Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 320
Filtrar
1.
Protein Sci ; 33(6): e5019, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38747396

RESUMEN

AF9 (MLLT3) and its paralog ENL(MLLT1) are members of the YEATS family of proteins with important role in transcriptional and epigenetic regulatory complexes. These proteins are two common MLL fusion partners in MLL-rearranged leukemias. The oncofusion proteins MLL-AF9/ENL recruit multiple binding partners, including the histone methyltransferase DOT1L, leading to aberrant transcriptional activation and enhancing the expression of a characteristic set of genes that drive leukemogenesis. The interaction between AF9 and DOT1L is mediated by an intrinsically disordered C-terminal ANC1 homology domain (AHD) in AF9, which undergoes folding upon binding of DOT1L and other partner proteins. We have recently reported peptidomimetics that disrupt the recruitment of DOT1L by AF9 and ENL, providing a proof-of-concept for targeting AHD and assessing its druggability. Intrinsically disordered proteins, such as AF9 AHD, are difficult to study and characterize experimentally on a structural level. In this study, we present a successful protein engineering strategy to facilitate structural investigation of the intrinsically disordered AF9 AHD domain in complex with peptidomimetic inhibitors by using maltose binding protein (MBP) as a crystallization chaperone connected with linkers of varying flexibility and length. The strategic incorporation of disulfide bonds provided diffraction-quality crystals of the two disulfide-bridged MBP-AF9 AHD fusion proteins in complex with the peptidomimetics. These successfully determined first series of 2.1-2.6 Å crystal complex structures provide high-resolution insights into the interactions between AHD and its inhibitors, shedding light on the role of AHD in recruiting various binding partner proteins. We show that the overall complex structures closely resemble the reported NMR structure of AF9 AHD/DOT1L with notable difference in the conformation of the ß-hairpin region, stabilized through conserved hydrogen bonds network. These first series of AF9 AHD/peptidomimetics complex structures are providing insights of the protein-inhibitor interactions and will facilitate further development of novel inhibitors targeting the AF9/ENL AHD domain.


Asunto(s)
Peptidomiméticos , Peptidomiméticos/química , Peptidomiméticos/metabolismo , Humanos , Proteínas Intrínsecamente Desordenadas/química , Proteínas Intrínsecamente Desordenadas/metabolismo , Proteínas Intrínsecamente Desordenadas/genética , Proteínas Intrínsecamente Desordenadas/antagonistas & inhibidores , N-Metiltransferasa de Histona-Lisina/química , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , N-Metiltransferasa de Histona-Lisina/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , Modelos Moleculares , Proteínas de Fusión Oncogénica/química , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Cristalografía por Rayos X , Dominios Proteicos , Proteína de la Leucemia Mieloide-Linfoide/química , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteína de la Leucemia Mieloide-Linfoide/antagonistas & inhibidores
2.
Anal Chim Acta ; 1287: 342108, 2024 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-38182385

RESUMEN

Accurate detection and classification of the three isoforms of PML/RARA genomic fragments are crucial for predicting disease progression, stratifying risk, and administering precise drug therapies in acute promyelocytic leukemia (APL). In this study, we have developed a highly specific nucleic acid detection platform capable of quantifying the long isoform of the three main PML-RARA isoforms at a constant temperature. This platform integrates the strengths of the CRISPR/Cas12a nuclease-based method and the rolling circle amplification (RCA) technique. Notably, the RCA-assisted CRISPR/Cas12a trans-cleavage system incorporates a spatial confinement effect by utilizing intermolecular G-quadruplex structures. This innovative design effectively enhances the local concentration of CRISPR/Cas12a, thereby accelerating its cleaving efficiency towards reporter nucleic acids and enabling the detection of PML/RARA fusion gene expression through spectroscopy. The robust detection of PML/RARA fusion gene from human serum samples validates the reliability and potential of this platform in the screening, diagnosis, and prognosis of APL cases. Our findings present an approach that holds significant potential for the further development of the robust CRISPR/Cas sensor system, offering a rapid and adaptable paradigm for APL diagnosis.


Asunto(s)
Sistemas CRISPR-Cas , G-Cuádruplex , Proteínas de Fusión Oncogénica , Humanos , Sistemas CRISPR-Cas/genética , Progresión de la Enfermedad , Endonucleasas , Isoformas de Proteínas , Reproducibilidad de los Resultados , Proteínas de Fusión Oncogénica/química
3.
Proteins ; 90(1): 282-298, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34414607

RESUMEN

Disruptor of telomeric silencing 1-like (DOT1L) is the only non-SET domain histone lysine methyltransferase (KMT) and writer of H3K79 methylation on nucleosomes marked by H2B ubiquitination. DOT1L has elicited significant attention because of its interaction or fusion with members of the AF protein family in blood cell biology and leukemogenic transformation. Here, our goal was to extend previous structural information by performing a robust molecular dynamic study of DOT1L and its leukemogenic partners combined with mutational analysis. We show that statically and dynamically, D161, G163, E186, and F223 make frequent time-dependent interactions with SAM, while additional residues T139, K187, and N241 interact with SAM only under dynamics. Dynamics models reveal DOT1L, SAM, and H4 moving as one and show that more than twice the number of DOT1L residues interacts with these partners, relative to the static structure. Mutational analyses indicate that six of these residues are intolerant to substitution. We describe the dynamic behavior of DOT1L interacting with AF10 and AF9. Studies on the dynamics of a heterotrimeric complex of DOT1L1-AF10 illuminated describe coordinated motions that impact the relative position of the DOT1L HMT domain to the nucleosome. The molecular motions of the DOT1L-AF9 complex are less extensive and highly dynamic, resembling a swivel-like mechanics. Through molecular dynamics and mutational analysis, we extend the knowledge previous provided by static measurements. These results are important to consider when describing the biochemical properties of DOT1L, under normal and in disease conditions, as well as for the development of novel therapeutic agents.


Asunto(s)
Carcinogénesis , N-Metiltransferasa de Histona-Lisina , Leucemia/metabolismo , Carcinogénesis/química , Carcinogénesis/metabolismo , N-Metiltransferasa de Histona-Lisina/química , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Simulación de Dinámica Molecular , Nucleosomas/química , Nucleosomas/metabolismo , Proteínas de Fusión Oncogénica/química , Proteínas de Fusión Oncogénica/metabolismo , S-Adenosilmetionina/química , S-Adenosilmetionina/metabolismo
4.
Theranostics ; 11(17): 8172-8184, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34373735

RESUMEN

Chromosome translocations involving mixed lineage leukemia (MLL) gene cause acute leukemia with a poor prognosis. MLL is frequently fused with transcription cofactors AF4 (~35%), AF9 (25%) or its paralog ENL (10%). The AHD domain of AF9/ENL binds to AF4, its paralog AFF4, or histone-H3 lysine-79 (H3K79) methyltransferase DOT1L. Formation of AF9/ENL/AF4/AFF4-containing super elongation complexes (SEC) and the catalytic activity of DOT1L are essential for MLL-rearranged leukemia. Protein-protein interactions (PPI) between AF9/ENL and DOT1L/AF4/AFF4 are therefore a potential drug target. Methods: Compound screening followed by medicinal chemistry was used to find inhibitors of such PPIs, which were examined for their biological activities against MLL-rearranged leukemia and other cancer cells. Results: Compound-1 was identified to be a novel small-molecule inhibitor of the AF9/ENL-DOT1L/AF4/AFF4 interaction with IC50s of 0.9-3.5 µM. Pharmacological inhibition of the PPIs significantly reduced SEC and DOT1L-mediated H3K79 methylation in the leukemia cells. Gene profiling shows compound-1 significantly suppressed the gene signatures related to onco-MLL, DOT1L, HoxA9 and Myc. It selectively inhibited proliferation of onco-MLL- or Myc-driven cancer cells and induced cell differentiation and apoptosis. Compound-1 exhibited strong antitumor activity in a mouse model of MLL-rearranged leukemia. Conclusions: The AF9/ENL-DOT1L/AF4/AFF4 interactions are validated to be an anticancer target and compound-1 is a useful in vivo probe for biological studies as well as a pharmacological lead for further drug development.


Asunto(s)
Antineoplásicos/farmacología , Leucemia Mieloide Aguda , Proteínas de Fusión Oncogénica , Animales , Expresión Génica/efectos de los fármacos , N-Metiltransferasa de Histona-Lisina/química , N-Metiltransferasa de Histona-Lisina/efectos de los fármacos , N-Metiltransferasa de Histona-Lisina/genética , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Ratones , Proteínas de Fusión Oncogénica/química , Proteínas de Fusión Oncogénica/efectos de los fármacos , Proteínas de Fusión Oncogénica/genética , Oncogenes/efectos de los fármacos , Dominios y Motivos de Interacción de Proteínas , Factores de Elongación Transcripcional/química , Factores de Elongación Transcripcional/efectos de los fármacos , Factores de Elongación Transcripcional/genética
5.
Cancer Res ; 81(19): 4939-4948, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34385184

RESUMEN

Chromosomal translocations generate oncogenic fusion proteins in approximately one-third of sarcomas, but how these proteins promote tumorigenesis is not well understood. Interestingly, some translocation-driven cancers exhibit dramatic clinical responses to therapy, such as radiotherapy, although the precise mechanism has not been elucidated. Here we reveal a molecular mechanism by which the fusion oncoprotein FUS-CHOP promotes tumor maintenance that also explains the remarkable sensitivity of myxoid liposarcomas to radiation therapy. FUS-CHOP interacted with chromatin remodeling complexes to regulate sarcoma cell proliferation. One of these chromatin remodelers, SNF2H, colocalized with FUS-CHOP genome-wide at active enhancers. Following ionizing radiation, DNA damage response kinases phosphorylated the prion-like domain of FUS-CHOP to impede these protein-protein interactions, which are required for transformation. Therefore, the DNA damage response after irradiation disrupted oncogenic targeting of chromatin remodelers required for FUS-CHOP-driven sarcomagenesis. This mechanism of disruption links phosphorylation of the prion-like domain of an oncogenic fusion protein to DNA damage after ionizing radiation and reveals that a dependence on oncogenic chromatin remodeling underlies sensitivity to radiation therapy in myxoid liposarcoma. SIGNIFICANCE: Prion-like domains, which are frequently translocated in cancers as oncogenic fusion proteins that drive global epigenetic changes, confer sensitivity to radiation via disruption of oncogenic interactions.


Asunto(s)
Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Dominios y Motivos de Interacción de Proteínas , Proteína FUS de Unión a ARN/genética , Proteína FUS de Unión a ARN/metabolismo , Radiación Ionizante , Factor de Transcripción CHOP/genética , Factor de Transcripción CHOP/metabolismo , Sitios de Unión , Línea Celular Tumoral , Transformación Celular Neoplásica/efectos de la radiación , Ensamble y Desensamble de Cromatina , Secuenciación de Inmunoprecipitación de Cromatina , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas de Fusión Oncogénica/química , Fosforilación/efectos de la radiación , Unión Proteica , Proteína FUS de Unión a ARN/química , Sarcoma/etiología , Sarcoma/metabolismo , Sarcoma/patología , Factor de Transcripción CHOP/química , Translocación Genética
6.
Mol Ther ; 29(11): 3305-3318, 2021 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-34274537

RESUMEN

FGFR3-TACC3 (F3-T3) gene fusions are regarded as a "low-hanging fruit" paradigm for precision therapy in human glioblastoma (GBM). Small molecules designed to target the kinase in FGFR currently serve as one form of potential treatment but cause off-target effects and toxicity. Here, CRISPR-Cas13a, which is known to directly suppress gene expression at the transcriptional level and induce a collateral effect in eukaryotes, was leveraged as a possible precision therapy in cancer cells harboring F3-T3 fusion genes. A library consisting of crRNAs targeting the junction site of F3-T3 was designed, and an in silico simulation scheme was created to select the optimal crRNA candidates. An optimal crRNA, crRNA1, showed efficiency and specificity in inducing the collateral effect in only U87 cells expressing F3-T3 (U87-F3-T3). Expression profiles obtained with microarray analysis were consistent with induction of the collateral effect by the CRISPR-Cas13a system. Tumor cell proliferation and colony formation were decreased in U87-F3-T3 cells expressing the Cas13a-based tool, and tumor growth was suppressed in an orthotopic tumor model in mice. These findings demonstrate that the CRISPR-Cas13a system induces the collateral damage effect in cancer cells and provides a viable strategy for precision tumor therapy based on the customized design of a CRISPR-Cas13a-based tool against F3-T3 fusion genes.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Glioblastoma/genética , Proteínas Asociadas a Microtúbulos/genética , Proteínas de Fusión Oncogénica/genética , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética , Animales , Biomarcadores de Tumor , Línea Celular Tumoral , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Expresión Génica , Perfilación de la Expresión Génica , Glioblastoma/patología , Xenoinjertos , Humanos , Enlace de Hidrógeno , Ratones , Proteínas Asociadas a Microtúbulos/química , Modelos Moleculares , Conformación de Ácido Nucleico , Proteínas de Fusión Oncogénica/química , Unión Proteica , Conformación Proteica , ARN Mensajero/química , ARN Mensajero/genética , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/química
7.
Cancer Cell ; 39(9): 1262-1278.e7, 2021 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-34329586

RESUMEN

Fusion-transcription factors (fusion-TFs) represent a class of driver oncoproteins that are difficult to therapeutically target. Recently, protein degradation has emerged as a strategy to target these challenging oncoproteins. The mechanisms that regulate fusion-TF stability, however, are generally unknown. Using CRISPR-Cas9 screening, we discovered tripartite motif-containing 8 (TRIM8) as an E3 ubiquitin ligase that ubiquitinates and degrades EWS/FLI, a driver fusion-TF in Ewing sarcoma. Moreover, we identified TRIM8 as a selective dependency in Ewing sarcoma compared with >700 other cancer cell lines. Mechanistically, TRIM8 knockout led to an increase in EWS/FLI protein levels that was not tolerated. EWS/FLI acts as a neomorphic substrate for TRIM8, defining the selective nature of the dependency. Our results demonstrate that fusion-TF protein stability is tightly regulated and highlight fusion oncoprotein-specific regulators as selective therapeutic targets. This study provides a tractable strategy to therapeutically exploit oncogene overdose in Ewing sarcoma and potentially other fusion-TF-driven cancers.


Asunto(s)
Neoplasias Óseas/mortalidad , Proteínas Portadoras/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Fusión Oncogénica/química , Proteína Proto-Oncogénica c-fli-1/química , Proteína Proto-Oncogénica c-fli-1/metabolismo , Proteína EWS de Unión a ARN/química , Proteína EWS de Unión a ARN/metabolismo , Sarcoma de Ewing/mortalidad , Neoplasias Óseas/metabolismo , Proteínas Portadoras/genética , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Técnicas de Inactivación de Genes , Células HEK293 , Humanos , Proteínas de Microfilamentos/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas de Fusión Oncogénica/metabolismo , Estabilidad Proteica , Proteolisis , Sarcoma de Ewing/metabolismo , Transactivadores/metabolismo
8.
Protein Sci ; 30(7): 1454-1466, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34018649

RESUMEN

Fusion transcription factors generated by genomic translocations are common drivers of several types of cancers including sarcomas and leukemias. Oncofusions of the FET (FUS, EWSR1, and TAF15) family proteins result from the fusion of the prion-like domain (PLD) of FET proteins to the DNA-binding domain (DBD) of certain transcription regulators and are implicated in aberrant transcriptional programs through interactions with chromatin remodelers. Here, we show that FUS-DDIT3, a FET oncofusion protein, undergoes PLD-mediated phase separation into liquid-like condensates. Nuclear FUS-DDIT3 condensates can recruit essential components of the global transcriptional machinery such as the chromatin remodeler SWI/SNF. The recruitment of mammalian SWI/SNF (mSWI/SNF) is driven by heterotypic PLD-PLD interactions between FUS-DDIT3 and core subunits of SWI/SNF, such as the catalytic component BRG1. Further experiments with single-molecule correlative force-fluorescence microscopy support a model wherein the fusion protein forms condensates on DNA surface and enrich BRG1 to activate transcription by ectopic chromatin remodeling. Similar PLD-driven co-condensation of mSWI/SNF with transcription factors can be employed by other oncogenic fusion proteins with a generic PLD-DBD domain architecture for global transcriptional reprogramming.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Priones/metabolismo , Proteína FUS de Unión a ARN/metabolismo , Factores de Transcripción/metabolismo , Línea Celular , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/genética , Humanos , Microscopía Fluorescente , Proteínas de Fusión Oncogénica/química , Proteínas de Fusión Oncogénica/genética , Priones/química , Priones/genética , Dominios Proteicos , Proteína FUS de Unión a ARN/química , Proteína FUS de Unión a ARN/genética , Factores de Transcripción/química , Factores de Transcripción/genética
9.
Clin Cancer Res ; 27(10): 2899-2909, 2021 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-33685866

RESUMEN

PURPOSE: Current standard initial therapy for advanced, ROS proto-oncogene 1, receptor tyrosine kinase fusion (ROS1)-positive (ROS1+) non-small cell lung cancer (NSCLC) is crizotinib or entrectinib. Lorlatinib, a next-generation anaplastic lymphoma kinase/ROS1 inhibitor, recently demonstrated efficacy in ROS1+ NSCLC, including in crizotinib-pretreated patients. However, mechanisms of lorlatinib resistance in ROS1+ disease remain poorly understood. Here, we assessed mechanisms of resistance to crizotinib and lorlatinib. EXPERIMENTAL DESIGN: Biopsies from patients with ROS1 + NSCLC progressing on crizotinib or lorlatinib were profiled by genetic sequencing. RESULTS: From 55 patients, 47 post-crizotinib and 32 post-lorlatinib biopsies were assessed. Among 42 post-crizotinib and 28 post-lorlatinib biopsies analyzed at distinct timepoints, ROS1 mutations were identified in 38% and 46%, respectively. ROS1 G2032R was the most commonly occurring mutation in approximately one third of cases. Additional ROS1 mutations included D2033N (2.4%) and S1986F (2.4%) post-crizotinib and L2086F (3.6%), G2032R/L2086F (3.6%), G2032R/S1986F/L2086F (3.6%), and S1986F/L2000V (3.6%) post-lorlatinib. Structural modeling predicted ROS1L2086F causes steric interference to lorlatinib, crizotinib, and entrectinib, while it may accommodate cabozantinib. In Ba/F3 models, ROS1L2086F, ROS1G2032R/L2086F, and ROS1S1986F/G2032R/L2086F were refractory to lorlatinib but sensitive to cabozantinib. A patient with disease progression on crizotinib and lorlatinib and ROS1 L2086F received cabozantinib for nearly 11 months with disease control. Among lorlatinib-resistant biopsies, we also identified MET amplification (4%), KRAS G12C (4%), KRAS amplification (4%), NRAS mutation (4%), and MAP2K1 mutation (4%). CONCLUSIONS: ROS1 mutations mediate resistance to crizotinib and lorlatinib in more than one third of cases, underscoring the importance of developing next-generation ROS1 inhibitors with potency against these mutations, including G2032R and L2086F. Continued efforts are needed to elucidate ROS1-independent resistance mechanisms.


Asunto(s)
Aminopiridinas/farmacología , Crizotinib/farmacología , Resistencia a Antineoplásicos/genética , Lactamas/farmacología , Neoplasias Pulmonares/genética , Proteínas de Fusión Oncogénica/genética , Proteínas Tirosina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Pirazoles/farmacología , Adulto , Anciano , Anciano de 80 o más Años , Alelos , Sustitución de Aminoácidos , Aminopiridinas/química , Aminopiridinas/uso terapéutico , Antígenos de Diferenciación de Linfocitos B/genética , Biopsia , Línea Celular Tumoral , Crizotinib/química , Crizotinib/uso terapéutico , Femenino , Antígenos de Histocompatibilidad Clase II/genética , Humanos , Lactamas/química , Lactamas/uso terapéutico , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/tratamiento farmacológico , Masculino , Persona de Mediana Edad , Modelos Moleculares , Mutación , Proteínas de Fusión Oncogénica/química , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/química , Proteínas Proto-Oncogénicas/química , Pirazoles/química , Pirazoles/uso terapéutico , Relación Estructura-Actividad , Adulto Joven
10.
J Comput Aided Mol Des ; 35(5): 629-642, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33748935

RESUMEN

Paired box 8 (PAX8)-peroxisome proliferator-activated receptor γ (PPARγ) rearrangement is believed to play an important role in tumorigenesis of PAX8-PPARγ fusion protein (PPFP) thyroid carcinomas, while without establishing any standard treatment, including drugs. Although PPFP is a potential promising target for therapeutic agents, the three-dimensional (3D) structure and functions have not yet been experimentally elucidated. In this study, we aimed to construct the 3D structure of PPFP and to aid in the development of therapies that can target PPFP for thyroid carcinomas. The 3D structure of PPFP was constructed by homology modeling based on crystallographic structure data. To validate the modeled structure, we analyzed the thermal fluctuations by molecular dynamics simulations and predicted the physical properties using bioinformatic analyses. We found that the modeled structure was stable under hydrated conditions and had features indicating the actual existence of the structure. Furthermore, the binding free energies of the ligand rosiglitazone with PPARγ and PPFP were evaluated by the molecular mechanics-Poisson-Boltzmann surface area method. We found that rosiglitazone has different binding affinities for the same binding pockets of PPARγ and PPFP, and the optimal compound for PPFP can differ from that of PPARγ. This suggests the need for the development of drugs targeting PPFP that allow for the fusion, rather than focusing on the PPARγ side of PPFP and searching for the best compounds for that pocket. Our findings are expected to lead to the development of new therapies for thyroid tumors.


Asunto(s)
Descubrimiento de Drogas , Proteínas de Fusión Oncogénica/química , Conformación Proteica/efectos de los fármacos , Neoplasias de la Tiroides/tratamiento farmacológico , Antineoplásicos/farmacología , Sitios de Unión/efectos de los fármacos , Humanos , Hipoglucemiantes/farmacología , Ligandos , Modelos Moleculares , Proteínas de Fusión Oncogénica/antagonistas & inhibidores , Proteínas de Fusión Oncogénica/metabolismo , Rosiglitazona/farmacología , Termodinámica , Neoplasias de la Tiroides/metabolismo
11.
Nat Struct Mol Biol ; 28(2): 190-201, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33479542

RESUMEN

NUP98 fusion proteins cause leukemia via unknown molecular mechanisms. All NUP98 fusion proteins share an intrinsically disordered region (IDR) in the NUP98 N terminus, featuring repeats of phenylalanine-glycine (FG), and C-terminal fusion partners often function in gene control. We investigated whether mechanisms of oncogenic transformation by NUP98 fusion proteins are hardwired in their protein interactomes. Affinity purification coupled to mass spectrometry (MS) and confocal imaging of five NUP98 fusion proteins expressed in human leukemia cells revealed that shared interactors were enriched for proteins involved in biomolecular condensation and that they colocalized with NUP98 fusion proteins in nuclear puncta. We developed biotinylated isoxazole-mediated condensome MS (biCon-MS) to show that NUP98 fusion proteins alter the global composition of biomolecular condensates. An artificial FG-repeat-containing fusion protein phenocopied the nuclear localization patterns of NUP98 fusion proteins and their capability to drive oncogenic gene expression programs. Thus, we propose that IDR-containing fusion proteins combine biomolecular condensation with transcriptional control to induce cancer.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas de Homeodominio , Leucemia , Proteínas de Complejo Poro Nuclear , Proteínas de Fusión Oncogénica , Animales , Expresión Génica , Regulación Leucémica de la Expresión Génica , Células HEK293 , Células HL-60 , Proteínas de Homeodominio/química , Proteínas de Homeodominio/fisiología , Humanos , Leucemia/metabolismo , Leucemia/patología , Ratones , Células 3T3 NIH , Proteínas de Complejo Poro Nuclear/química , Proteínas de Complejo Poro Nuclear/fisiología , Proteínas de Fusión Oncogénica/química , Proteínas de Fusión Oncogénica/fisiología
12.
Nat Struct Mol Biol ; 27(9): 836-845, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32747783

RESUMEN

Interactions between chromatin-associated proteins and the histone landscape play major roles in dictating genome topology and gene expression. Cancer-specific fusion oncoproteins, which display unique chromatin localization patterns, often lack classical DNA-binding domains, presenting challenges in identifying mechanisms governing their site-specific chromatin targeting and function. Here we identify a minimal region of the human SS18-SSX fusion oncoprotein (the hallmark driver of synovial sarcoma) that mediates a direct interaction between the mSWI/SNF complex and the nucleosome acidic patch. This binding results in altered mSWI/SNF composition and nucleosome engagement, driving cancer-specific mSWI/SNF complex targeting and gene expression. Furthermore, the C-terminal region of SSX confers preferential affinity to repressed, H2AK119Ub-marked nucleosomes, underlying the selective targeting to polycomb-marked genomic regions and synovial sarcoma-specific dependency on PRC1 function. Together, our results describe a functional interplay between a key nucleosome binding hub and a histone modification that underlies the disease-specific recruitment of a major chromatin remodeling complex.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Histonas/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/metabolismo , Sarcoma Sinovial/metabolismo , Factores de Transcripción/metabolismo , Ubiquitinas/metabolismo , Línea Celular Tumoral , Proteínas Cromosómicas no Histona/química , Células HEK293 , Humanos , Modelos Moleculares , Proteínas de Neoplasias/química , Nucleosomas/metabolismo , Nucleosomas/patología , Proteínas de Fusión Oncogénica/química , Conformación Proteica , Proteínas Proto-Oncogénicas/química , Proteínas Represoras/química , Sarcoma Sinovial/patología , Factores de Transcripción/química , Ubiquitinación
13.
Genes Dev ; 34(15-16): 1051-1064, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32675324

RESUMEN

YAP1 is a transcriptional coactivator and the principal effector of the Hippo signaling pathway, which is causally implicated in human cancer. Several YAP1 gene fusions have been identified in various human cancers and identifying the essential components of this family of gene fusions has significant therapeutic value. Here, we show that the YAP1 gene fusions YAP1-MAMLD1, YAP1-FAM118B, YAP1-TFE3, and YAP1-SS18 are oncogenic in mice. Using reporter assays, RNA-seq, ChIP-seq, and loss-of-function mutations, we can show that all of these YAP1 fusion proteins exert TEAD-dependent YAP activity, while some also exert activity of the C'-terminal fusion partner. The YAP activity of the different YAP1 fusions is resistant to negative Hippo pathway regulation due to constitutive nuclear localization and resistance to degradation of the YAP1 fusion proteins. Genetic disruption of the TEAD-binding domain of these oncogenic YAP1 fusions is sufficient to inhibit tumor formation in vivo, while pharmacological inhibition of the YAP1-TEAD interaction inhibits the growth of YAP1 fusion-expressing cell lines in vitro. These results highlight TEAD-dependent YAP activity found in these gene fusions as critical for oncogenesis and implicate these YAP functions as potential therapeutic targets in YAP1 fusion-positive tumors.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Carcinogénesis/genética , Proteínas de Fusión Oncogénica/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Células Cultivadas , Regulación de la Expresión Génica , Humanos , Ratones , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Señales de Localización Nuclear , Motivos de Nucleótidos , Proteínas de Fusión Oncogénica/antagonistas & inhibidores , Proteínas de Fusión Oncogénica/química , Complejo de la Endopetidasa Proteasomal/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Transcripción Genética
14.
J Vis Exp ; (160)2020 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-32658189

RESUMEN

Many cancers are characterized by chromosomal translocations which result in the expression of oncogenic fusion transcription factors. Typically, these proteins contain an intrinsically disordered domain (IDD) fused with the DNA-binding domain (DBD) of another protein and orchestrate widespread transcriptional changes to promote malignancy. These fusions are often the sole recurring genomic aberration in the cancers they cause, making them attractive therapeutic targets. However, targeting oncogenic transcription factors requires a better understanding of the mechanistic role that low-complexity, IDDs play in their function. The N-terminal domain of EWSR1 is an IDD involved in a variety of oncogenic fusion transcription factors, including EWS/FLI, EWS/ATF, and EWS/WT1. Here, we use RNA-sequencing to investigate the structural features of the EWS domain important for transcriptional function of EWS/FLI in Ewing sarcoma. First shRNA-mediated depletion of the endogenous fusion from Ewing sarcoma cells paired with ectopic expression of a variety of EWS-mutant constructs is performed. Then RNA-sequencing is used to analyze the transcriptomes of cells expressing these constructs to characterize the functional deficits associated with mutations in the EWS domain. By integrating the transcriptomic analyses with previously published information about EWS/FLI DNA binding motifs, and genomic localization, as well as functional assays for transforming ability, we were able to identify structural features of EWS/FLI important for oncogenesis and define a novel set of EWS/FLI target genes critical for Ewing sarcoma. This paper demonstrates the use of RNA-sequencing as a method to map the structure-function relationship of the intrinsically disordered domain of oncogenic transcription factors.


Asunto(s)
Perfilación de la Expresión Génica , Proteínas de Fusión Oncogénica/química , Proteínas de Fusión Oncogénica/metabolismo , Proteína Proto-Oncogénica c-fli-1/química , Proteína Proto-Oncogénica c-fli-1/metabolismo , Proteína EWS de Unión a ARN/química , Proteína EWS de Unión a ARN/metabolismo , Relación Estructura-Actividad , Sitios de Unión , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Humanos , Mutación , Proteínas de Fusión Oncogénica/genética , Dominios Proteicos , Proteína Proto-Oncogénica c-fli-1/genética , Proteína EWS de Unión a ARN/genética , Sarcoma de Ewing/genética , Sarcoma de Ewing/metabolismo , Sarcoma de Ewing/patología
15.
Blood ; 136(1): 11-23, 2020 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-32276273

RESUMEN

E2A, a basic helix-loop-helix transcription factor, plays a crucial role in determining tissue-specific cell fate, including differentiation of B-cell lineages. In 5% of childhood acute lymphoblastic leukemia (ALL), the t(1,19) chromosomal translocation specifically targets the E2A gene and produces an oncogenic E2A-PBX1 fusion protein. Although previous studies have shown the oncogenic functions of E2A-PBX1 in cell and animal models, the E2A-PBX1-enforced cistrome, the E2A-PBX1 interactome, and related mechanisms underlying leukemogenesis remain unclear. Here, by unbiased genomic profiling approaches, we identify the direct target sites of E2A-PBX1 in t(1,19)-positive pre-B ALL cells and show that, compared with normal E2A, E2A-PBX1 preferentially binds to a subset of gene loci cobound by RUNX1 and gene-activating machineries (p300, MED1, and H3K27 acetylation). Using biochemical analyses, we further document a direct interaction of E2A-PBX1, through a region spanning the PBX1 homeodomain, with RUNX1. Our results also show that E2A-PBX1 binding to gene enhancers is dependent on the RUNX1 interaction but not the DNA-binding activity harbored within the PBX1 homeodomain of E2A-PBX1. Transcriptome analyses and cell transformation assays further establish a significant RUNX1 requirement for E2A-PBX1-mediated target gene activation and leukemogenesis. Notably, the RUNX1 locus itself is also directly activated by E2A-PBX1, indicating a multilayered interplay between E2A-PBX1 and RUNX1. Collectively, our study provides the first unbiased profiling of the E2A-PBX1 cistrome in pre-B ALL cells and reveals a previously unappreciated pathway in which E2A-PBX1 acts in concert with RUNX1 to enforce transcriptome alterations for the development of pre-B ALL.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/fisiología , Regulación Leucémica de la Expresión Génica/genética , Proteínas de Homeodominio/fisiología , Proteínas de Neoplasias/metabolismo , Proteínas de Fusión Oncogénica/fisiología , Secuencias de Aminoácidos , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/química , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , ADN/metabolismo , Elementos de Facilitación Genéticos , Código de Histonas , Proteínas de Homeodominio/química , Humanos , Complejo Mediador/metabolismo , Proteínas de Fusión Oncogénica/química , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , Dominios Proteicos , Mapeo de Interacción de Proteínas , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Neoplásico/biosíntesis , ARN Neoplásico/genética , Relación Estructura-Actividad , Transcriptoma , Factores de Transcripción p300-CBP/metabolismo
16.
PLoS One ; 15(4): e0232036, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32343715

RESUMEN

The NUP98 and NUP214 nucleoporins (NUPs) are recurrently fused to heterologous proteins in leukemia. The resulting chimeric oncoproteins retain the phenylalanine-glycine (FG) repeat motifs of the NUP moiety that mediate interaction with the nuclear export receptor Crm1. NUP fusion leukemias are characterized by HOXA gene upregulation; however, their molecular pathogenesis remains poorly understood. To investigate the role of Crm1 in mediating the leukemogenic properties of NUP chimeric proteins, we took advantage of the Sequestosome-1 (SQSTM1)-NUP214 fusion. SQSTM1-NUP214 retains only a short C-terminal portion of NUP214 which contains FG motifs that mediate interaction with Crm1. We introduced point mutations targeting these FG motifs and found that the ability of the resulting SQSTM1-NUP214FGmut protein to interact with Crm1 was reduced by more than 50% compared with SQSTM1-NUP214. Mutation of FG motifs affected transforming potential: while SQSTM1-NUP214 impaired myeloid maturation and conferred robust colony formation to transduced hematopoietic progenitors in a serial replating assay, the effect of SQSTM1-NUP214FGmut was considerably diminished. Moreover, SQSTM1-NUP214 caused myeloid leukemia in all transplanted mice, whereas none of the SQSTM1-NUP214FGmut reconstituted mice developed leukemia. These oncogenic effects coincided with the ability of SQSTM1-NUP214 and SQSTM1-NUP214FGmut to upregulate the expression of Hoxa and Meis1 genes in hematopoietic progenitors. Indeed, chromatin immunoprecipitation assays demonstrated that impaired SQSTM1-NUP214 interaction with Crm1 correlated with impaired binding of the fusion protein to Hoxa and Meis1 genes. These findings highlight the importance of Crm1 in mediating the leukemogenic properties of SQSTM1-NUP214, and suggest a conserved role of Crm1 in recruiting oncoproteins to their effector genes.


Asunto(s)
Proteínas de Homeodominio/genética , Carioferinas/metabolismo , Leucemia/metabolismo , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/genética , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Fusión Oncogénica/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteína Sequestosoma-1/genética , Secuencias de Aminoácidos , Animales , Línea Celular , Regulación Neoplásica de la Expresión Génica , Leucemia/genética , Leucemia/patología , Ratones , Mutagénesis Sitio-Dirigida , Trasplante de Neoplasias , Proteínas de Complejo Poro Nuclear/química , Proteínas de Fusión Oncogénica/química , Proteínas de Fusión Oncogénica/genética , Mutación Puntual , Regulación hacia Arriba , Proteína Exportina 1
17.
Dis Model Mech ; 13(4)2020 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-32102783

RESUMEN

Fibrolamellar carcinoma (FLC) is a rare liver cancer that affects adolescents and young adults. Genomic analysis of FLC has revealed a 400 kb deletion in chromosome 19 that leads to the chimeric transcript DNAJB1-PRKACA (DnaJ-PKAc), comprised of the first exon of heat shock protein 40 (DNAJB1) and exons 2-10 of the catalytic subunit of protein kinase A (PRKACA). Here, we report a new zebrafish model of FLC induced by ectopic expression of zebrafish Dnaja-Pkaca (zfDnaJa-Pkaca) in hepatocytes that is amenable to live imaging of early innate immune inflammation. Expression of zfDnaJa-Pkaca in hepatocytes induces hepatomegaly and increased hepatocyte size. In addition, FLC larvae exhibit early innate immune inflammation characterized by early infiltration of neutrophils and macrophages into the liver microenvironment. Increased Caspase-a (the zebrafish homolog for human caspase-1) activity was also found in the liver of FLC larvae, and pharmacological inhibition of Tnfα and caspase-a decreased liver size and inflammation. Overall, these findings show that innate immune inflammation is an early feature in a zebrafish model of FLC and that pharmacological inhibition of TNFα or caspase-1 activity might be targets to treat inflammation and progression in FLC patients.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Inmunidad Innata , Inflamación/patología , Hígado/patología , Proteínas de Fusión Oncogénica/metabolismo , Pez Cebra/metabolismo , Envejecimiento/patología , Secuencia de Aminoácidos , Animales , Carcinoma Hepatocelular/inmunología , Caspasas/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Hepatocitos/metabolismo , Hepatocitos/patología , Hígado/inmunología , Macrófagos/patología , Proteínas de Fusión Oncogénica/química , Factor de Necrosis Tumoral alfa/metabolismo
18.
J Biol Chem ; 295(13): 4303-4315, 2020 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-32098872

RESUMEN

The E-protein transcription factors guide immune cell differentiation, with E12 and E47 (hereafter called E2A) being essential for B-cell specification and maturation. E2A and the oncogenic chimera E2A-PBX1 contain three transactivation domains (ADs), with AD1 and AD2 having redundant, independent, and cooperative functions in a cell-dependent manner. AD1 and AD2 both mediate their functions by binding to the KIX domain of the histone acetyltransferase paralogues CREB-binding protein (CBP) and E1A-binding protein P300 (p300). This interaction is necessary for B-cell maturation and oncogenesis by E2A-PBX1 and occurs through conserved ΦXXΦΦ motifs (with Φ denoting a hydrophobic amino acid) in AD1 and AD2. However, disruption of this interaction via mutation of the KIX domain in CBP/p300 does not completely abrogate binding of E2A and E2A-PBX1. Here, we determined that E2A-AD1 and E2A-AD2 also interact with the TAZ2 domain of CBP/p300. Characterization of the TAZ2:E2A-AD1(1-37) complex indicated that E2A-AD1 adopts an α-helical structure and uses its ΦXXΦΦ motif to bind TAZ2. Whereas this region overlapped with the KIX recognition region, key KIX-interacting E2A-AD1 residues were exposed, suggesting that E2A-AD1 could simultaneously bind both the KIX and TAZ2 domains. However, we did not detect a ternary complex involving E2A-AD1, KIX, and TAZ2 and found that E2A containing both intact AD1 and AD2 is required to bind to CBP/p300. Our findings highlight the structural plasticity and promiscuity of E2A-AD1 and suggest that E2A binds both the TAZ2 and KIX domains of CBP/p300 through AD1 and AD2.


Asunto(s)
Proteína de Unión a CREB/química , Proteína p300 Asociada a E1A/genética , Dominios Proteicos/genética , Factor de Transcripción 3/química , Linfocitos B/química , Linfocitos B/metabolismo , Proteína de Unión a CREB/genética , Proteína de Unión a CREB/ultraestructura , Proteína p300 Asociada a E1A/química , Proteína p300 Asociada a E1A/ultraestructura , Proteínas de Homeodominio/química , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/ultraestructura , Humanos , Mutación/genética , Proteínas de Fusión Oncogénica/química , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/ultraestructura , Unión Proteica/genética , Conformación Proteica , Factor de Transcripción 3/genética , Factor de Transcripción 3/ultraestructura
19.
PLoS One ; 14(12): e0225977, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31826021

RESUMEN

The oncogenic fusion protein RUNX1-ETO is a product of the t(8;21) translocation and consists of the hematopoietic transcriptional master regulator RUNX1 and the repressor ETO. RUNX1-ETO is found in 10-15% of acute myeloid leukemia and interferes with the expression of genes that are essential for myeloid differentiation. The neutrophil serine protease Cathepsin G is one of the genes suppressed by RUNX1-ETO, but little is known about its impact on the regulation of other lysosomal proteases. By lentiviral transduction of the t(8;21) positive cell line Kasumi-1 with an RUNX1-ETO specific shRNA, we analyzed long-term effects of stable RUNX1-ETO silencing on cellular phenotypes and target gene expression. Stable anti RUNX1-ETO RNAi reduces both proliferation and apoptosis in Kasumi-1 cells. In addition, long-term knockdown of RUNX1-ETO leads to an upregulation of proteolytic activity in Kasumi-1 cells, which may be released in vitro upon cell lysis leading to massive degradation of cellular proteins. We therefore propose that protein expression data of RUNX1-ETO-silenced Kasumi-1 cells must be analyzed with caution, as cell lysis conditions can heavily influence the results of studies on protein expression. Next, a mass spectrometry-based approach was used to identify protease cleavage patterns in RUNX1-ETO-depleted Kasumi-1 cells and Neutrophil Elastase has been identified as a RUNX1-ETO candidate target. Finally, proteolytic activity of Neutrophil Elastase and Cathepsin G was functionally confirmed by si/shRNA-mediated knockdown in Kasumi-1 cells.


Asunto(s)
Catepsina G/metabolismo , Expresión Génica , Elastasa de Leucocito/metabolismo , Proteínas de Fusión Oncogénica/genética , Catepsina G/química , Línea Celular Tumoral , Cromatografía Liquida , Silenciador del Gen , Humanos , Elastasa de Leucocito/química , Proteínas de Fusión Oncogénica/química , Proteínas de Fusión Oncogénica/metabolismo , Proteolisis , ARN Largo no Codificante , Espectrometría de Masas en Tándem
20.
Metallomics ; 11(12): 2089-2096, 2019 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-31670356

RESUMEN

Arsenic trioxide (As2O3) is one of the most effective drugs for the treatment of acute promyelocytic leukemia (APL), and induces the degradation of chimeric oncoprotein PML/RARα (P/R) and APL cell differentiation. Recent evidence has suggested that P/R fusion protein degradation by arsenic occurs through two steps, namely, rapid solubility change/shift of the P/R fusion protein following arsenic treatment (i.e., transfer of P/R protein from the soluble fraction to the insoluble pellet fraction), and subsequent degradation of these insoluble proteins. However, there is little information regarding the reversibility of arsenic induced P/R fusion protein solubility change as well as protein degradation in the insoluble fraction after removing arsenic. In this study, we used APL cell line NB4 or P/R and PML over-expressed 293T cells as well as HeLa cells to reveal the solubility change of P/R and PML by arsenic exposure, and further determined the fate of these insoluble proteins after the removal of arsenic. Here, for the first time, we found that arsenic induced P/R or PML protein solubility change is an irreversible process. Once arsenic induces a P/R or PML protein solubility change, these insoluble proteins could be degraded by the proteasomal pathway even without continuous arsenic treatment. However, PML and P/R proteins can be newly synthesized after the removal of arsenic, suggesting that great caution should be taken in the clinical therapy of APL patients before ending arsenic treatment.


Asunto(s)
Trióxido de Arsénico/farmacología , Diferenciación Celular/efectos de los fármacos , Leucemia Promielocítica Aguda/tratamiento farmacológico , Proteínas de Fusión Oncogénica/metabolismo , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Células HEK293 , Células HeLa , Humanos , Leucemia Promielocítica Aguda/metabolismo , Leucemia Promielocítica Aguda/patología , Proteínas de Fusión Oncogénica/química , Proteínas de Fusión Oncogénica/genética , Solubilidad/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...