Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros

Base de datos
Tipo de estudio
Tipo del documento
Asunto de la revista
País de afiliación
Intervalo de año de publicación
1.
Nat Cell Biol ; 26(6): 946-961, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38745030

RESUMEN

RNA-binding proteins (RBPs) are pivotal in acute myeloid leukaemia (AML), a lethal disease. Although specific phase separation-competent RBPs are recognized in AML, the effect of their condensate formation on AML leukaemogenesis, and the therapeutic potential of inhibition of phase separation are underexplored. In our in vivo CRISPR RBP screen, fibrillarin (FBL) emerges as a crucial nucleolar protein that regulates AML cell survival, primarily through its phase separation domains rather than methyltransferase or acetylation domains. These phase separation domains, with specific features, coordinately drive nucleoli formation and early processing of pre-rRNA (including efflux, cleavage and methylation), eventually enhancing the translation of oncogenes such as MYC. Targeting the phase separation capability of FBL with CGX-635 leads to elimination of AML cells, suggesting an additional mechanism of action for CGX-635 that complements its established therapeutic effects. We highlight the potential of PS modulation of critical proteins as a possible therapeutic strategy for AML.


Asunto(s)
Proteínas Cromosómicas no Histona , Leucemia Mieloide Aguda , Precursores del ARN , Procesamiento Postranscripcional del ARN , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Leucemia Mieloide Aguda/metabolismo , Precursores del ARN/metabolismo , Precursores del ARN/genética , Proteínas Cromosómicas no Histona/metabolismo , Proteínas Cromosómicas no Histona/genética , Animales , Línea Celular Tumoral , Biosíntesis de Proteínas , Nucléolo Celular/metabolismo , Nucléolo Celular/genética , Ratones , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Regulación Leucémica de la Expresión Génica , Separación de Fases
2.
Blood ; 144(7): 742-756, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-38657191

RESUMEN

ABSTRACT: Hematopoietic differentiation is controlled by intrinsic regulators and the extrinsic hematopoietic niche. Activating transcription factor 4 (ATF4) plays a crucial role in the function of fetal and adult hematopoietic stem cell maintenance. However, the precise function of ATF4 in the bone marrow (BM) niche and the mechanism by which ATF4 regulates adult hematopoiesis remain largely unknown. Here, we used 4 cell-type-specific mouse Cre lines to achieve conditional knockout of Atf4 in Cdh5+ endothelial cells, Prx1+ BM stromal cells, Osx+ osteoprogenitor cells, and Mx1+ hematopoietic cells and uncovered the role of Atf4 in niche cells and hematopoiesis. Intriguingly, depletion of Atf4 in niche cells did not affect hematopoiesis; however, Atf4-deficient hematopoietic cells exhibited erythroid differentiation defects, leading to hypoplastic anemia. Mechanistically, ATF4 mediated direct regulation of Rps19bp1 transcription, which is, in turn, involved in 40 S ribosomal subunit assembly to coordinate ribosome biogenesis and promote erythropoiesis. Finally, we demonstrate that under conditions of 5-fluorouracil-induced stress, Atf4 depletion impedes the recovery of hematopoietic lineages, which requires efficient ribosome biogenesis. Taken together, our findings highlight the indispensable role of the ATF4-RPS19BP1 axis in the regulation of erythropoiesis.


Asunto(s)
Factor de Transcripción Activador 4 , Eritropoyesis , Ribosomas , Animales , Factor de Transcripción Activador 4/metabolismo , Factor de Transcripción Activador 4/genética , Ratones , Ribosomas/metabolismo , Proteínas Ribosómicas/metabolismo , Proteínas Ribosómicas/genética , Ratones Noqueados , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/citología
3.
Cell Discov ; 10(1): 35, 2024 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-38548771

RESUMEN

Microplastics (MPs) are contaminants ubiquitously found in the global biosphere that enter the body through inhalation or ingestion, posing significant risks to human health. Recent studies emerge that MPs are present in the bone marrow and damage the hematopoietic system. However, it remains largely elusive about the specific mechanisms by which MPs affect hematopoietic stem cells (HSCs) and their clinical relevance in HSC transplantation (HSCT). Here, we established a long-term MPs intake mouse model and found that MPs caused severe damage to the hematopoietic system. Oral gavage administration of MPs or fecal transplantation of microbiota from MPs-treated mice markedly undermined the self-renewal and reconstitution capacities of HSCs. Mechanistically, MPs did not directly kill HSCs but disrupted gut structure and permeability, which eventually ameliorated the abundance of Rikenellaceae and hypoxanthine in the intestine and inactivated the HPRT-Wnt signaling in bone marrow HSCs. Furthermore, administration of Rikenellaceae or hypoxanthine in mice as well as treatment of WNT10A in the culture system substantially rescued the MPs-induced HSC defects. Finally, we validated in a cohort of human patients receiving allogenic HSCT from healthy donors, and revealed that the survival time of patients was negatively correlated with levels of MPs, while positively with the abundance of Rikenellaceae, and hypoxanthine in the HSC donors' feces and blood. Overall, our study unleashes the detrimental roles and mechanisms of MPs in HSCs, which provides potential strategies to prevent hematopoietic damage from MPs and serves as a fundamental critique for selecting suitable donors for HSCT in clinical practice.

4.
Cell Rep Med ; 5(2): 101400, 2024 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-38307031

RESUMEN

Chimeric antigen receptor (CAR)-T therapy has shown superior efficacy against hematopoietic malignancies. However, many patients failed to achieve sustainable tumor control partially due to CAR-T cell exhaustion and limited persistence. In this study, by performing single-cell multi-omics data analysis on patient-derived CAR-T cells, we identify CD38 as a potential hallmark of exhausted CAR-T cells, which is positively correlated with exhaustion-related transcription factors and further confirmed with in vitro exhaustion models. Moreover, inhibiting CD38 activity reverses tonic signaling- or tumor antigen-induced exhaustion independent of single-chain variable fragment design or costimulatory domain, resulting in improved CAR-T cell cytotoxicity and antitumor response. Mechanistically, CD38 inhibition synergizes the downregulation of CD38-cADPR -Ca2+ signaling and activation of the CD38-NAD+-SIRT1 axis to suppress glycolysis. Collectively, our findings shed light on the role of CD38 in CAR-T cell exhaustion and suggest potential clinical applications of CD38 inhibition in enhancing the efficacy and persistence of CAR-T cell therapy.


Asunto(s)
Neoplasias , Anticuerpos de Cadena Única , Humanos , Linfocitos T , Inmunoterapia Adoptiva/métodos , Antígenos de Neoplasias/metabolismo
5.
Cell Metab ; 36(1): 176-192.e10, 2024 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-38171332

RESUMEN

The efficacy of chimeric antigen receptor (CAR) T cell therapy is hampered by relapse in hematologic malignancies and by hyporesponsiveness in solid tumors. Long-lived memory CAR T cells are critical for improving tumor clearance and long-term protection. However, during rapid ex vivo expansion or in vivo tumor eradication, metabolic shifts and inhibitory signals lead to terminal differentiation and exhaustion of CAR T cells. Through a mitochondria-related compound screening, we find that the FDA-approved isocitrate dehydrogenase 2 (IDH2) inhibitor enasidenib enhances memory CAR T cell formation and sustains anti-leukemic cytotoxicity in vivo. Mechanistically, IDH2 impedes metabolic fitness of CAR T cells by restraining glucose utilization via the pentose phosphate pathway, which alleviates oxidative stress, particularly in nutrient-restricted conditions. In addition, IDH2 limits cytosolic acetyl-CoA levels to prevent histone acetylation that promotes memory cell formation. In combination with pharmacological IDH2 inhibition, CAR T cell therapy is demonstrated to have superior efficacy in a pre-clinical model.


Asunto(s)
Antioxidantes , Neoplasias , Humanos , Antioxidantes/farmacología , Antioxidantes/metabolismo , Isocitrato Deshidrogenasa , Histonas/metabolismo , Acetilación , Linfocitos T , Neoplasias/metabolismo , Mitocondrias/metabolismo
6.
Front Med (Lausanne) ; 10: 1321490, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38105896

RESUMEN

Background: Severe fever with thrombocytopenia syndrome (SFTS) is an emerging and life-threatening infectious disease caused by SFTS virus. Although recent studies have reported the use of nomograms based on demographic and laboratory data to predict the prognosis of SFTS, no study has included viral load, which is an important factor that influences the prognosis, when compared with other risk factors. Therefore, this study aimed to develop a model that predicts SFTS prognosis before it reaches the critical illness stage and to compare the predictive ability of groups with and without viral load. Methods: Two hundred patients with SFTS were enrolled between June 2018 and August 2023. Data were sourced from the first laboratory results at admission, and two nomograms for mortality risk were developed using multivariate logistic regression to identify the risk variables for poor prognosis in these patients. We calculated the area under the receiver operating characteristic curve (AUC) for the two nomograms to assess their discrimination, and predictive abilities were compared using net reclassification improvement (NRI) and integrated discrimination improvement (IDI). Results: The multivariate logistic regression analysis identified four independent risk factors: age, bleeding manifestations, prolonged activated partial thromboplastin time, and viral load. Based on these factors, a final nomogram predicting mortality risk in patients with SFTS was constructed; in addition, a simplified nomogram was constructed excluding the viral load. The AUC [0.926, 95% confidence interval (CI): 0.882-0.970 vs. 0.882, 95% CI: 35 0.823-0.942], NRI (0.143, 95% CI, 0.036-0.285), and IDI (0.124, 95% CI, 0.061-0.186) were calculated and compared between the two models. The calibration curves of the two models showed excellent concordance, and decision curve analysis was used to quantify the net benefit at different threshold probabilities. Conclusion: Two critical risk nomograms were developed based on the indicators for early prediction of mortality risk in patients with SFTS, and enhanced predictive accuracy was observed in the model that incorporated the viral load. The models developed will provide frontline clinicians with a convenient tool for early identification of critically ill patients and initiation of a better personalized treatment in a timely manner.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA