Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Haematologica ; 108(10): 2715-2729, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37102608

RESUMEN

Inflammation is associated with the pathogenesis of myelodysplastic syndromes (MDS) and emerging evidence suggests that MDS hematopoietic stem and progenitor cells (HSPC) exhibit an altered response to inflammation. Deletion of chromosome 5 (del(5q)) is the most common chromosomal abnormality in MDS. Although this MDS subtype contains several haploinsufficient genes that impact innate immune signaling, the effects of inflammation on del(5q) MDS HSPC remains undefined. Utilizing a model of del(5q)-like MDS, inhibiting the IRAK1/4-TRAF6 axis improved cytopenias, suggesting that activation of innate immune pathways contributes to certain clinical features underlying the pathogenesis of low-risk MDS. However, low-grade inflammation in the del(5q)-like MDS model did not contribute to more severe disease but instead impaired the del(5q)-like HSPC as indicated by their diminished numbers, premature attrition and increased p53 expression. Del(5q)-like HSPC exposed to inflammation became less quiescent, but without affecting cell viability. Unexpectedly, the reduced cellular quiescence of del(5q) HSPC exposed to inflammation was restored by p53 deletion. These findings uncovered that inflammation confers a competitive advantage of functionally defective del(5q) HSPC upon loss of p53. Since TP53 mutations are enriched in del(5q) AML following an MDS diagnosis, increased p53 activation in del(5q) MDS HSPC due to inflammation may create a selective pressure for genetic inactivation of p53 or expansion of a pre-existing TP53-mutant clone.


Asunto(s)
Síndromes Mielodisplásicos , Proteína p53 Supresora de Tumor , Humanos , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Deleción Cromosómica , Síndromes Mielodisplásicos/patología , Células Madre Hematopoyéticas/metabolismo , Transducción de Señal , Cromosomas Humanos Par 5/genética , Cromosomas Humanos Par 5/metabolismo
2.
Sci Transl Med ; 14(635): eabb7695, 2022 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-35263148

RESUMEN

Dysregulation of innate immune signaling pathways is implicated in various hematologic malignancies. However, these pathways have not been systematically examined in acute myeloid leukemia (AML). We report that AML hematopoietic stem and progenitor cells (HSPCs) exhibit a high frequency of dysregulated innate immune-related and inflammatory pathways, referred to as oncogenic immune signaling states. Through gene expression analyses and functional studies in human AML cell lines and patient-derived samples, we found that the ubiquitin-conjugating enzyme UBE2N is required for leukemic cell function in vitro and in vivo by maintaining oncogenic immune signaling states. It is known that the enzyme function of UBE2N can be inhibited by interfering with thioester formation between ubiquitin and the active site. We performed in silico structure-based and cellular-based screens and identified two related small-molecule inhibitors UC-764864/65 that targeted UBE2N at its active site. Using these small-molecule inhibitors as chemical probes, we further revealed the therapeutic efficacy of interfering with UBE2N function. This resulted in the blocking of ubiquitination of innate immune- and inflammatory-related substrates in human AML cell lines. Inhibition of UBE2N function disrupted oncogenic immune signaling by promoting cell death of leukemic HSPCs while sparing normal HSPCs in vitro. Moreover, baseline oncogenic immune signaling states in leukemic cells derived from discrete subsets of patients with AML exhibited a selective dependency on UBE2N function in vitro and in vivo. Our study reveals that interfering with UBE2N abrogates leukemic HSPC function and underscores the dependency of AML cells on UBE2N-dependent oncogenic immune signaling states.


Asunto(s)
Leucemia Mieloide Aguda , Enzimas Ubiquitina-Conjugadoras , Proliferación Celular/genética , Humanos , Leucemia Mieloide Aguda/metabolismo , Oncogenes , Transducción de Señal/genética , Enzimas Ubiquitina-Conjugadoras/antagonistas & inhibidores , Enzimas Ubiquitina-Conjugadoras/genética , Enzimas Ubiquitina-Conjugadoras/metabolismo
3.
Leukemia ; 36(2): 438-451, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34465865

RESUMEN

Ubiquitin-specific peptidase 15 (USP15) is a deubiquitinating enzyme implicated in critical cellular and oncogenic processes. We report that USP15 mRNA and protein are overexpressed in human acute myeloid leukemia (AML) as compared to normal hematopoietic progenitor cells. This high expression of USP15 in AML correlates with KEAP1 protein and suppression of NRF2. Knockdown or deletion of USP15 in human and mouse AML models significantly impairs leukemic progenitor function and viability and de-represses an antioxidant response through the KEAP1-NRF2 axis. Inhibition of USP15 and subsequent activation of NRF2 leads to redox perturbations in AML cells, coincident with impaired leukemic cell function. In contrast, USP15 is dispensable for human and mouse normal hematopoietic cells in vitro and in vivo. A preclinical small-molecule inhibitor of USP15 induced the KEAP1-NRF2 axis and impaired AML cell function, suggesting that targeting USP15 catalytic function can suppress AML. Based on these findings, we report that USP15 drives AML cell function, in part, by suppressing a critical oxidative stress sensor mechanism and permitting an aberrant redox state. Furthermore, we postulate that inhibition of USP15 activity with small molecule inhibitors will selectively impair leukemic progenitor cells by re-engaging homeostatic redox responses while sparing normal hematopoiesis.


Asunto(s)
Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Leucemia Mieloide Aguda/patología , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo , Proteasas Ubiquitina-Específicas/metabolismo , Proteasas Ubiquitina-Específicas/fisiología , Animales , Apoptosis , Proliferación Celular , Femenino , Humanos , Proteína 1 Asociada A ECH Tipo Kelch/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Factor 2 Relacionado con NF-E2/genética , Oxidación-Reducción , Pronóstico , Transducción de Señal , Células Tumorales Cultivadas , Proteasas Ubiquitina-Específicas/genética , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Exp Hematol ; 90: 18-29, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32910997

RESUMEN

Forkhead-associated (FHA) domain-containing proteins are widely expressed across eubacteria and in eukaryotes. FHA domains contain phosphopeptide recognition motifs, which operate in a variety of phosphorylation-dependent and -independent biological processes, including the DNA damage response, signal transduction, and regulation of the cell cycle. More recently, two FHA domain-containing proteins were discovered in mammalian cells as tumor necrosis factor receptor-associated factor (TRAF)-interacting proteins: TIFA and TIFAB. TIFA and TIFAB are important modifiers of the innate immune signaling through their regulation of TRAF proteins. Recent studies have also revealed distinct roles for TIFA and TIFAB in the context of immune cell function, chronic inflammation, hematopoiesis, and hematologic disorders. Collectively, these studies indicate the important role of TIFA- and TIFAB-dependent signaling in hematopoietic cells and their dysregulation in several human diseases. In this review, we summarize the molecular mechanisms and biological role of these FHA-domain homologues, placing them into the context of human disease.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Enfermedades Hematológicas/inmunología , Hematopoyesis/inmunología , Péptidos y Proteínas de Señalización Intracelular/inmunología , Transducción de Señal/inmunología , Animales , Humanos , Inflamación/inmunología , Inflamación/patología
5.
Cell Rep ; 30(8): 2776-2790.e6, 2020 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-32101751

RESUMEN

TRAF-interacting protein with a forkhead-associated domain B (TIFAB) is implicated in myeloid malignancies with deletion of chromosome 5q. Employing a combination of proteomic and genetic approaches, we find that TIFAB regulates ubiquitin-specific peptidase 15 (USP15) ubiquitin hydrolase activity. Expression of TIFAB in hematopoietic stem/progenitor cells (HSPCs) permits USP15 signaling to substrates, including MDM2 and KEAP1, and mitigates p53 expression. Consequently, TIFAB-deficient HSPCs exhibit compromised USP15 signaling and are sensitized to hematopoietic stress by derepression of p53. In MLL-AF9 leukemia, deletion of TIFAB increases p53 signaling and correspondingly decreases leukemic cell function and development of leukemia. Restoring USP15 expression partially rescues the function of TIFAB-deficient MLL-AF9 cells. Conversely, elevated TIFAB represses p53, increases leukemic progenitor function, and correlates with MLL gene expression programs in leukemia patients. Our studies uncover a function of TIFAB as an effector of USP15 activity and rheostat of p53 signaling in stressed and malignant HSPCs.


Asunto(s)
Hematopoyesis , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo , Proteasas Ubiquitina-Específicas/metabolismo , Animales , Dominio Catalítico , Línea Celular Tumoral , Modelos Animales de Enfermedad , Células HEK293 , Células Madre Hematopoyéticas/metabolismo , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Ratones , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Unión Proteica , Estrés Fisiológico , Ubiquitina/metabolismo , Proteasas Ubiquitina-Específicas/química , Ubiquitinación
6.
Cell Stem Cell ; 20(5): 579-581, 2017 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-28475879

RESUMEN

Hematopoietic stem cells (HSCs) in the bone marrow (BM) form mature blood cells of all lineages through expansion of lineage-biased progenitors. In a recent issue of Nature, Hérault et al. (2017) uncover a unique spatiotemporal mechanism of granulocyte-macrophage progenitors (GMPs) employed in emergency hematopoiesis that is also hijacked in leukemia.


Asunto(s)
Células Progenitoras de Granulocitos y Macrófagos/citología , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Leucemia Mieloide Aguda/metabolismo , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Humanos , Modelos Biológicos , Mielopoyesis/fisiología
7.
J Exp Med ; 212(11): 1967-85, 2015 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-26458771

RESUMEN

TRAF-interacting protein with forkhead-associated domain B (TIFAB) is a haploinsufficient gene in del(5q) myelodysplastic syndrome (MDS). Deletion of Tifab results in progressive bone marrow (BM) and blood defects, including skewed hematopoietic stem/progenitor cell (HSPC) proportions and altered myeloid differentiation. A subset of mice transplanted with Tifab knockout (KO) HSPCs develop a BM failure with neutrophil dysplasia and cytopenia. In competitive transplants, Tifab KO HSPCs are out-competed by wild-type (WT) cells, suggesting a cell-intrinsic defect. Gene expression analysis of Tifab KO HSPCs identified dysregulation of immune-related signatures, and hypersensitivity to TLR4 stimulation. TIFAB forms a complex with TRAF6, a mediator of immune signaling, and reduces TRAF6 protein stability by a lysosome-dependent mechanism. In contrast, TIFAB loss increases TRAF6 protein and the dynamic range of TLR4 signaling, contributing to ineffective hematopoiesis. Moreover, combined deletion of TIFAB and miR-146a, two genes associated with del(5q) MDS/AML, results in a cooperative increase in TRAF6 expression and hematopoietic dysfunction. Re-expression of TIFAB in del(5q) MDS/AML cells results in attenuated TLR4 signaling and reduced viability. These findings underscore the importance of efficient regulation of innate immune/TRAF6 signaling within HSPCs by TIFAB, and its cooperation with miR-146a as it relates to the pathogenesis of hematopoietic malignancies, such as del(5q) MDS/AML.


Asunto(s)
Hematopoyesis , Proteínas/fisiología , Transducción de Señal/fisiología , Factor 6 Asociado a Receptor de TNF/fisiología , Receptores Toll-Like/fisiología , Animales , Apoptosis , Trasplante de Médula Ósea , Diferenciación Celular , Cromosomas Humanos Par 5 , Humanos , Péptidos y Proteínas de Señalización Intracelular , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/fisiología , FN-kappa B/antagonistas & inhibidores , FN-kappa B/fisiología , Proteínas/genética
8.
Exp Hematol ; 43(8): 587-598, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26143580

RESUMEN

Overexpression of immune-related genes is widely reported in myelodysplastic syndromes (MDSs), and chronic immune stimulation increases the risk for developing MDS. Aberrant innate immune activation, such as that caused by increased toll-like receptor (TLR) signaling, in MDS can contribute to systemic effects on hematopoiesis, in addition to cell-intrinsic defects on hematopoietic stem/progenitor cell (HSPC) function. This review will deconstruct aberrant function of TLR signaling mediators within MDS HSPCs that may contribute to cell-intrinsic consequences on hematopoiesis and disease pathogenesis. We will discuss the contribution of chronic TLR signaling to the pathogenesis of MDS based on evidence from patients and mouse genetic models.


Asunto(s)
Hematopoyesis/inmunología , Células Madre Hematopoyéticas/inmunología , Inmunidad Innata , Síndromes Mielodisplásicos/inmunología , Transducción de Señal/inmunología , Receptores Toll-Like/inmunología , Animales , Modelos Animales de Enfermedad , Hematopoyesis/genética , Células Madre Hematopoyéticas/patología , Humanos , Ratones , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/patología , Transducción de Señal/genética , Receptores Toll-Like/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...