Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 186(19): 4189-4203.e22, 2023 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-37633268

RESUMEN

Thrombopoietin (THPO or TPO) is an essential cytokine for hematopoietic stem cell (HSC) maintenance and megakaryocyte differentiation. Here, we report the 3.4 Å resolution cryoelectron microscopy structure of the extracellular TPO-TPO receptor (TpoR or MPL) signaling complex, revealing the basis for homodimeric MPL activation and providing a structural rationalization for genetic loss-of-function thrombocytopenia mutations. The structure guided the engineering of TPO variants (TPOmod) with a spectrum of signaling activities, from neutral antagonists to partial- and super-agonists. Partial agonist TPOmod decoupled JAK/STAT from ERK/AKT/CREB activation, driving a bias for megakaryopoiesis and platelet production without causing significant HSC expansion in mice and showing superior maintenance of human HSCs in vitro. These data demonstrate the functional uncoupling of the two primary roles of TPO, highlighting the potential utility of TPOmod in hematology research and clinical HSC transplantation.


Asunto(s)
Receptores de Trombopoyetina , Trombopoyetina , Animales , Humanos , Ratones , Ciclo Celular , Microscopía por Crioelectrón , Receptores de Trombopoyetina/genética , Trombopoyesis , Metilación de ADN
2.
Biomacromolecules ; 24(11): 4646-4652, 2023 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-37792488

RESUMEN

Thiol-reactive Michael acceptors are commonly used for the formation of chemically cross-linked hydrogels. In this paper, we address the drawbacks of many Michael acceptors by introducing pyridazinediones as new cross-linking agents. Through the use of pyridazinediones and their mono- or dibrominated analogues, we show that the mechanical strength, swelling ratio, and rate of gelation can all be controlled in a pH-sensitive manner. Moreover, we demonstrate that the degradation of pyridazinedione-gels can be induced by the addition of thiols, thus providing a route to responsive or dynamic gels, and that monobromo-pyridazinedione gels are able to support the proliferation of human cells. We anticipate that our results will provide a valuable and complementary addition to the existing toolkit of cross-linking agents, allowing researchers to tune and rationally design the properties of biomedical hydrogels.


Asunto(s)
Hidrogeles , Compuestos de Sulfhidrilo , Humanos , Hidrogeles/química , Compuestos de Sulfhidrilo/química , Reactivos de Enlaces Cruzados/química
3.
Biochim Biophys Acta Mol Cell Res ; 1865(1): 105-116, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28939057

RESUMEN

The Cdc28 protein kinase subunits, Cks1 and Cks2, play dual roles in Cdk-substrate specificity and Cdk-independent protein degradation, in concert with the E3 ubiquitin ligase complexes SCFSkp2 and APCCdc20. Notable targets controlled by Cks include p27 and Cyclin A. Here, we demonstrate that Cks1 and Cks2 proteins interact with both the MllN and MllC subunits of Mll1 (Mixed-lineage leukaemia 1), and together, the Cks proteins define Mll1 levels throughout the cell cycle. Overexpression of CKS1B and CKS2 is observed in multiple human cancers, including various MLL-rearranged (MLLr) AML subtypes. To explore the importance of MLL-Fusion Protein regulation by CKS1/2, we used small molecule inhibitors (MLN4924 and C1) to modulate their protein degradation functions. These inhibitors specifically reduced the proliferation of MLLr cell lines compared to primary controls. Altogether, this study uncovers a novel regulatory pathway for MLL1, which may open a new therapeutic approach to MLLr leukaemia.


Asunto(s)
Quinasas CDC2-CDC28/fisiología , Proteínas Portadoras/fisiología , Proteínas de Ciclo Celular/fisiología , N-Metiltransferasa de Histona-Lisina/genética , Leucemia/genética , Leucemia/patología , Proteína de la Leucemia Mieloide-Linfoide/genética , Animales , Proteína Quinasa CDC28 de Saccharomyces cerevisiae/fisiología , Supervivencia Celular/genética , Células Cultivadas , Embrión de Mamíferos , Regulación Leucémica de la Expresión Génica , Reordenamiento Génico , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Ratones , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Transducción de Señal/genética
5.
Cereb Cortex ; 27(1): 11-23, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-28365778

RESUMEN

In mitotic cells, the cyclin-dependent kinase (CDK) subunit protein CKS1 regulates S phase entry by mediating degradation of the CDK inhibitor p27. Although mature neurons lack mitotic CDKs, we found that CKS1 was actively expressed in post-mitotic neurons of the adult hippocampus. Interestingly, Cks1 knockout (Cks1-/-) mice exhibited poor long-term memory, and diminished maintenance of long-term potentiation in the hippocampal circuits. Furthermore, there was neuronal accumulation of cofilin-actin rods or cofilin aggregates, which are associated with defective dendritic spine maturation and synaptic loss. We further demonstrated that it was the increased p27 level that activated cofilin by suppressing the RhoA kinase-mediated inhibitory phosphorylation of cofilin, resulting in the formation of cofilin aggregates in the Cks1-/- neuronal cells. Consistent with reports that the peptidyl-prolyl-isomerase PIN1 competes with CKS1 for p27 binding, we found that inhibition of PIN1 diminished the formation of cofilin aggregates through decreasing p27 levels, thereby activating RhoA and increasing cofilin phosphorylation. Our results revealed that CKS1 is involved in normal glutamatergic synapse development and dendritic spine maturation in adult hippocampus through modulating p27 stability.


Asunto(s)
Factores Despolimerizantes de la Actina/metabolismo , Quinasas CDC2-CDC28/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Hipocampo/metabolismo , Trastornos de la Memoria/metabolismo , Memoria a Largo Plazo , Neuronas/metabolismo , Animales , Quinasas CDC2-CDC28/genética , Ciclo Celular , Espinas Dendríticas , Hipocampo/patología , Potenciación a Largo Plazo , Masculino , Trastornos de la Memoria/patología , Ratones , Ratones Noqueados , Agregado de Proteínas , Aprendizaje Espacial
6.
Hum Mutat ; 34(6): 864-8, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23505216

RESUMEN

Germline mutations in the cyclin-dependent kinase inhibitor, CDKN1B, have been described in patients with multiple endocrine neoplasia (MEN), a cancer predisposition syndrome with adult onset neoplasia and no additional phenotypes. Here, we describe the first human case of CDKN1B deficiency, which recapitulates features of the murine CDKN1B knockout mouse model, including gigantism and neurodevelopmental defects. Decreased mRNA and protein expression of CDKN1B were confirmed in the proband's peripheral blood, which is not seen in MEN syndrome patients. We ascribed the decreased protein level to a maternally derived deletion on chromosome 12p13 encompassing the CDKN1B locus (which reduced mRNA expression) and a de novo allelic variant (c.-73G>A) in the CDKN1B promoter (which reduced protein translation). We propose a recessive model where decreased dosage of CDKN1B during development in humans results in a neuronal phenotype akin to that described in mice, placing CDKN1B as a candidate gene involved in developmental delay.


Asunto(s)
Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Discapacidades del Desarrollo/genética , Trastorno Autístico/genética , Preescolar , Hibridación Genómica Comparativa , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/deficiencia , Discapacidades del Desarrollo/diagnóstico , Femenino , Expresión Génica , Humanos , Masculino , Mutación , Linaje , Polimorfismo de Nucleótido Simple
7.
Hemasphere ; 7(3): e853, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36874381

RESUMEN

Long-term hematopoietic stem cells are rare, highly quiescent stem cells of the hematopoietic system with life-long self-renewal potential and the ability to transplant and reconstitute the entire hematopoietic system of conditioned recipients. Most of our understanding of these rare cells has relied on cell surface identification, epigenetic, and transcriptomic analyses. Our knowledge of protein synthesis, folding, modification, and degradation-broadly termed protein homeostasis or "proteostasis"-in these cells is still in its infancy, with very little known about how the functional state of the proteome is maintained in hematopoietic stem cells. We investigated the requirement of the small phospho-binding adaptor proteins, the cyclin-dependent kinase subunits (CKS1 and CKS2), for maintaining ordered hematopoiesis and long-term hematopoietic stem cell reconstitution. CKS1 and CKS2 are best known for their roles in p27 degradation and cell cycle regulation, and by studying the transcriptome and proteome of Cks1 -/- and Cks2 -/- mice, we demonstrate regulation of key signaling pathways that govern hematopoietic stem cell biology including AKT, FOXO1, and NFκB, together balancing protein homeostasis and restraining reactive oxygen species to ensure healthy hematopoietic stem cell function.

8.
Front Physiol ; 13: 1009160, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36246104

RESUMEN

The haematopoietic system is a classical stem cell hierarchy that maintains all the blood cells in the body. Haematopoietic stem cells (HSCs) are rare, highly potent cells that reside at the apex of this hierarchy and are historically some of the most well studied stem cells in humans and laboratory models, with haematopoiesis being the original system to define functional cell types by cell surface markers. Whilst it is possible to isolate HSCs to near purity, we know very little about the functional activity of markers to purify HSCs. This review will focus on the historical efforts to purify HSCs in humans based on cell surface markers, their putative functions and recent advances in finding functional markers on HSCs.

9.
Ultrasonics ; 118: 106557, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34509048

RESUMEN

Polymer tanks made of Polypropylene (PP) sub-assemblies are commonly used for their ease of formability, surface and optical quality. The tightness of an assembly is classically assessed by pressurizing the vessel and measuring a pressure drop after a given period. In order to avoid this long and imprecise method, active ultrasound methods can be envisioned, but should be carefully designed in order to derive the proper transducer configurations, frequencies and assess the performances of the method in terms of repeatability and detectability. In this article, a thermoviscous Finite Element Model (FEM) is derived in order to predict the effect of realistic damages on the measured transmitted acoustic field in bonded polymer joints. Three damage scenarios are considered, namely through holes, flat-bottom holes and internal voids that may impair the tightness and durability of a polymer assembly. Numerical results in terms of on-axis sound pressure spectra and directivity diagrams are presented and verified experimentally on a flat panel. These results allow the derivation of design rules for the active inspection of polymer jointed structures.

10.
Sci Transl Med ; 14(650): eabn3248, 2022 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-35731890

RESUMEN

Acute myeloid leukemia (AML) is an aggressive hematological disorder comprising a hierarchy of quiescent leukemic stem cells (LSCs) and proliferating blasts with limited self-renewal ability. AML has a dismal prognosis, with extremely low 2-year survival rates in the poorest cytogenetic risk patients, primarily due to the failure of intensive chemotherapy protocols to deplete LSCs and toxicity of therapy toward healthy hematopoietic cells. We studied the role of cyclin-dependent kinase regulatory subunit 1 (CKS1)-dependent protein degradation in primary human AML and healthy hematopoiesis xenograft models in vivo. Using a small-molecule inhibitor (CKS1i), we demonstrate a dual role for CKS1-dependent protein degradation in reducing patient-derived AML blasts in vivo and, importantly, depleting LSCs, whereas inhibition of CKS1 has the opposite effect on normal hematopoiesis, protecting normal hematopoietic stem cells from chemotherapeutic toxicity. Proteomic analysis of responses to CKS1i in our patient-derived xenograft mouse model demonstrate that inhibition of CKS1 in AML leads to hyperactivation of RAC1 and accumulation of lethal reactive oxygen species, whereas healthy hematopoietic cells enter quiescence in response to CKS1i, protecting hematopoietic stem cells. Together, these findings demonstrate that CKS1-dependent proteostasis is a key vulnerability in malignant stem cell biology.


Asunto(s)
Quinasas CDC2-CDC28 , Leucemia Mieloide Aguda , Animales , Quinasas CDC2-CDC28/metabolismo , Quinasas CDC2-CDC28/farmacología , Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Ratones , Células Madre Neoplásicas , Proteómica
11.
Exp Hematol ; 95: 23-30, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33497761

RESUMEN

Acute myeloid leukemia (AML) is a heterogeneous disease, yet clinically most patients present with pancytopenia resulting from bone marrow failure, predisposing them to life-threatening infections and bleeding. The mechanisms by which AML mediates hematopoietic suppression is not well known. Indeed, much effort has so far been focused on how AML remodels the bone marrow niche to make it a more permissive environment, with less focus on how the remodeled niche affects normal hematopoietic cells. In this perspective, we present evidence of the key role of the bone marrow niche in suppressing hematopoietic stem cells (HSCs) during leukemic progression and provide perspectives on how future research on this topic may be exploited to provide treatments for one of the key complications of AML.


Asunto(s)
Células Madre Hematopoyéticas/citología , Leucemia Mieloide Aguda/patología , Células Madre Neoplásicas/citología , Nicho de Células Madre , Microambiente Tumoral , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Comunicación Celular , Hipoxia de la Célula , Deferoxamina/farmacología , Progresión de la Enfermedad , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Exosomas/fisiología , Hematopoyesis , Células Madre Hematopoyéticas/efectos de los fármacos , Homeostasis , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Neoplásicas/efectos de los fármacos , Recurrencia , Células Tumorales Cultivadas
12.
Stem Cell Reports ; 16(3): 428-436, 2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33581053

RESUMEN

We document here that intensive care COVID-19 patients suffer a profound decline in hemoglobin levels but show an increase of circulating nucleated red cells, suggesting that SARS-CoV-2 infection either directly or indirectly induces stress erythropoiesis. We show that ACE2 expression peaks during erythropoiesis and renders erythroid progenitors vulnerable to infection by SARS-CoV-2. Early erythroid progenitors, defined as CD34-CD117+CD71+CD235a-, show the highest levels of ACE2 and constitute the primary target cell to be infected during erythropoiesis. SARS-CoV-2 causes the expansion of colony formation by erythroid progenitors and can be detected in these cells after 2 weeks of the initial infection. Our findings constitute the first report of SARS-CoV-2 infectivity in erythroid progenitor cells and can contribute to understanding both the clinical symptoms of severe COVID-19 patients and how the virus can spread through the circulation to produce local inflammation in tissues, including the bone marrow.


Asunto(s)
COVID-19/virología , Células Precursoras Eritroides/virología , Eritropoyesis/fisiología , SARS-CoV-2/patogenicidad , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , COVID-19/metabolismo , Línea Celular , Chlorocebus aethiops , Células Precursoras Eritroides/metabolismo , Humanos , Inflamación/metabolismo , Inflamación/virología , Células Vero
13.
Nat Commun ; 11(1): 1792, 2020 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-32286289

RESUMEN

Continuous cancer growth is driven by subsets of self-renewing malignant cells. Targeting of uncontrolled self-renewal through inhibition of stem cell-related signaling pathways has proven challenging. Here, we show that cancer cells can be selectively deprived of self-renewal ability by interfering with their epigenetic state. Re-expression of histone H1.0, a tumor-suppressive factor that inhibits cancer cell self-renewal in many cancer types, can be broadly induced by the clinically well-tolerated compound Quisinostat. Through H1.0, Quisinostat inhibits cancer cell self-renewal and halts tumor maintenance without affecting normal stem cell function. Quisinostat also hinders expansion of cells surviving targeted therapy, independently of the cancer types and the resistance mechanism, and inhibits disease relapse in mouse models of lung cancer. Our results identify H1.0 as a major mediator of Quisinostat's antitumor effect and suggest that sequential administration of targeted therapy and Quisinostat may be a broadly applicable strategy to induce a prolonged response in patients.


Asunto(s)
Autorrenovación de las Células , Histonas/metabolismo , Ácidos Hidroxámicos/farmacología , Neoplasias/metabolismo , Neoplasias/patología , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Autorrenovación de las Células/efectos de los fármacos , Autorrenovación de las Células/genética , Supervivencia Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Ratones , Neoplasias/genética , Recurrencia
14.
Environ Entomol ; 38(2): 387-94, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19389287

RESUMEN

A complex of Fusarium spp., including F. pseudograminearum, F. culmorum, F. avenaceum, F. equiseti, and F. acuminatum, was isolated from field-collected larval cadavers of wheat stem sawfly at two locations for 2 yr. The Fusarium spp. isolates caused mortality in both diapausing larvae in a topical bioassay and in developing larvae feeding in infected stems in a greenhouse experiment. Larval mortality was >90% in both experiments at the highest dose. The pattern of correlation between integument discoloration, hyphal growth, and larval mortality showed that the Fusarium spp. isolates actively infect larvae and kill them, rather than colonizing larval tissue as secondary postmortem invaders. The versatility of Fusarium spp. as plant and insect pathogens enables colonization that results in disease in wheat plants and subsequent mortality of the wheat stem sawfly larvae developing within the same tissue.


Asunto(s)
Fusarium/fisiología , Himenópteros/microbiología , Triticum/microbiología , Animales , Fusarium/clasificación , Fusarium/aislamiento & purificación , Himenópteros/crecimiento & desarrollo , Himenópteros/fisiología , Larva/crecimiento & desarrollo , Larva/microbiología , Larva/fisiología , Enfermedades de las Plantas/microbiología
15.
Blood Cancer J ; 9(3): 28, 2019 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-30820024

RESUMEN

Since the publication of the original article the authors noticed the the affiliation details for Paresh Vyas are incorrect. The correct affiliation details for this author are given below.

16.
J Exp Med ; 215(3): 729-743, 2018 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-29453226

RESUMEN

Xenotransplantation of patient-derived samples in mouse models has been instrumental in depicting the role of hematopoietic stem and progenitor cells in the establishment as well as progression of hematological malignancies. The foundations for this field of research have been based on the development of immunodeficient mouse models, which provide normal and malignant human hematopoietic cells with a supportive microenvironment. Immunosuppressed and genetically modified mice expressing human growth factors were key milestones in patient-derived xenograft (PDX) models, highlighting the importance of developing humanized microenvironments. The latest major improvement has been the use of human bone marrow (BM) niche-forming cells to generate human-mouse chimeric BM tissues in PDXs, which can shed light on the interactions between human stroma and hematopoietic cells. Here, we summarize the methods used for human hematopoietic cell xenotransplantation and their milestones and review the latest approaches in generating humanized BM tissues in mice to study human normal and malignant hematopoiesis.


Asunto(s)
Bioingeniería/métodos , Trasplante de Médula Ósea , Médula Ósea/metabolismo , Nicho de Células Madre , Animales , Hematopoyesis , Humanos , Ratones , Trasplante Heterólogo
17.
Blood Cancer J ; 8(8): 68, 2018 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-30061630

RESUMEN

In acute myeloid leukemia (AML), risk stratification based on cytogenetics and mutation profiling is essential but remains insufficient to select the optimal therapy. Accurate biomarkers are needed to improve prognostic assessment. We analyzed RNA sequencing and survival data of 430 AML patients and identified HMGA2 as a novel prognostic marker. We validated a quantitative PCR test to study the association of HMGA2 expression with clinical outcomes in 358 AML samples. In this training cohort, HMGA2 was highly expressed in 22.3% of AML, mostly in patients with intermediate or adverse cytogenetics. High expression levels of HMGA2 (H + ) were associated with a lower frequency of complete remission (58.8% vs 83.4%, P < 0.001), worse 3-year overall survival (OS, 13.2% vs 43.5%, P < 0.001) and relapse-free survival (RFS, 10.8% vs 44.2%, P < 0.001). A positive HMGA2 test also identified a subgroup of patients unresponsive to standard treatments. Multivariable analyses showed that H + was independently associated with significantly worse OS and RFS, including in the intermediate cytogenetic risk category. These associations were confirmed in a validation cohort of 260 patient samples from the UK NCRI AML17 trial. The HMGA2 test could be implemented in clinical trials developing novel therapeutic strategies for high-risk AML.

18.
Endocr Pathol ; 28(1): 41-48, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27704398

RESUMEN

The REarranged during Transfection (RET) proto-oncogene is a receptor tyrosine kinase involved in growth and differentiation during embryogenesis and maintenance of the urogenital and nervous systems in mammals. Distinct mutations across hotspot RET exons can cause Multiple Endocrine Neoplasia Type 2A (MEN2A) characterised by development of medullary thyroid cancer (MTC), phaeochromocytoma (PCC) and primary hyperparathyroidism (PHPT), with a strong correlation between genotype and phenotype. Here, we report a 42-year-old man presented in the clinic with a unilateral PCC, with subsequent investigations revealing a nodular and cystic thyroid gland. He proceeded to thyroidectomy, which showed bilateral C-cell hyperplasia (CCH) without evidence of MTC. His brother had neonatal Hirschsprung disease (HSCR). Genetic testing revealed the presence of a heterozygous variant of unknown significance (VUS) in the cysteine-rich region of exon 10 in the RET gene (c.1846G>C, p.E616Q), in both affected siblings and their unaffected mother. Exon 10 RET mutations are known to be associated with HSCR and MEN2. Variants in the cysteine-rich region of the RET gene, outside of the key cysteine residues, may contribute to the development of MEN2 in a less aggressive manner, with a lower penetrance of MTC. Currently, a VUS in RET cannot be used to inform clinical management and direct future care. Analysis of RETE616Q reveals a gain of function mutant phenotype for this variant, which has not previously been reported, indicating that this VUS should be considered at risk for future clinical management.


Asunto(s)
Neoplasia Endocrina Múltiple Tipo 2a/genética , Proteínas Proto-Oncogénicas c-ret/genética , Adulto , Western Blotting , Femenino , Humanos , Masculino , Mutagénesis Sitio-Dirigida , Mutación , Linaje , Proto-Oncogenes Mas
19.
Nat Commun ; 8(1): 1679, 2017 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-29162833

RESUMEN

Cytarabine (AraC) represents the most effective single agent treatment for AML. Nevertheless, overriding AraC resistance in AML remains an unmet medical need. Here we show that the CHK1 inhibitor (CHK1i) GDC-0575 enhances AraC-mediated killing of AML cells both in vitro and in vivo, thus abrogating any potential chemoresistance mechanisms involving DNA repair. Importantly, this combination of drugs does not affect normal long-term hematopoietic stem/progenitors. Moreover, the addition of CHK1i to AraC does not generate de novo mutations and in patients' samples where AraC is mutagenic, addition of CHK1i appears to eliminate the generation of mutant clones. Finally, we observe that persistent residual leukemic cells are quiescent and can become responsive to the treatment when forced into cycle via granulocyte colony-stimulating factor (G-CSF) administration. This drug combination (AraC+CHK1i+G-CSF) will open the doors for a more efficient treatment of AML in the clinic.


Asunto(s)
Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/antagonistas & inhibidores , Citarabina/administración & dosificación , Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Leucemia Mieloide Aguda/tratamiento farmacológico , Piperidinas/administración & dosificación , Inhibidores de Proteínas Quinasas/administración & dosificación , Piridinas/administración & dosificación , Pirroles/administración & dosificación , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Línea Celular Tumoral , Resistencia a Antineoplásicos , Femenino , Células HL-60 , Hematopoyesis/efectos de los fármacos , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mutación/efectos de los fármacos , Células U937 , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA