Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
2.
Blood ; 140(23): 2463-2476, 2022 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-35960849

RESUMEN

Peripheral T-cell lymphoma (PTCL) is a heterogeneous group of hematological cancers arising from the malignant transformation of mature T cells. In a cohort of 28 PTCL cases, we identified recurrent overexpression of MYCN, a member of the MYC family of oncogenic transcription factors. Approximately half of all PTCL cases was characterized by a MYC expression signature. Inducible expression of MYCN in lymphoid cells in a mouse model caused T-cell lymphoma that recapitulated human PTCL with an MYC expression signature. Integration of mouse and human expression data identified EZH2 as a key downstream target of MYCN. Remarkably, EZH2 was found to be an essential cofactor for the transcriptional activation of the MYCN-driven gene expression program, which was independent of methyltransferase activity but dependent on phosphorylation by CDK1. MYCN-driven T-cell lymphoma was sensitive to EZH2 degradation or CDK1 inhibition, which displayed synergy with US Food and Drug Administration-approved histone deacetylase (HDAC) inhibitors.


Asunto(s)
Proteína Potenciadora del Homólogo Zeste 2 , Linfoma de Células T Periférico , Proteína Proto-Oncogénica N-Myc , Humanos , Proteína Potenciadora del Homólogo Zeste 2/genética , Linfoma de Células T Periférico/genética , Proteína Proto-Oncogénica N-Myc/genética
4.
J Hematol Oncol ; 14(1): 97, 2021 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-34167562

RESUMEN

BACKGROUND: T cell acute lymphoblastic leukemia (T-ALL) is a high-risk subtype that comprises 10-15% of childhood and 20-25% of adult ALL cases. Over 70% of T-ALL patients harbor activating mutations in the NOTCH1 signaling pathway and are predicted to be sensitive to gamma-secretase inhibitors. We have recently demonstrated that selective inhibition of PSEN1-containing gamma-secretase complexes can overcome the dose-limiting toxicity associated with broad gamma-secretase inhibitors. In this study, we developed combination treatment strategies with the PSEN1-selective gamma-secretase inhibitor MRK-560 and other targeted agents (kinase inhibitors ruxolitinib and imatinib; XPO-1 inhibitor KPT-8602/eltanexor) for the treatment of T-ALL. METHODS: We treated T-ALL cell lines in vitro and T-ALL patient-derived xenograft (PDX) models in vivo with MRK-560 alone or in combination with other targeted inhibitors (ruxolitinib, imatinib or KPT-8602/eltanexor). We determined effects on proliferation of the cell lines and leukemia development and survival in the PDX models. RESULTS: All NOTCH1-signaling-dependent T-ALL cell lines were sensitive to MRK-560 and its combination with ruxolitinib or imatinib in JAK1- or ABL1-dependent cell lines synergistically inhibited leukemia proliferation. We also observed strong synergy between MRK-560 and KPT-8602 (eltanexor) in all NOTCH1-dependent T-ALL cell lines. Such synergy was also observed in vivo in a variety of T-ALL PDX models with NOTCH1 or FBXW7 mutations. Combination treatment significantly reduced leukemic infiltration in vivo and resulted in a survival benefit when compared to single treatment groups. We did not observe weight loss or goblet cell hyperplasia in single drug or combination treated mice when compared to control. CONCLUSIONS: These data demonstrate that the antileukemic effect of PSEN1-selective gamma-secretase inhibition can be synergistically enhanced by the addition of other targeted inhibitors. The combination of MRK-560 with KPT-8602 is a highly effective treatment combination, which circumvents the need for the identification of additional mutations and provides a clear survival benefit in vivo. These promising preclinical data warrant further development of combination treatment strategies for T-ALL based on PSEN1-selective gamma-secretase inhibition.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Antineoplásicos/farmacología , Carioferinas/antagonistas & inhibidores , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Sulfonamidas/farmacología , Animales , Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Mesilato de Imatinib/uso terapéutico , Ratones , Terapia Molecular Dirigida , Nitrilos/uso terapéutico , Presenilina-1/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirazoles/uso terapéutico , Pirimidinas/uso terapéutico , Sulfonamidas/uso terapéutico , Proteína Exportina 1
5.
Nat Commun ; 12(1): 3705, 2021 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-34140493

RESUMEN

Peripheral T-cell lymphoma (PTCL) is a heterogeneous group of non-Hodgkin lymphomas with poor prognosis. Up to 30% of PTCL lack distinctive features and are classified as PTCL, not otherwise specified (PTCL-NOS). To further improve our understanding of the genetic landscape and biology of PTCL-NOS, we perform RNA-sequencing of 18 cases and validate results in an independent cohort of 37 PTCL cases. We identify FYN-TRAF3IP2, KHDRBS1-LCK and SIN3A-FOXO1 as new in-frame fusion transcripts, with FYN-TRAF3IP2 as a recurrent fusion detected in 8 of 55 cases. Using ex vivo and in vivo experiments, we demonstrate that FYN-TRAF3IP2 and KHDRBS1-LCK activate signaling pathways downstream of the T cell receptor (TCR) complex and confer therapeutic vulnerability to clinically available drugs.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas de Unión al ADN/genética , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/genética , Linfoma de Células T Periférico/genética , Proteínas de Fusión Oncogénica/metabolismo , Proteínas Proto-Oncogénicas c-fyn/genética , Proteínas de Unión al ARN/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Línea Celular Tumoral , Membrana Celular/metabolismo , Estudios de Cohortes , Proteínas de Unión al ADN/metabolismo , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Estimación de Kaplan-Meier , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/metabolismo , Linfoma de Células T Periférico/metabolismo , Linfoma de Células T Periférico/patología , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Proteínas de Fusión Oncogénica/genética , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Proteínas de Unión al ARN/metabolismo , RNA-Seq , Transducción de Señal/genética , Complejo Correpresor Histona Desacetilasa y Sin3/genética , Complejo Correpresor Histona Desacetilasa y Sin3/metabolismo , Proteína bcl-X/antagonistas & inhibidores , Proteína bcl-X/metabolismo
6.
Clin Cancer Res ; 26(21): 5747-5758, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32826328

RESUMEN

PURPOSE: KPT-8602 (Eltanexor) is a second-generation exportin-1 (XPO1) inhibitor with potent activity against acute lymphoblastic leukemia (ALL) in preclinical models and with minimal effects on normal cells. In this study, we evaluated whether KPT-8602 would synergize with dexamethasone, vincristine, or doxorubicin, three drugs currently used for the treatment of ALL. EXPERIMENTAL DESIGN: First, we searched for the most synergistic combination of KPT-8602 with dexamethasone, vincristine, or doxorubicin in vitro in both B-ALL and T-ALL cell lines using proliferation and apoptosis as a readout. Next, we validated this synergistic effect by treatment of clinically relevant B- and T-ALL patient-derived xenograft models in vivo. Finally, we performed RNA-sequencing (RNA-seq) and chromatin immunoprecipitation sequencing (ChIP-seq) to determine the mechanism of synergy. RESULTS: KPT-8602 showed strong synergism with dexamethasone on human B-ALL and T-ALL cell lines as well as in vivo in three patient-derived ALL xenografts. Compared with single-drug treatment, the drug combination caused increased apoptosis and led to histone depletion. Mechanistically, integration of ChIP-seq and RNA-seq data revealed that addition of KPT-8602 to dexamethasone enhanced the activity of the glucocorticoid receptor (NR3C1) and led to increased inhibition of E2F-mediated transcription. We observed strong inhibition of E2F target genes related to cell cycle, DNA replication, and transcriptional regulation. CONCLUSIONS: Our preclinical study demonstrates that KPT-8602 enhances the effects of dexamethasone to inhibit B-ALL and T-ALL cells via NR3C1- and E2F-mediated transcriptional complexes, allowing to achieve increased dexamethasone effects for patients.


Asunto(s)
Dexametasona/farmacología , Doxorrubicina/farmacología , Carioferinas/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Receptores Citoplasmáticos y Nucleares/genética , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Sinergismo Farmacológico , Humanos , Carioferinas/antagonistas & inhibidores , Ratones , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Vincristina/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína Exportina 1
7.
Blood ; 134(16): 1323-1336, 2019 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-31492675

RESUMEN

The polycomb repressive complex 2, with core components EZH2, SUZ12, and EED, is responsible for writing histone 3 lysine 27 trimethylation histone marks associated with gene repression. Analysis of sequence data from 419 T-cell acute lymphoblastic leukemia (T-ALL) cases demonstrated a significant association between SUZ12 and JAK3 mutations. Here we show that CRISPR/Cas9-mediated inactivation of Suz12 cooperates with mutant JAK3 to drive T-cell transformation and T-ALL development. Gene expression profiling integrated with ChIP-seq and ATAC-seq data established that inactivation of Suz12 led to increased PI3K/mammalian target of rapamycin (mTOR), vascular endothelial growth factor (VEGF), and WNT signaling. Moreover, a drug screen revealed that JAK3/Suz12 mutant leukemia cells were more sensitive to histone deacetylase (HDAC)6 inhibition than JAK3 mutant leukemia cells. Among the broad genome and gene expression changes observed on Suz12 inactivation, our integrated analysis identified the PI3K/mTOR, VEGF/VEGF receptor, and HDAC6/HSP90 pathways as specific vulnerabilities in T-ALL cells with combined JAK3 and SUZ12 mutations.


Asunto(s)
Transformación Celular Neoplásica/genética , Complejo Represivo Polycomb 2/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Transducción de Señal/fisiología , Animales , Humanos , Janus Quinasa 3/genética , Ratones , Mutación , Proteínas de Neoplasias , Factores de Transcripción
10.
Blood ; 131(4): 421-425, 2018 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-29187379

RESUMEN

The Janus kinase 3 (JAK3) tyrosine kinase is mutated in 10% to 16% of T-cell acute lymphoblastic leukemia (T-ALL) cases. JAK3 mutants induce constitutive JAK/STAT signaling and cause leukemia when expressed in the bone marrow cells of mice. Surprisingly, we observed that one third of JAK3-mutant T-ALL cases harbor 2 JAK3 mutations, some of which are monoallelic and others that are biallelic. Our data suggest that wild-type JAK3 competes with mutant JAK3 (M511I) for binding to the common γ chain and thereby suppresses its oncogenic potential. We demonstrate that JAK3 (M511I) can increase its limited oncogenic potential through the acquisition of an additional mutation in the mutant JAK3 allele. These double JAK3 mutants show increased STAT5 activation and increased potential to transform primary mouse pro-T cells to interleukin-7-independent growth and were not affected by wild-type JAK3 expression. These data extend our insight into the oncogenic properties of JAK3 mutations and provide an explanation of why progression of JAK3-mutant T-ALL cases can be associated with the accumulation of additional JAK3 mutations.


Asunto(s)
Janus Quinasa 3/genética , Mutación Puntual , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Transducción de Señal , Alelos , Línea Celular Tumoral , Humanos , Janus Quinasa 3/metabolismo , Modelos Moleculares , Tasa de Mutación , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo
11.
Haematologica ; 102(9): 1605-1616, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28659337

RESUMEN

Anaplastic lymphoma kinase (ALK)-positive anaplastic large cell lymphoma is characterized by 2p23/ALK aberrations, including the classic t(2;5)(p23;q35)/NPM1-ALK rearrangement present in ~80% of cases and several variant t(2p23/ALK) occurring in the remaining cases. The ALK fusion partners play a key role in the constitutive activation of the chimeric protein and its subcellular localization. Using various molecular technologies, we have characterized ALK fusions in eight recently diagnosed anaplastic large cell lymphoma cases with cytoplasmic-only ALK expression. The identified partner genes included EEF1G (one case), RNF213/ALO17 (one case), ATIC (four cases) and TPM3 (two cases). Notably, all cases showed copy number gain of the rearranged ALK gene, which is never observed in NPM1-ALK-positive lymphomas. We hypothesized that this could be due to lower expression levels and/or lower oncogenic potential of the variant ALK fusions. Indeed, all partner genes, except EEF1G, showed lower expression in normal and malignant T cells, in comparison with NPM1 In addition, we investigated the transformation potential of endogenous Npm1-Alk and Atic-Alk fusions generated by clustered regularly interspaced short palindromic repeats/Cas9 genome editing in Ba/F3 cells. We found that Npm1-Alk has a stronger transformation potential than Atic-Alk, and we observed a subclonal gain of Atic-Alk after a longer culture period, which was not observed for Npm1-Alk Taken together, our data illustrate that lymphomas driven by the variant ATIC-ALK fusion (and likely by RNF213-ALK and TPM3-ALK), but not the classic NPM1-ALK, require an increased dosage of the ALK hybrid gene to compensate for the relatively low and insufficient expression and signaling properties of the chimeric gene.


Asunto(s)
Adenosina Trifosfatasas/genética , Reordenamiento Génico , Transferasas de Hidroximetilo y Formilo/genética , Linfoma Anaplásico de Células Grandes/genética , Complejos Multienzimáticos/genética , Nucleótido Desaminasas/genética , Proteínas de Fusión Oncogénica/genética , Proteínas Tirosina Quinasas Receptoras/genética , Translocación Genética , Tropomiosina/genética , Ubiquitina-Proteína Ligasas/genética , Adolescente , Anciano , Quinasa de Linfoma Anaplásico , Preescolar , Femenino , Humanos , Linfoma Anaplásico de Células Grandes/patología , Masculino , Persona de Mediana Edad , Nucleofosmina
12.
PLoS One ; 11(1): e0146100, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26745281

RESUMEN

The four members of the epidermal growth factor receptor (EGFR/ERBB) family form homo- and heterodimers which mediate ligand-specific regulation of many key cellular processes in normal and cancer tissues. While signaling through the EGFR has been extensively studied on the molecular level, signal transduction through ERBB3/ERBB4 heterodimers is less well understood. Here, we generated isogenic mouse Ba/F3 cells that express full-length and functional membrane-integrated ERBB3 and ERBB4 or ERBB4 alone, to serve as a defined cellular model for biological and phosphoproteomics analysis of ERBB3/ERBB4 signaling. ERBB3 co-expression significantly enhanced Ba/F3 cell proliferation upon neuregulin-1 (NRG1) treatment. For comprehensive signaling studies we performed quantitative mass spectrometry (MS) experiments to compare the basal ERBB3/ERBB4 cell phosphoproteome to NRG1 treatment of ERBB3/ERBB4 and ERBB4 cells. We employed a workflow comprising differential isotope labeling with mTRAQ reagents followed by chromatographic peptide separation and final phosphopeptide enrichment prior to MS analysis. Overall, we identified 9686 phosphorylation sites which could be confidently localized to specific residues. Statistical analysis of three replicate experiments revealed 492 phosphorylation sites which were significantly changed in NRG1-treated ERBB3/ERBB4 cells. Bioinformatics data analysis recapitulated regulation of mitogen-activated protein kinase and Akt pathways, but also indicated signaling links to cytoskeletal functions and nuclear biology. Comparative assessment of NRG1-stimulated ERBB4 Ba/F3 cells revealed that ERBB3 did not trigger defined signaling pathways but more broadly enhanced phosphoproteome regulation in cells expressing both receptors. In conclusion, our data provide the first global picture of ERBB3/ERBB4 signaling and provide numerous potential starting points for further mechanistic studies.


Asunto(s)
Linfocitos B/metabolismo , Fosfoproteínas/genética , Proteoma/genética , Receptor ErbB-3/genética , Receptor ErbB-4/genética , Transducción de Señal , Secuencia de Aminoácidos , Animales , Linfocitos B/citología , Linfocitos B/efectos de los fármacos , Línea Celular , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Ingeniería Genética , Humanos , Ratones , Datos de Secuencia Molecular , Neurregulina-1/metabolismo , Neurregulina-1/farmacología , Fosfoproteínas/metabolismo , Fosforilación , Unión Proteica , Mapeo de Interacción de Proteínas , Proteoma/metabolismo , Receptor ErbB-3/metabolismo , Receptor ErbB-4/metabolismo
13.
Blood ; 124(20): 3092-100, 2014 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-25193870

RESUMEN

JAK3 is a tyrosine kinase that associates with the common γ chain of cytokine receptors and is recurrently mutated in T-cell acute lymphoblastic leukemia (T-ALL). We tested the transforming properties of JAK3 pseudokinase and kinase domain mutants using in vitro and in vivo assays. Most, but not all, JAK3 mutants transformed cytokine-dependent Ba/F3 or MOHITO cell lines to cytokine-independent proliferation. JAK3 pseudokinase mutants were dependent on Jak1 kinase activity for cellular transformation, whereas the JAK3 kinase domain mutant could transform cells in a Jak1 kinase-independent manner. Reconstitution of the IL7 receptor signaling complex in 293T cells showed that JAK3 mutants required receptor binding to mediate downstream STAT5 phosphorylation. Mice transplanted with bone marrow progenitor cells expressing JAK3 mutants developed a long-latency transplantable T-ALL-like disease, characterized by an accumulation of immature CD8(+) T cells. In vivo treatment of leukemic mice with the JAK3 selective inhibitor tofacitinib reduced the white blood cell count and caused leukemic cell apoptosis. Our data show that JAK3 mutations are drivers of T-ALL and require the cytokine receptor complex for transformation. These results warrant further investigation of JAK1/JAK3 inhibitors for the treatment of T-ALL.


Asunto(s)
Transformación Celular Neoplásica/genética , Modelos Animales de Enfermedad , Janus Quinasa 1/metabolismo , Janus Quinasa 3/genética , Leucemia de Células T/genética , Ratones , Enfermedad Aguda , Animales , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Activación Enzimática/efectos de los fármacos , Janus Quinasa 3/antagonistas & inhibidores , Janus Quinasa 3/metabolismo , Leucemia de Células T/tratamiento farmacológico , Leucemia de Células T/metabolismo , Leucemia de Células T/patología , Masculino , Ratones/genética , Ratones/metabolismo , Ratones Endogámicos BALB C , Mutación , Piperidinas/uso terapéutico , Pirimidinas/uso terapéutico , Pirroles/uso terapéutico , Transducción de Señal/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Linfocitos T/patología
14.
Eur J Med Chem ; 63: 713-21, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23567961

RESUMEN

FLT3 and PDGFR tyrosine kinases are important targets for therapy of different types of leukemia. Several FLT3/PDGFR inhibitors are currently under clinical investigation for combination with standard therapy for treatment of acute myeloid leukemia (AML), however these agents only induce partial remission and development of resistance has been reported. In this work we describe the identification of potent and novel dual FLT3/PDGFR inhibitors that resulted from our efforts to screen a library of 25,607 small molecules against the FLT3 dependent cell line MOLM-13 and the PDGFR dependent cell line EOL-1. This effort led to the identification of five compounds that were confirmed to be active on additional FLT3 dependent cell lines (cellular EC50 values between 35 and 700 nM), while having no significant effect on 24 other tyrosine kinases.


Asunto(s)
Inhibidores de Proteínas Quinasas/farmacología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Tirosina Quinasa 3 Similar a fms/antagonistas & inhibidores , Western Blotting , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Síndrome Hipereosinofílico/metabolismo , Síndrome Hipereosinofílico/patología , Leucemia Monocítica Aguda/metabolismo , Leucemia Monocítica Aguda/patología , Estructura Molecular , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/química , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad , Tirosina Quinasa 3 Similar a fms/metabolismo
15.
Nat Genet ; 45(2): 186-90, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23263491

RESUMEN

T-cell acute lymphoblastic leukemia (T-ALL) is caused by the cooperation of multiple oncogenic lesions. We used exome sequencing on 67 T-ALLs to gain insight into the mutational spectrum in these leukemias. We detected protein-altering mutations in 508 genes, with an average of 8.2 mutations in pediatric and 21.0 mutations in adult T-ALL. Using stringent filtering, we predict seven new oncogenic driver genes in T-ALL. We identify CNOT3 as a tumor suppressor mutated in 7 of 89 (7.9%) adult T-ALLs, and its knockdown causes tumors in a sensitized Drosophila melanogaster model. In addition, we identify mutations affecting the ribosomal proteins RPL5 and RPL10 in 12 of 122 (9.8%) pediatric T-ALLs, with recurrent alterations of Arg98 in RPL10. Yeast and lymphoid cells expressing the RPL10 Arg98Ser mutant showed a ribosome biogenesis defect. Our data provide insights into the mutational landscape of pediatric versus adult T-ALL and identify the ribosome as a potential oncogenic factor.


Asunto(s)
Exoma/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Proteínas Ribosómicas/genética , Factores de Transcripción/genética , Animales , Secuencia de Bases , Drosophila melanogaster , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Ratones , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación/genética , Polirribosomas/genética , Interferencia de ARN , Proteína Ribosómica L10 , Saccharomyces cerevisiae , Alineación de Secuencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...