combined strategy boosted NK cell effector function through enhancing the Akt/mTORC1 axis and c-MYC signaling, resulting in increased aerobic glycolysis. When tested in a lymphoma mouse model, this combined approach improved NK cell antitumor activity more than either alteration alone, eradicating lymphoma xenografts without signs of any measurable toxicity. We conclude that targeting a cytokine checkpoint further enhances the antitumor activity of IL-15-secreting armored CAR-NK cells by promoting their metabolic fitness and antitumor activity. This combined approach represents a promising milestone in the development of the next generation of NK cells for cancer immunotherapy.


Asunto(s)
Sangre Fetal/citología , Inmunoterapia Adoptiva , Interleucina-15/genética , Células Asesinas Naturales/efectos de los fármacos , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas Supresoras de la Señalización de Citocinas/antagonistas & inhibidores , Aerobiosis , Animales , Antígenos CD19/inmunología , Linfoma de Burkitt/patología , Linfoma de Burkitt/terapia , Sistemas CRISPR-Cas , Línea Celular Tumoral , Técnicas de Inactivación de Genes , Glucólisis , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Interleucina-15/metabolismo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/trasplante , Diana Mecanicista del Complejo 1 de la Rapamicina/fisiología , Ratones , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Receptores Quiméricos de Antígenos , Transducción de Señal/fisiología , Proteínas Supresoras de la Señalización de Citocinas/genética , Proteínas Supresoras de la Señalización de Citocinas/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Blood ; 136(15): 1707-1721, 2020 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-32609823

RESUMEN

The nucleophosmin (NPM1) gene encodes for a multifunctional protein with prominent nucleolar localization that shuttles between nucleus and cytoplasm. NPM1 mutations represent the most common genetic lesion in adult acute myeloid leukemia (AML; about one third of cases), and they act deterministically to cause the aberrant cytoplasmic delocalization of NPM1 mutants. Because of its unique features, NPM1-mutated AML is recognized as a distinct entity in the 2017 World Health Organization (WHO) classification of hematopoietic neoplasms. Here, we focus on recently identified functions of wild-type NPM1 in the nucleolus and address new biological and clinical issues related to NPM1-mutated AML. The relevance of the cooperation between NPM1 and other mutations in driving AML with different outcomes is presented. We also discuss the importance of eradicating NPM1-mutated clones to achieve AML cure and the impact of preleukemic clonal hematopoiesis persistence in predisposing to second AML. The contribution of HOX genes' expression to the development of NPM1-mutated AML is also highlighted. Clinically, yet unsolved diagnostic issues in the 2017 WHO classification of myeloid neoplasms and the importance of NPM1 mutations in defining the framework of European LeukemiaNet genetic-based risk stratification are discussed. Finally, we address the value and limits of NPM1-based measurable residual disease assessment for treatment guidance and present the results of promising preclinical studies with XPO1 and menin-MLL inhibitors.


Asunto(s)
Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Leucemia Mieloide Aguda/genética , Mutación , Proteínas Nucleares/genética , Animales , Transformación Celular Neoplásica/genética , Hematopoyesis Clonal/genética , Manejo de la Enfermedad , Humanos , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/terapia , Nucleofosmina , Lesiones Precancerosas/genética , Lesiones Precancerosas/metabolismo , Pronóstico , Investigación Biomédica Traslacional
7.
Haematologica ; 107(4): 887-898, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-34092059

RESUMEN

Tatton-Brown-Rahman syndrome (TBRS) is an overgrowth disorder caused by germline heterozygous mutations in the DNA methyltransferase DNMT3A. DNMT3A is a critical regulator of hematopoietic stem cell (HSC) differentiation and somatic DNMT3A mutations are frequent in hematologic malignancies and clonal hematopoiesis. Yet, the impact of constitutive DNMT3A mutation on hematopoiesis in TBRS is undefined. In order to establish how constitutive mutation of DNMT3A impacts blood development in TBRS we gathered clinical data and analyzed blood parameters in 18 individuals with TBRS. We also determined the distribution of major peripheral blood cell lineages by flow cytometric analyses. Our analyses revealed non-anemic macrocytosis, a relative decrease in lymphocytes and increase in neutrophils in TBRS individuals compared to unaffected controls. We were able to recapitulate these hematologic phenotypes in multiple murine models of TBRS and identified rare hematological and non-hematological malignancies associated with constitutive Dnmt3a mutation. We further show that loss of DNMT3A in TBRS is associated with an altered DNA methylation landscape in hematopoietic cells affecting regions critical to stem cell function and tumorigenesis. Overall, our data identify key hematopoietic effects driven by DNMT3A mutation with clinical implications for individuals with TBRS and DNMT3A-associated clonal hematopoiesis or malignancies.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas , Discapacidad Intelectual , Animales , ADN (Citosina-5-)-Metiltransferasas/genética , ADN Metiltransferasa 3A , Células Germinativas/patología , Hematopoyesis/genética , Humanos , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Ratones
8.
Proc Natl Acad Sci U S A ; 115(7): E1511-E1519, 2018 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-29382756

RESUMEN

Capicua (CIC) regulates a transcriptional network downstream of the RAS/MAPK signaling cascade. In Drosophila, CIC is important for many developmental processes, including embryonic patterning and specification of wing veins. In humans, CIC has been implicated in neurological diseases, including spinocerebellar ataxia type 1 (SCA1) and a neurodevelopmental syndrome. Additionally, we and others have reported mutations in CIC in several cancers. However, whether CIC is a tumor suppressor remains to be formally tested. In this study, we found that deletion of Cic in adult mice causes T cell acute lymphoblastic leukemia/lymphoma (T-ALL). Using hematopoietic-specific deletion and bone marrow transplantation studies, we show that loss of Cic from hematopoietic cells is sufficient to drive T-ALL. Cic-null tumors show up-regulation of the KRAS pathway as well as activation of the NOTCH1 and MYC transcriptional programs. In sum, we demonstrate that loss of CIC causes T-ALL, establishing it as a tumor suppressor for lymphoid malignancies. Moreover, we show that mouse models lacking CIC in the hematopoietic system are robust models for studying the role of RAS signaling as well as NOTCH1 and MYC transcriptional programs in T-ALL.


Asunto(s)
Diferenciación Celular , Susceptibilidad a Enfermedades , Leucemia-Linfoma Linfoblástico de Células T Precursoras/etiología , Proteínas Represoras/fisiología , Linfocitos T/patología , Animales , Células Cultivadas , Ratones , Ratones Noqueados , Mutación , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Receptor Notch1/genética , Receptor Notch1/metabolismo , Transducción de Señal , Linfocitos T/metabolismo , Proteínas ras/genética , Proteínas ras/metabolismo
12.
Blood ; 125(22): 3455-65, 2015 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-25795919

RESUMEN

Nucleophosmin (NPM1) mutations represent an attractive therapeutic target in acute myeloid leukemia (AML) because they are common (∼30% AML), stable, and behave as a founder genetic lesion. Oncoprotein targeting can be a successful strategy to treat AML, as proved in acute promyelocytic leukemia by treatment with all-trans retinoic acid (ATRA) plus arsenic trioxide (ATO), which degrade the promyelocytic leukemia (PML)-retinoic acid receptor fusion protein. Adjunct of ATRA to chemotherapy was reported to be beneficial for NPM1-mutated AML patients. Leukemic cells with NPM1 mutation also showed sensibility to ATO in vitro. Here, we explore the mechanisms underlying these observations and show that ATO/ATRA induce proteasome-dependent degradation of NPM1 leukemic protein and apoptosis in NPM1-mutated AML cell lines and primary patients' cells. We also show that PML intracellular distribution is altered in NPM1-mutated AML cells and reverted by arsenic through oxidative stress induction. Interestingly, similarly to what was described for PML, oxidative stress also mediates ATO-induced degradation of the NPM1 mutant oncoprotein. Strikingly, NPM1 mutant downregulation by ATO/ATRA was shown to potentiate response to the anthracyclin daunorubicin. These findings provide experimental evidence for further exploring ATO/ATRA in preclinical NPM1-mutated AML in vivo models and a rationale for exploiting these compounds in chemotherapeutic regimens in clinics.


Asunto(s)
Apoptosis/efectos de los fármacos , Arsenicales/farmacología , Leucemia Mieloide Aguda/metabolismo , Proteínas Nucleares/metabolismo , Óxidos/farmacología , Tretinoina/farmacología , Animales , Apoptosis/genética , Trióxido de Arsénico , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Ratones , Ratones SCID , Proteínas Mutantes/efectos de los fármacos , Proteínas Mutantes/metabolismo , Mutación , Proteínas Nucleares/efectos de los fármacos , Nucleofosmina , Proteínas Oncogénicas/efectos de los fármacos , Proteínas Oncogénicas/metabolismo , Células Tumorales Cultivadas , Células U937 , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Br J Haematol ; 170(3): 305-22, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25891481

RESUMEN

The acute myeloid leukaemia (AML) genome contains more than 20 driver recurrent mutations. Here, we review the potential for therapeutic targeting of the most common mutations associated with normal cytogenetics AML, focusing on those affecting the FLT3, NPM1 and epigenetic modifier genes (DNMT3A, IDH1/2, TET2). As compared to early compounds, second generation FLT3 inhibitors are more specific and have better pharmacokinetics. They also show higher anti-leukaemic activity, leading to about 50% of composite complete remissions in refractory/relapsed FLT3-internal tandem duplication-mutated AML. However, rapid relapses invariably occur due to various mechanisms of resistance to FLT3 inhibitors. This issue and the best way for using FLT3 inhibitors in combination with other therapeutic modalities are discussed. Potential approaches for therapeutic targeting of NPM1-mutated AML include: (i) reverting the aberrant nuclear export of NPM1 mutant using exportin-1 inhibitors; (ii) disruption of the nucleolus with drugs blocking the oligomerization of wild-type nucleophosmin or inducing nucleolar stress; and (iii) immunotherapeutic targeting of highly expressed CD33 and IL3RA (CD123) antigens. Finally, we discuss the role of demethylating agents (decitabine and azacitidine) and IDH1/2 inhibitors in the treatment of AML patients carrying mutations of genes (DNMT3A, IDH1/2 and TET2) involved in the epigenetic regulation of transcription.


Asunto(s)
Antineoplásicos/uso terapéutico , Sistemas de Liberación de Medicamentos/métodos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Animales , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN Metiltransferasa 3A , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Dioxigenasas , Epigénesis Genética/efectos de los fármacos , Epigénesis Genética/genética , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Regulación Leucémica de la Expresión Génica/genética , Humanos , Subunidad alfa del Receptor de Interleucina-3/biosíntesis , Subunidad alfa del Receptor de Interleucina-3/genética , Isocitrato Deshidrogenasa/antagonistas & inhibidores , Isocitrato Deshidrogenasa/genética , Isocitrato Deshidrogenasa/metabolismo , Leucemia Mieloide Aguda/metabolismo , Mutación , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Nucleofosmina , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Lectina 3 Similar a Ig de Unión al Ácido Siálico/biosíntesis , Lectina 3 Similar a Ig de Unión al Ácido Siálico/genética , Transcripción Genética/efectos de los fármacos , Transcripción Genética/genética , Tirosina Quinasa 3 Similar a fms/antagonistas & inhibidores , Tirosina Quinasa 3 Similar a fms/genética , Tirosina Quinasa 3 Similar a fms/metabolismo
18.
Prof Inferm ; 66(2): 109-16, 2013.
Artículo en Italiano | MEDLINE | ID: mdl-23900147

RESUMEN

Workplace violence is a widespread phenomenon in every kind of settings. Among these ones there are emergency departments (ED), that have distinctive features as like the large daily number of patients' presentations, and high emotional content or stressing situations related to the management of diagnostic-therapeutic priorities. We reviewed the medical and nursing literature to quantify the international widespread of aggressions towards nurses working in EDs, distinguish the typologies and the perpetrators, and identify the consequences on victims and healthcare organizations. Original papers were searched using Medline, CINHAL, and Medscape databases.  35 research articles met the inclusion criteria, but 6 were not retrieved. The rate of verbal abuses reported by ED nurses varies from 50% to 100% of those who were surveyed, while physical violence ranges between 16.7% and 72%. Patients and relatives are the main perpetrators, followed by doctors, and, only in lower percentages, by nurses colleagues. Alcohol, drugs abuse, and overcrowding in EDs are acknowledged as motivating factors for violent events. Under-reporting of aggressions is frequent up to the 80% of victims, and some papers report that nurses consider assaults as a normal part of their work. There is a direct relation between aggressions and symptoms of post-traumatic stress disturb syndrome. Moreover there is a sense of continuous fear in nurses, causing the likelihood of workplace leaving. Special educational courses seem to be effective in diminishing the number of aggressions and to adopt adequate adaptive behaviors. 


Asunto(s)
Agresión , Servicio de Urgencia en Hospital , Enfermeras y Enfermeros , Violencia , Lugar de Trabajo , Humanos , Internacionalidad , Violencia/estadística & datos numéricos
19.
Cancer Discov ; 13(3): 746-765, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36455613

RESUMEN

The dysregulation of developmental and stem cell-associated genes is a common phenomenon during cancer development. Around half of patients with acute myeloid leukemia (AML) express high levels of HOXA cluster genes and MEIS1. Most of these AML cases harbor an NPM1 mutation (NPM1c), which encodes for an oncoprotein mislocalized from the nucleolus to the cytoplasm. How NPM1c expression in hematopoietic cells leads to its characteristic gene-expression pattern remains unclear. Here, we show that NPM1c directly binds to specific chromatin targets, which are co-occupied by the histone methyltransferase KMT2A (MLL1). Targeted degradation of NPM1c leads to a rapid decrease in gene expression and loss of RNA polymerase II, as well as activating histone modifications at its targets. We demonstrate that NPM1c directly regulates oncogenic gene expression in collaboration with the MLL1 complex and define the mechanism by which MLL1-Menin small-molecule inhibitors produce clinical responses in patients with NPM1-mutated AML. SIGNIFICANCE: We uncovered an important functional role of mutant NPM1 as a crucial direct driver of oncogenic gene expression in AML. NPM1c can bind to chromatin and cooperate with the MLL complex, providing the first functional insight into the mechanism of Menin-MLL inhibition in NPM1c leukemias. See related article by Wang et al., p. 724. This article is highlighted in the In This Issue feature, p. 517.


Asunto(s)
Leucemia Mieloide Aguda , Proteínas Nucleares , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Nucleofosmina , Mutación , Leucemia Mieloide Aguda/patología , Cromatina/genética
20.
Cell Rep ; 42(12): 113484, 2023 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-37999976

RESUMEN

The nucleolar scaffold protein NPM1 is a multifunctional regulator of cellular homeostasis, genome integrity, and stress response. NPM1 mutations, known as NPM1c variants promoting its aberrant cytoplasmic localization, are the most frequent genetic alterations in acute myeloid leukemia (AML). A hallmark of AML cells is their dependency on elevated autophagic flux. Here, we show that NPM1 and NPM1c induce the autophagy-lysosome pathway by activating the master transcription factor TFEB, thereby coordinating the expression of lysosomal proteins and autophagy regulators. Importantly, both NPM1 and NPM1c bind to autophagy modifiers of the GABARAP subfamily through an atypical binding module preserved within its N terminus. The propensity of NPM1c to induce autophagy depends on this module, likely indicating that NPM1c exerts its pro-autophagic activity by direct engagement with GABARAPL1. Our data report a non-canonical binding mode of GABARAP family members that drives the pro-autophagic potential of NPM1c, potentially enabling therapeutic options.


Asunto(s)
Leucemia Mieloide Aguda , Proteínas Nucleares , Humanos , Proteínas Nucleares/metabolismo , Leucemia Mieloide Aguda/metabolismo , Autofagia/fisiología , Mutación/genética , Lisosomas/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA