Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 143
Filtrar
1.
J Med Chem ; 64(18): 13902-13917, 2021 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-34515477

RESUMEN

The nucleosome remodeling factor (NURF) alters chromatin accessibility through interactions with its largest subunit,the bromodomain PHD finger transcription factor BPTF. BPTF is overexpressed in several cancers and is an emerging anticancer target. Targeting the BPTF bromodomain presents a potential strategy for its inhibition and the evaluation of its functional significance; however, inhibitor development for BPTF has lagged behind those of other bromodomains. Here we describe the development of pyridazinone-based BPTF inhibitors. The lead compound, BZ1, possesses a high potency (Kd = 6.3 nM) and >350-fold selectivity over BET bromodomains. We identify an acidic triad in the binding pocket to guide future designs. We show that our inhibitors sensitize 4T1 breast cancer cells to doxorubicin but not BPTF knockdown cells, suggesting a specificity to BPTF. Given the high potency and good physicochemical properties of these inhibitors, we anticipate that they will be useful starting points for chemical tool development to explore the biological roles of BPTF.


Asunto(s)
Antineoplásicos/farmacología , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Piridazinas/farmacología , Factores de Transcripción/antagonistas & inhibidores , Secuencia de Aminoácidos , Animales , Antígenos Nucleares/química , Antineoplásicos/química , Antineoplásicos/toxicidad , Línea Celular Tumoral , Diseño de Fármacos , Ratones , Estructura Molecular , Proteínas del Tejido Nervioso/química , Dominios Proteicos , Piridazinas/química , Piridazinas/toxicidad , Relación Estructura-Actividad , Factores de Transcripción/química
2.
J Med Chem ; 64(16): 12075-12088, 2021 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-34375106

RESUMEN

The dysfunctional bromodomain PHD finger transcription factor (BPTF) exerts a pivotal influence in the occurrence and development of many human diseases, particularly cancers. Herein, through the structural decomposition of the reported BPTF inhibitor TP-238, the effective structural fragments were synthetically modified to obtain our lead compound DC-BPi-03. DC-BPi-03 was identified as a novel BPTF-BRD inhibitor with a moderate potency (IC50 = 698.3 ± 21.0 nM). A structure-guided structure-activity relationship exploration gave rise to two BPTF inhibitors with much higher affinities, DC-BPi-07 and DC-BPi-11. Notably, DC-BPi-07 and DC-BPi-11 show selectivities 100-fold higher than those of other BRD targets. The cocrystal structures of BPTF in complex with DC-BPi-07 and DC-BPi-11 demonstrate the rationale of chemical efforts from the atomic level. Further study showed that DC-BPi-11 significantly inhibited leukemia cell proliferation.


Asunto(s)
Antineoplásicos/farmacología , Indoles/farmacología , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Pirimidinas/farmacología , Factores de Transcripción/antagonistas & inhibidores , Antígenos Nucleares/química , Antígenos Nucleares/metabolismo , Antineoplásicos/síntesis química , Antineoplásicos/metabolismo , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Descubrimiento de Drogas , Ensayos de Selección de Medicamentos Antitumorales , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Humanos , Indoles/síntesis química , Indoles/metabolismo , Estructura Molecular , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Unión Proteica , Dominios Proteicos , Pirimidinas/síntesis química , Pirimidinas/metabolismo , Relación Estructura-Actividad , Factores de Transcripción/química , Factores de Transcripción/metabolismo
3.
Curr Opin Chem Biol ; 63: 57-67, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33706239

RESUMEN

Nucleosome remodeling provides access to genomic DNA for recruitment of the transcriptional machinery to mediate gene expression. The aberrant function of nucleosome remodeling complexes has been correlated to human cancer, making them emerging therapeutic targets. The bromodomain PHD finger transcription factor, BPTF, is the largest member of the human nucleosome remodeling factor NURF. Over the last five years, BPTF has become increasingly identified as a protumorigenic factor, prompting investigations into the molecular mechanisms associated with BPTF function. Despite a druggable bromodomain, small molecule discovery is at an early stage. Here we highlight recent investigations into the biology being discovered for BPTF, chemical biology approaches used to study its function, and small molecule inhibitors being designed as future chemical probes and therapeutics.


Asunto(s)
Antígenos Nucleares/química , Inhibidores Enzimáticos/química , Técnicas de Inactivación de Genes/métodos , Proteínas del Tejido Nervioso/química , Nucleosomas/química , Bibliotecas de Moléculas Pequeñas/química , Factores de Transcripción/química , Antígenos Nucleares/genética , Sitios de Unión , Epigénesis Genética , Histonas/química , Humanos , Lisina/química , Proteínas del Tejido Nervioso/genética , Nucleosomas/metabolismo , Unión Proteica , Dominios Proteicos , Relación Estructura-Actividad , Factores de Transcripción/genética
4.
Biochem Biophys Res Commun ; 545: 125-131, 2021 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-33548625

RESUMEN

Bromodomain and PHD finger containing transcription factor (BPTF) is a multidomain protein that regulates the transcription of chromatin and is related to many cancers. Herein, we report the screening-based discovery of Cpd1, a compound with micromolar affinity to the BPTF bromodomain. Through structure-guided optimization, we synthesized a variety of new inhibitors. Among these compounds, Cpd8 and Cpd10 were highly potent and selective inhibitors, with KD values of 428 nM and 655 nM in ITC assays, respectively. The high activity was explained by the cocrystal structure of Cpd8 in complex with the BPTF bromodomain protein. Cpd8 and Cpd10 were able to stabilize the BPTF bromodomain protein in cells in a cellular thermal shift assay (CETSA). Cpd8 downregulated c-MYC expression in A549 cells. All experiments prove that these two compounds are potential BPTF inhibitors.


Asunto(s)
Proteínas del Tejido Nervioso/antagonistas & inhibidores , Factores de Transcripción/antagonistas & inhibidores , Células A549 , Antígenos Nucleares/química , Antígenos Nucleares/genética , Calorimetría , Cristalografía por Rayos X , Diseño de Fármacos , Descubrimiento de Drogas , Evaluación Preclínica de Medicamentos , Fluorometría , Regulación de la Expresión Génica/efectos de los fármacos , Genes myc , Células HEK293 , Humanos , Modelos Moleculares , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Dominios Proteicos , Estabilidad Proteica/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/efectos de los fármacos , Proteínas Recombinantes/genética , Relación Estructura-Actividad , Factores de Transcripción/química , Factores de Transcripción/genética
5.
Molecules ; 25(17)2020 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-32872491

RESUMEN

As fragment-based drug discovery has become mainstream, there has been an increase in various screening methodologies. Protein-observed 19F (PrOF) NMR and 1H CPMG NMR are two fragment screening assays that have complementary advantages. Here, we sought to combine these two NMR-based assays into a new screening workflow. This combination of protein- and ligand-observed experiments allows for a time- and resource-efficient multiplexed screen of mixtures of fragments and proteins. PrOF NMR is first used to screen mixtures against two proteins. Hit mixtures for each protein are identified then deconvoluted using 1H CPMG NMR. We demonstrate the benefit of this fragment screening method by conducting the first reported fragment screens against the bromodomains of BPTF and Plasmodium falciparum (Pf) GCN5 using 467 3D-enriched fragments. The hit rates were 6%, 5% and 4% for fragments binding BPTF, PfGCN5, and fragments binding both proteins, respectively. Select hits were characterized, revealing a broad range of affinities from low µM to mM dissociation constants. Follow-up experiments supported a low-affinity second binding site on PfGCN5. This approach can be used to bias fragment screens towards more selective hits at the onset of inhibitor development in a resource- and time-efficient manner.


Asunto(s)
Antígenos Nucleares/química , Descubrimiento de Drogas/métodos , Histona Acetiltransferasas/química , Proteínas del Tejido Nervioso/química , Proteínas Protozoarias/química , Factores de Transcripción/química , Sitios de Unión , Humanos , Ligandos , Imagen por Resonancia Magnética , Modelos Moleculares , Plasmodium falciparum , Unión Proteica , Dominios Proteicos , Bibliotecas de Moléculas Pequeñas
6.
Org Biomol Chem ; 18(27): 5174-5182, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32588860

RESUMEN

Bromodomain-containing proteins regulate transcription through protein-protein interactions with chromatin and serve as scaffolding proteins for recruiting essential members of the transcriptional machinery. One such protein is the bromodomain and PHD-containing transcription factor (BPTF), the largest member of the nucleosome remodeling complex, NURF. Despite an emerging role for BPTF in regulating a diverse set of cancers, small molecule development for inhibiting the BPTF bromodomain has been lacking. Here we cross-validate three complementary biophysical assays to further the discovery of BPTF bromodomain inhibitors for chemical probe development: two direct binding assays (protein-observed 19F (PrOF) NMR and surface plasmon resonance (SPR)) and a competitive inhibition assay (AlphaScreen). We first compare the assays using three small molecules and acetylated histone peptides with reported affinity for the BPTF bromodomain. Using SPR with both unlabeled and fluorinated BPTF, we further determine that there is a minimal effect of 19F incorporation on ligand binding for future PrOF NMR experiments. To guide medicinal chemistry efforts towards chemical probe development, we subsequently evaluate two new BPTF inhibitor scaffolds with our suite of biophysical assays and rank-order compound affinities which could not otherwise be determined by PrOF NMR. Finally, we cocrystallize a subset of small molecule inhibitors and present the first published small molecule-protein structures with the BPTF bromodomain. We envision the biophysical assays described here and the structural insights from the crystallography will guide researchers towards developing selective and potent BPTF bromodomain inhibitors.


Asunto(s)
Proteínas del Tejido Nervioso/antagonistas & inhibidores , Factores de Transcripción/antagonistas & inhibidores , Antígenos Nucleares/química , Fenómenos Biofísicos , Espectroscopía de Resonancia Magnética , Proteínas del Tejido Nervioso/química , Dominios Proteicos , Resonancia por Plasmón de Superficie , Factores de Transcripción/química
7.
J Biol Chem ; 295(13): 4114-4123, 2020 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-32047112

RESUMEN

Ether-a-go-go (EAG) potassium selective channels are major regulators of neuronal excitability and cancer progression. EAG channels contain a Per-Arnt-Sim (PAS) domain in their intracellular N-terminal region. The PAS domain is structurally similar to the PAS domains in non-ion channel proteins, where these domains frequently function as ligand-binding domains. Despite the structural similarity, it is not known whether the PAS domain can regulate EAG channel function via ligand binding. Here, using surface plasmon resonance, tryptophan fluorescence, and analysis of EAG currents recorded in Xenopus laevis oocytes, we show that a small molecule chlorpromazine (CH), widely used as an antipsychotic medication, binds to the isolated PAS domain of EAG channels and inhibits currents from these channels. Mutant EAG channels that lack the PAS domain show significantly lower inhibition by CH, suggesting that CH affects currents from EAG channels directly through the binding to the PAS domain. Our study lends support to the hypothesis that there are previously unaccounted steps in EAG channel gating that could be activated by ligand binding to the PAS domain. This has broad implications for understanding gating mechanisms of EAG and related ERG and ELK K+ channels and places the PAS domain as a new target for drug discovery in EAG and related channels. Up-regulation of EAG channel activity is linked to cancer and neurological disorders. Our study raises the possibility of repurposing the antipsychotic drug chlorpromazine for treatment of neurological disorders and cancer.


Asunto(s)
Clorpromazina/farmacología , Canal de Potasio ERG1/genética , Canales de Potasio Éter-A-Go-Go/genética , Neuronas/efectos de los fármacos , Secuencia de Aminoácidos/genética , Animales , Antígenos de Neoplasias/química , Antígenos de Neoplasias/genética , Antígenos Nucleares/química , Antígenos Nucleares/genética , Sitios de Unión/efectos de los fármacos , Excitabilidad Cortical/efectos de los fármacos , Excitabilidad Cortical/genética , Canal de Potasio ERG1/química , Canales de Potasio Éter-A-Go-Go/química , Canales de Potasio Éter-A-Go-Go/metabolismo , Humanos , Ligandos , Neuronas/metabolismo , Oocitos/crecimiento & desarrollo , Oocitos/metabolismo , Dominios Proteicos/efectos de los fármacos , Resonancia por Plasmón de Superficie , Xenopus laevis/genética
8.
Immunotherapy ; 11(14): 1205-1219, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31478431

RESUMEN

Colorectal cancer is the third commonest malignancy in Asia including Malaysia. The immunogenic cancer-testis antigens, which are expressed in a variety of cancers but with limited expression in normal tissues except the testis, represent an attractive approach to improve treatment options for colorectal cancer. We aimed to validate four PASD1 peptides as the immunotherapeutic targets in colorectal cancer. First, PASD1 mRNA and protein expression were determined via real-time polymerase chain reaction (RT-PCR) and immunohistochemistry. The PASD1 peptides specific to HLA-A*24:02 were investigated using IFN-y-ELISpot assay, followed by the cytolytic and granzyme-B-ELISpot assays to analyze the cytolytic effects of CD8+ T cells. Gene and protein expressions of PASD1 were detected in 20% and 17.3% of colorectal cancer samples, respectively. PASD1(4) peptide was shown to be immunogenic in colorectal cancer samples. CD8+ T cells raised against PASD1(4) peptide were able to lyze HLA-A*24:02+ PASD1+ cells. Our results reveal that PASD1(4) peptide represents a potential target for colorectal cancer.


Asunto(s)
Antígenos de Neoplasias , Antígenos Nucleares , Neoplasias Colorrectales , Antígeno HLA-A24/inmunología , Inmunoterapia , Proteínas de Neoplasias , Péptidos , Antígenos de Neoplasias/química , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/farmacología , Antígenos Nucleares/química , Antígenos Nucleares/inmunología , Antígenos Nucleares/farmacología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/terapia , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/inmunología , Células HCT116 , Humanos , Inmunidad Celular/efectos de los fármacos , Masculino , Proteínas de Neoplasias/química , Proteínas de Neoplasias/inmunología , Proteínas de Neoplasias/farmacología , Péptidos/química , Péptidos/inmunología , Péptidos/farmacología
9.
Nat Commun ; 10(1): 2208, 2019 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-31101817

RESUMEN

Cortical force generators connect epithelial polarity sites with astral microtubules, allowing dynein movement to orient the mitotic spindle as astral microtubules depolymerize. Complexes of the LGN and NuMA proteins, fundamental components of force generators, are recruited to the cortex by Gαi-subunits of heterotrimeric G-proteins. They associate with dynein/dynactin and activate the motor activity pulling on astral microtubules. The architecture of cortical force generators is unknown. Here we report the crystal structure of NuMA:LGN hetero-hexamers, and unveil their role in promoting the assembly of active cortical dynein/dynactin motors that are required in orchestrating oriented divisions in polarized cells. Our work elucidates the basis for the structural organization of essential spindle orientation motors.


Asunto(s)
Antígenos Nucleares/metabolismo , Polaridad Celular , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Asociadas a Matriz Nuclear/metabolismo , Huso Acromático/metabolismo , Antígenos Nucleares/química , Antígenos Nucleares/genética , Antígenos Nucleares/aislamiento & purificación , Células CACO-2 , Proteínas de Ciclo Celular , Cristalografía por Rayos X , Complejo Dinactina/metabolismo , Dineínas/metabolismo , Técnicas de Silenciamiento del Gen , Células HEK293 , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/aislamiento & purificación , Microtúbulos/metabolismo , Proteínas Asociadas a Matriz Nuclear/química , Proteínas Asociadas a Matriz Nuclear/genética , Proteínas Asociadas a Matriz Nuclear/aislamiento & purificación , Unión Proteica/fisiología , Multimerización de Proteína/fisiología , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo
10.
Biochim Biophys Acta Gen Subj ; 1863(2): 456-465, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30465816

RESUMEN

BACKGROUND: Human Sp140 protein is a leukocyte-specific member of the speckled protein (Sp) family (Sp100, Sp110, Sp140, Sp140L), a class of multi-domain nuclear proteins involved in intrinsic immunity and transcriptional regulation. Sp140 regulates macrophage transcriptional program and is implicated in several haematologic malignancies. Little is known about Sp140 structural domains and its post-translational modifications. METHODS: We used mass spectrometry and biochemical experiments to investigate endogenous Sp140 SUMOylation in Burkitt's Lymphoma cells and Sp140 SUMOylation sites in HEK293T cells, FLAG-Sp140 transfected and His6-SUMO-1T95K infected. NMR spectroscopy and in vitro SUMOylation reactions were applied to investigate the role of Sp140 PHD finger in the SUMOylation of the adjacent BRD. RESULTS: Endogenous Sp140 is a SUMO-1 target, whereby FLAG-Sp140 harbors at least 13 SUMOylation sites distributed along the protein sequence, including the BRD. NMR experiments prove direct binding of the SUMO E2 ligase Ubc9 and SUMO-1 to PHD-BRDSp140. In vitro SUMOylation reactions show that the PHDSp140 behaves as SUMO E3 ligase, assisting intramolecular SUMOylation of the adjacent BRD. CONCLUSIONS: Sp140 is multi-SUMOylated and its PHD finger works as versatile protein-protein interaction platform promoting intramolecular SUMOylation of the adjacent BRD. Thus, combinatorial association of Sp140 chromatin binding domains generates a multifaceted interaction scaffold, whose function goes beyond the canonical histone recognition. GENERAL SIGNIFICANCE: The addition of Sp140 to the increasing lists of multi-SUMOylated proteins opens new perspectives for molecular studies on Sp140 transcriptional activity, where SUMOylation could represent a regulatory route and a docking surface for the recruitment and assembly of leukocyte-specific transcription regulators.


Asunto(s)
Antígenos Nucleares/química , Antígenos Nucleares/metabolismo , Dedos de Zinc PHD , Dominios Proteicos , Sumoilación , Factores de Transcripción/química , Factores de Transcripción/metabolismo , Células HEK293 , Humanos , Unión Proteica
11.
J Proteome Res ; 18(3): 1064-1077, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30585729

RESUMEN

The Ku heterodimer, composed of Ku70 and Ku80, is best characterized for its role in repairing double-stranded DNA breaks but is also known to participate in other regulatory processes. Despite our understanding of Ku protein interplay during DNA repair, the extent of Ku's protein interactions in other processes has never been fully determined. Using proximity-dependent biotin identification (BioID) and affinity purification coupled to mass spectrometry (AP-MS) with wild-type Ku70, we identified candidate proteins that interact with the Ku heterodimer in HEK293 cells, in the absence of exogenously induced DNA damage. BioID analysis identified approximately 250 nuclear proteins, appearing in at least two replicates, including known Ku-interacting factors such as MRE11A, WRN, and NCOA6. Meanwhile, AP-MS analysis identified approximately 50 candidate proteins. Of the novel protein interactors identified, many were involved in functions already suspected to involve Ku such as transcriptional regulation, DNA replication, and DNA repair, while several others suggest that Ku may be involved in additional functions such as RNA metabolism, chromatin-remodeling, and microtubule dynamics. Using a combination of BioID and AP-MS, this is the first report that comprehensively characterizes the Ku protein interaction landscape, revealing new cellular processes and protein complexes involving the Ku complex.


Asunto(s)
Antígenos Nucleares/genética , Reparación del ADN/genética , Autoantígeno Ku/genética , Proteínas Nucleares/genética , Antígenos Nucleares/química , Biotina/química , Daño del ADN/genética , Replicación del ADN/genética , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Células HEK293 , Humanos , Autoantígeno Ku/química , Proteína Homóloga de MRE11/genética , Proteínas Nucleares/química , Coactivadores de Receptor Nuclear/genética , Multimerización de Proteína/genética , Helicasa del Síndrome de Werner/genética
12.
Elife ; 72018 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-29848445

RESUMEN

To position the mitotic spindle within the cell, dynamic plus ends of astral microtubules are pulled by membrane-associated cortical force-generating machinery. However, in contrast to the chromosome-bound kinetochore structure, how the diffusion-prone cortical machinery is organized to generate large spindle-pulling forces remains poorly understood. Here, we develop a light-induced reconstitution system in human cells. We find that induced cortical targeting of NuMA, but not dynein, is sufficient for spindle pulling. This spindle-pulling activity requires dynein-dynactin recruitment by NuMA's N-terminal long arm, dynein-based astral microtubule gliding, and NuMA's direct microtubule-binding activities. Importantly, we demonstrate that cortical NuMA assembles specialized focal structures that cluster multiple force-generating modules to generate cooperative spindle-pulling forces. This clustering activity of NuMA is required for spindle positioning, but not for spindle-pole focusing. We propose that cortical Dynein-Dynactin-NuMA (DDN) clusters act as the core force-generating machinery that organizes a multi-arm ensemble reminiscent of the kinetochore.


Asunto(s)
Antígenos Nucleares/metabolismo , Complejo Dinactina/metabolismo , Dineínas/metabolismo , Proteínas Asociadas a Matriz Nuclear/metabolismo , Huso Acromático/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Antígenos Nucleares/química , Proteínas de Ciclo Celular , Línea Celular , Humanos , Ácidos Indolacéticos/farmacología , Luz , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Mutación/genética , Proteínas Asociadas a Matriz Nuclear/química , Optogenética , Paclitaxel/farmacología , Fenotipo , Dominios Proteicos
13.
J Clin Invest ; 128(7): 2951-2965, 2018 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-29649003

RESUMEN

A synthetic lethality-based strategy has been developed to identify therapeutic targets in cancer harboring tumor-suppressor gene mutations, as exemplified by the effectiveness of poly ADP-ribose polymerase (PARP) inhibitors in BRCA1/2-mutated tumors. However, many synthetic lethal interactors are less reliable due to the fact that such genes usually do not perform fundamental or indispensable functions in the cell. Here, we developed an approach to identifying the "essential lethality" arising from these mutated/deleted essential genes, which are largely tolerated in cancer cells due to genetic redundancy. We uncovered the cohesion subunit SA1 as a putative synthetic-essential target in cancers carrying inactivating mutations of its paralog, SA2. In SA2-deficient Ewing sarcoma and bladder cancer, further depletion of SA1 profoundly and specifically suppressed cancer cell proliferation, survival, and tumorigenic potential. Mechanistically, inhibition of SA1 in the SA2-mutated cells led to premature chromatid separation, dramatic extension of mitotic duration, and consequently, lethal failure of cell division. More importantly, depletion of SA1 rendered those SA2-mutated cells more susceptible to DNA damage, especially double-strand breaks (DSBs), due to reduced functionality of DNA repair. Furthermore, inhibition of SA1 sensitized the SA2-deficient cancer cells to PARP inhibitors in vitro and in vivo, providing a potential therapeutic strategy for patients with SA2-deficient tumors.


Asunto(s)
Antígenos Nucleares/genética , Proteínas de Ciclo Celular/genética , Proteínas Cromosómicas no Histona/genética , Mutación , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Proteínas Nucleares/genética , Animales , Antígenos Nucleares/química , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/química , Línea Celular Tumoral , Proteínas Cromosómicas no Histona/antagonistas & inhibidores , Proteínas Cromosómicas no Histona/química , Roturas del ADN de Doble Cadena , Femenino , Técnicas de Silenciamiento del Gen , Genes Esenciales , Humanos , Ratones , Ratones Desnudos , Neoplasias/patología , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/química , Ftalazinas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Subunidades de Proteína/antagonistas & inhibidores , Subunidades de Proteína/química , Subunidades de Proteína/genética , Sarcoma de Ewing/tratamiento farmacológico , Sarcoma de Ewing/genética , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Cohesinas
14.
Biochemistry ; 57(3): 300-304, 2018 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-29111671

RESUMEN

Methyllysine analogues (MLAs), furnished by aminoethylation of engineered cysteine residues, are widely used surrogates of histone methyllysine and are considered to be effective proxies for studying these epigenetic marks in vitro. Here we report the first structure of a trimethyllysine MLA histone in complex with a protein binding partner, quantify the thermodynamic distinctions between MLAs and their native methyllysine counterparts, and demonstrate that these differences can compromise qualitative interpretations of binding at the nucleosome level. Quantitative measurements with two methyllysine binding protein modules reveal substantial affinity losses for the MLA peptides versus the corresponding native methyllysine species in both cases, although the thermodynamic underpinnings are distinct. MLA and methyllysine adopt distinct conformational geometries when in complex with the BPTF PHD finger, a well-established H3K4me3 binding partner. In this case, an ∼13-fold Kd difference at the peptide level translates to nucleosomal affinities for MLA analogues that fall outside of the detectable range in a pull-down format, whereas the methyllysine species installed by native chemical ligation demonstrates robust binding. Thus, despite their facile production and commercial availability, there is a significant caveat of potentially altered binding affinity when MLAs are used in place of native methyllysine residues.


Asunto(s)
Antígenos Nucleares/química , Histonas/química , Lisina/análogos & derivados , Proteínas del Tejido Nervioso/química , Dedos de Zinc PHD , Factores de Transcripción/química , Secuencia de Aminoácidos , Humanos , Lisina/química , Unión Proteica , Procesamiento Proteico-Postraduccional , Termodinámica
15.
J Cell Biol ; 216(11): 3453-3462, 2017 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-28939615

RESUMEN

Ran-guanosine triphosphatase orchestrates mitotic spindle assembly by modulation of the interaction between Importin-α/-ß and spindle assembly factors (SAFs). The inhibition of SAFs performed by importins needs to be done without much sequestration from abundant nuclear localization signal (NLS) -containing proteins. However, the molecular mechanisms that determine NLS-binding selectivity and that inhibit activity of Importin-ß-regulated SAFs (e.g., nuclear mitotic apparatus protein [NuMA]) remain undefined. Here, we present a crystal structure of the Importin-α-NuMA C terminus complex showing a novel binding pattern that accounts for selective NLS recognition. We demonstrate that, in the presence of Importin-α, Importin-ß inhibits the microtubule-binding function of NuMA. Further, we have identified a high-affinity microtubule-binding region that lies carboxyl-terminal to the NLS, which is sterically masked by Importin-ß on being bound by Importin-α. Our study provides mechanistic evidence of how Importin-α/-ß regulates the NuMA functioning required for assembly of higher-order microtubule structures, further illuminating how Ran-governed transport factors regulate diverse SAFs and accommodate various cell demands.


Asunto(s)
Antígenos Nucleares/metabolismo , Proteínas Asociadas a Matriz Nuclear/metabolismo , Huso Acromático/metabolismo , beta Carioferinas/metabolismo , Animales , Antígenos Nucleares/química , Antígenos Nucleares/genética , Proteínas de Ciclo Celular , Humanos , Microtúbulos/metabolismo , Modelos Moleculares , Complejos Multiproteicos , Proteínas Asociadas a Matriz Nuclear/química , Proteínas Asociadas a Matriz Nuclear/genética , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Huso Acromático/química , Huso Acromático/genética , Relación Estructura-Actividad , Xenopus , alfa Carioferinas/metabolismo , beta Carioferinas/química , beta Carioferinas/genética , Proteína de Unión al GTP ran/metabolismo
16.
Nat Commun ; 8: 13996, 2017 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-28045117

RESUMEN

Both cell-cell adhesion and oriented cell division play prominent roles in establishing tissue architecture, but it is unclear how they might be coordinated. Here, we demonstrate that the cell-cell adhesion protein E-cadherin functions as an instructive cue for cell division orientation. This is mediated by the evolutionarily conserved LGN/NuMA complex, which regulates cortical attachments of astral spindle microtubules. We show that LGN, which adopts a three-dimensional structure similar to cadherin-bound catenins, binds directly to the E-cadherin cytosolic tail and thereby localizes at cell-cell adhesions. On mitotic entry, NuMA is released from the nucleus and competes LGN from E-cadherin to locally form the LGN/NuMA complex. This mediates the stabilization of cortical associations of astral microtubules at cell-cell adhesions to orient the mitotic spindle. Our results show how E-cadherin instructs the assembly of the LGN/NuMA complex at cell-cell contacts, and define a mechanism that couples cell division orientation to intercellular adhesion.


Asunto(s)
Antígenos Nucleares/química , Cadherinas/química , Células Epiteliales/metabolismo , Péptidos y Proteínas de Señalización Intracelular/química , Microtúbulos/metabolismo , Proteínas Asociadas a Matriz Nuclear/química , Huso Acromático/metabolismo , Animales , Antígenos CD , Antígenos Nucleares/genética , Antígenos Nucleares/metabolismo , Sitios de Unión , Cadherinas/genética , Cadherinas/metabolismo , Adhesión Celular , Comunicación Celular , Proteínas de Ciclo Celular , División Celular , Línea Celular , Perros , Drosophila melanogaster/citología , Drosophila melanogaster/metabolismo , Células Epiteliales/ultraestructura , Expresión Génica , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Células de Riñón Canino Madin Darby , Microtúbulos/ultraestructura , Modelos Moleculares , Proteínas Asociadas a Matriz Nuclear/genética , Proteínas Asociadas a Matriz Nuclear/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estructura Secundaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Huso Acromático/ultraestructura
17.
J Phys Chem B ; 121(15): 3565-3573, 2017 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-27976577

RESUMEN

Phosphorylation is one of the most frequent post-translational modifications on proteins. It regulates many cellular processes by modulation of phosphorylation on protein structure and dynamics. However, the mechanism of phosphorylation-induced conformational changes of proteins is still poorly understood. Here, we report a computational study of three representative groups of tyrosine in ADP-ribosylhydrolase 1, serine in BTG2, and serine in Sp100C by using six molecular dynamics (MD) simulations and quantum chemical calculations. Added phosphorylation was found to disrupt hydrogen bond, and increase new weak interactions (hydrogen bond and hydrophobic interaction) during MD simulations, leading to conformational changes. Quantum chemical calculations further indicate that the phosphorylation on tyrosine, threonine, and serine could decrease the optical band gap energy (Egap), which can trigger electronic transitions to form or disrupt interactions easily. Our results provide an atomic and electronic description of how phosphorylation facilitates conformational and dynamic changes in proteins, which may be useful for studying protein function and protein design.


Asunto(s)
Simulación de Dinámica Molecular , Teoría Cuántica , Serina/química , Tirosina/química , ADP Ribosa Transferasas/química , ADP Ribosa Transferasas/metabolismo , Antígenos Nucleares/química , Autoantígenos/química , Proteínas Ligadas a GPI/química , Proteínas Ligadas a GPI/metabolismo , Humanos , Proteínas Inmediatas-Precoces/química , Fosforilación , Proteínas Supresoras de Tumor/química
18.
Bioconjug Chem ; 27(5): 1373-81, 2016 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-27070736

RESUMEN

Cell-penetrating peptides (CPPs) have been successfully applied to deliver various functional macromolecules into cells in recent times. Here, we describe a novel CPP designated as hPP3 (KPKRKRRKKKGHGWSR), which were derived from human nuclear body protein SP140-like protein. The location of hPP3-FITC in cells was investigated using the fluorescence microscopy, and the internalization of hPP3 was quantitatively measured using a fluorescence spectrophotometer. The results showed that hPP3-FITC could enter into culturing cells, following a concentration-, incubation time-, serum-, and temperature-dependent manner. Uptake of hPP3-FITC into cells was significantly enhanced by DMSO pretreatment, and inhibited by heparin and the endocytosis inhibitors (chlorpromazine and sodium azide), while the potent lysosomotropic agent, chloroquine, showed small positive effects on hPP3-FITC penetrating. Moreover, hPP3 could mediate functional GFP, KLA, or NBD penetration. The findings of this study showed that human origin peptide hPP3 has the potential to act as a macromolecular carrier penetrating cellular membranes and promising delivery peptide as drug delivery vectors.


Asunto(s)
Antígenos Nucleares/química , Péptidos de Penetración Celular/química , Portadores de Fármacos/química , Factores de Transcripción/química , Secuencia de Aminoácidos , Apoptosis/efectos de los fármacos , Péptidos de Penetración Celular/metabolismo , Dimetilsulfóxido/química , Liberación de Fármacos , Células HeLa , Humanos , L-Lactato Deshidrogenasa/química , Transporte de Proteínas
19.
J Biol Chem ; 291(24): 12786-12798, 2016 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-27129259

RESUMEN

The decoding of histone post-translational modifications by chromatin-binding modules ("readers") constitutes one major mechanism of epigenetic regulation. Nuclear antigen Sp100 (SPECKLED, 100 kDa), a constitutive component of the promyelocytic leukemia nuclear bodies, plays key roles in intrinsic immunity and transcriptional repression. Sp100C, a splicing isoform specifically up-regulated upon interferon stimulation, harbors a unique tandem plant homeodomain (PHD) finger and bromodomain at its C terminus. Combining structural, quantitative binding, and cellular co-localization studies, we characterized Sp100C PHD finger as an unmethylated histone H3 Lys(4) (H3K4me0) reader that tolerates histone H3 Thr(3) phosphorylation (H3T3ph), histone H3 Lys(9) trimethylation (H3K9me3), and histone H3 Ser(10) phosphorylation (H3S10ph), hallmarks associated with the mitotic chromosome. In contrast, whereas H3K4me0 reader activity is conserved in Sp140, an Sp100C paralog, the multivalent tolerance of H3T3ph, H3K9me3, and H3S10ph was lost for Sp140. The complex structure determined at 2.1 Šrevealed a highly coordinated lysine ϵ-amine recognition sphere formed by an extended N-terminal motif for H3K4me0 readout. Interestingly, reader pocket rigidification by disulfide bond formation enhanced H3K4me0 binding by Sp100C. An additional complex structure solved at 2.7 Šrevealed that H3T3ph is recognized by the arginine residue, Arg(713), that is unique to the PHD finger of Sp100C. Consistent with a restrictive cellular role of Sp100C, these results establish a direct chromatin targeting function of Sp100C that may regulate transcriptional gene silencing and promyelocytic leukemia nuclear body-mediated intrinsic immunity in response to interferon stimulation.


Asunto(s)
Antígenos Nucleares/metabolismo , Autoantígenos/metabolismo , Histonas/metabolismo , Lisina/metabolismo , Secuencia de Aminoácidos , Antígenos Nucleares/química , Antígenos Nucleares/genética , Autoantígenos/química , Autoantígenos/genética , Sitios de Unión/genética , Calorimetría/métodos , Cristalografía por Rayos X , Células HeLa , Histonas/química , Humanos , Enlace de Hidrógeno , Lisina/química , Metilación , Microscopía Confocal , Modelos Moleculares , Mutación , Fosforilación , Unión Proteica , Dominios Proteicos , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Homología de Secuencia de Aminoácido
20.
Exp Cell Res ; 342(2): 135-44, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26976509

RESUMEN

Ku protein is a heterodimer composed of two subunits, and is capable of both sequence-independent and sequence-specific DNA binding. The former mode of DNA binding plays a crucial role in DNA repair. The biological role of Ku protein during apoptosis remains unclear. Here, we show characterization of Ku protein during apoptosis. In order to study the DNA binding properties of Ku, we used two methods for the electrophoresis mobility shift assay (EMSA). One method, RI-EMSA, which is commonly used, employed radiolabeled DNA probes. The other method, WB-EMSA, employed unlabeled DNA followed by western blot and detection with anti-Ku antiserum. In this study, Ku-DNA probe binding activity was found to dramatically decrease upon etoposide treatment, when examined by the RI-EMSA method. In addition, pre-treatment with apoptotic cell extracts inhibited Ku-DNA probe binding activity in the non-treated cell extract. The inhibitory effect of the apoptotic cell extract was reduced by DNase I treatment. WB-EMSA showed that the Ku in the apoptotic cell extract bound to fragmented endogenous DNA. Interestingly, Ku in the apoptotic cell extract purified by the Resource Q column bound 15-bp DNA in both RI-EMSA and WB-EMSA, whereas Ku in unpurified apoptotic cell extracts did not bind additional DNA. These results suggest that Ku binds cleaved chromosomal DNA and/or nucleosomes in apoptotic cells. In conclusion, Ku is intact and retains DNA binding activity in early apoptotic cells.


Asunto(s)
Antígenos Nucleares/metabolismo , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , Etopósido/farmacología , Antígenos Nucleares/química , Antineoplásicos/química , ADN/química , Proteína Quinasa Activada por ADN/metabolismo , Proteínas de Unión al ADN/química , Ensayo de Cambio de Movilidad Electroforética , Etopósido/química , Células HL-60 , Humanos , Autoantígeno Ku , Unión Proteica , Transporte de Proteínas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...