Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 86
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 300(5): 107144, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38458397

RESUMEN

Echinoderm microtubule-associated protein-like 4 (EML4)-anaplastic lymphoma kinase (ALK) oncogenic fusion proteins are found in approximately 5% of non-small cell lung cancers. Different EML4-ALK fusion variants exist with variant 3 (V3) being associated with a significantly higher risk than other common variants, such as variant 1 (V1). Patients with V3 respond less well to targeted ALK inhibitors, have accelerated rates of metastasis, and have poorer overall survival. A pathway has been described downstream of EML4-ALK V3 that is independent of ALK catalytic activity but dependent on the NEK9 and NEK7 kinases. It has been proposed that assembly of an EML4-ALK V3-NEK9-NEK7 complex on microtubules leads to cells developing a mesenchymal-like morphology and exhibiting enhanced migration. However, downstream targets of this complex remain unknown. Here, we show that the microtubule-based kinesin, Eg5, is recruited to interphase microtubules in cells expressing EML4-ALK V3, whereas chemical inhibition of Eg5 reverses the mesenchymal morphology of cells. Furthermore, we show that depletion of NEK7 interferes with Eg5 recruitment to microtubules in cells expressing EML4-ALK V3 and cell length is reduced, but this is reversed by coexpression of a phosphomimetic mutant of Eg5, in a site, S1033, phosphorylated by NEK7. Intriguingly, we also found that expression of Eg5-S1033D led to cells expressing EML4-ALK V1 adopting a more mesenchymal-like morphology. Together, we propose that Eg5 acts as a substrate of NEK7 in cells expressing EML4-ALK V3 and Eg5 phosphorylation promotes the mesenchymal morphology typical of these cells.


Asunto(s)
Cinesinas , Quinasas Relacionadas con NIMA , Proteínas de Fusión Oncogénica , Quinasas Relacionadas con NIMA/metabolismo , Quinasas Relacionadas con NIMA/genética , Humanos , Fosforilación , Proteínas de Fusión Oncogénica/metabolismo , Proteínas de Fusión Oncogénica/genética , Cinesinas/metabolismo , Cinesinas/genética , Microtúbulos/metabolismo , Microtúbulos/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/genética , Mesodermo/metabolismo , Mesodermo/patología , Línea Celular Tumoral , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética
2.
J Biol Chem ; 300(6): 107409, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38796063

RESUMEN

About 18% of all human cancers carry a mutation in the KRAS gene making it among the most sought-after anticancer targets. However, mutant KRas protein has proved remarkably undruggable. The recent approval of the first generation of RAS inhibitors therefore marks a seminal milestone in the history of cancer research. It also raises the predictable challenges of limited drug efficacies and acquired resistance. Hence, new approaches that improve our understanding of the tumorigenic mechanisms of oncogenic RAS within more physiological settings continue to be essential. Here, we have used the near-diploid hTERT RPE-1 cells to generate isogenic cell lines in which one of the endogenous KRAS alleles carries an oncogenic KRAS mutation at glycine 12. Cells with a KRASG12V/+, KRASG12C/+, or KRASG12D/+ genotype, together with WT KRASG12G(WT)/+ cells, reveal that oncogenic KRAS.G12X mutations increase cell proliferation rate and cell motility and reduced focal adhesions in KRASG12V/+ cells. Epidermal growth factor -induced phosphorylation of ERK and AKT was comparable between KRASG12V/+, KRASG12C/+, KRASG12D/+, and KRASG12G(WT)/+ cells. Interestingly, KRASG12X/+ cells showed varying responses to distinct inhibitors with the KRASG12V/+ and KRASG12D/+ cells more sensitive to hydroxyurea and MEK inhibitors, U0126 and trametinib, but more resistant to PI3K inhibitor, PIK-90, than the KRASG12G(WT)/+ cells. A combination of low doses of hydroxyurea and U0126 showed an additive inhibition on growth rate that was greater in KRASG12V/+ than WT cells. Collectively, these cell lines will be a valuable resource for studying oncogenic RAS signaling and developing effective anti-KRAS reagents with minimum cytotoxicity on WT cells.


Asunto(s)
Movimiento Celular , Proliferación Celular , Proteínas Proto-Oncogénicas p21(ras) , Humanos , Movimiento Celular/efectos de los fármacos , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Proliferación Celular/efectos de los fármacos , Telomerasa/genética , Telomerasa/metabolismo , Proteínas ras/metabolismo , Proteínas ras/genética , Pirimidinonas/farmacología , Piridonas/farmacología , Mutación Missense , Línea Celular , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas/genética , Nitrilos/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Butadienos/farmacología , Sustitución de Aminoácidos , Mutación
3.
Nucleic Acids Res ; 50(22): 13045-13062, 2022 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-36537190

RESUMEN

Sam68, also known as KHDRBS1, is a member of the STAR family of proteins that directly link signal transduction with post-transcriptional gene regulation. Sam68 controls the alternative splicing of many oncogenic proteins and its role is modulated by post-translational modifications, including serine/threonine phosphorylation, that differ at various stages of the cell cycle. However, the molecular basis and mechanisms of these modulations remain largely unknown. Here, we combined mass spectrometry, nuclear magnetic resonance spectroscopy and cell biology techniques to provide a comprehensive post-translational modification mapping of Sam68 at different stages of the cell cycle in HEK293 and HCT116 cells. We established that Sam68 is specifically phosphorylated at T33 and T317 by Cdk1, and demonstrated that these phosphorylation events reduce the binding of Sam68 to RNA, control its cellular localization and reduce its alternative splicing activity, leading to a reduction in the induction of apoptosis and an increase in the proliferation of HCT116 cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Empalme Alternativo , Humanos , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Empalme Alternativo/genética , Proteína Quinasa CDC2/genética , Proteína Quinasa CDC2/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/genética , Células HEK293 , Fosforilación , ARN/metabolismo , Proteínas de Unión al ARN/metabolismo , Células HCT116
4.
EMBO Rep ; 22(12): e53693, 2021 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-34661367

RESUMEN

Variants of the oncogenic EML4-ALK fusion protein contain a similar region of ALK encompassing the kinase domain, but different portions of EML4. Here, we show that EML4-ALK V1 and V3 proteins form cytoplasmic foci that contain components of the MAPK, PLCγ and PI3K signalling pathways. The ALK inhibitors ceritinib and lorlatinib dissolve these foci and EML4-ALK V3 but not V1 protein re-localises to microtubules, an effect recapitulated in a catalytically inactive EML4-ALK mutant. Mutations that promote a constitutively active ALK stabilise the cytoplasmic foci even in the presence of these inhibitors. In contrast, the inhibitor alectinib increases foci formation of both wild-type and catalytically inactive EML4-ALK V3 proteins, but not a Lys-Glu salt bridge mutant. We propose that EML4-ALK foci formation occurs as a result of transient association of stable EML4-ALK trimers mediated through an active conformation of the ALK kinase domain. Our results demonstrate the formation of EML4-ALK cytoplasmic foci that orchestrate oncogenic signalling and reveal that their assembly depends upon the conformational state of the catalytic domain and can be differentially modulated by structurally divergent ALK inhibitors.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Quinasa de Linfoma Anaplásico/genética , Humanos , Neoplasias Pulmonares/genética , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Conformación Proteica , Inhibidores de Proteínas Quinasas/farmacología
5.
Proc Natl Acad Sci U S A ; 117(29): 17195-17203, 2020 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-32606248

RESUMEN

The vast majority of intracellular protein targets are refractory toward small-molecule therapeutic engagement, and additional therapeutic modalities are needed to overcome this deficiency. Here, the identification and characterization of a natural product, WDB002, reveals a therapeutic modality that dramatically expands the currently accepted limits of druggability. WDB002, in complex with the FK506-binding protein (FKBP12), potently and selectively binds the human centrosomal protein 250 (CEP250), resulting in disruption of CEP250 function in cells. The recognition mode is unprecedented in that the targeted domain of CEP250 is a coiled coil and is topologically featureless, embodying both a structural motif and surface topology previously considered on the extreme limits of "undruggability" for an intracellular target. Structural studies reveal extensive protein-WDB002 and protein-protein contacts, with the latter being distinct from those seen in FKBP12 ternary complexes formed by FK506 and rapamycin. Outward-facing structural changes in a bound small molecule can thus reprogram FKBP12 to engage diverse, otherwise "undruggable" targets. The flat-targeting modality demonstrated here has the potential to expand the druggable target range of small-molecule therapeutics. As CEP250 was recently found to be an interaction partner with the Nsp13 protein of the SARS-CoV-2 virus that causes COVID-19 disease, it is possible that WDB002 or an analog may exert useful antiviral activity through its ability to form high-affinity ternary complexes containing CEP250 and FKBP12.


Asunto(s)
Actinobacteria/genética , Antivirales/farmacología , Genoma Bacteriano , Macrólidos/farmacología , Dominios y Motivos de Interacción de Proteínas/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Proteína 1A de Unión a Tacrolimus/química , Proteína 1A de Unión a Tacrolimus/metabolismo , Actinobacteria/metabolismo , Secuencia de Aminoácidos , Antivirales/química , Antivirales/metabolismo , Autoantígenos/genética , Autoantígenos/metabolismo , Calcineurina/genética , Calcineurina/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Evolución Molecular , Células HEK293 , Humanos , Macrólidos/química , Macrólidos/metabolismo , Modelos Moleculares , Conformación Proteica , Homología de Secuencia , Sirolimus/química , Sirolimus/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/metabolismo , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
6.
J Cell Sci ; 133(9)2020 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-32184261

RESUMEN

EML4-ALK is an oncogenic fusion present in ∼5% of non-small cell lung cancers. However, alternative breakpoints in the EML4 gene lead to distinct variants of EML4-ALK with different patient outcomes. Here, we show that, in cell models, EML4-ALK variant 3 (V3), which is linked to accelerated metastatic spread, causes microtubule stabilization, formation of extended cytoplasmic protrusions and increased cell migration. EML4-ALK V3 also recruits the NEK9 and NEK7 kinases to microtubules via the N-terminal EML4 microtubule-binding region. Overexpression of wild-type EML4, as well as constitutive activation of NEK9, also perturbs cell morphology and accelerates migration in a microtubule-dependent manner that requires the downstream kinase NEK7 but does not require ALK activity. Strikingly, elevated NEK9 expression is associated with reduced progression-free survival in EML4-ALK patients. Hence, we propose that EML4-ALK V3 promotes microtubule stabilization through NEK9 and NEK7, leading to increased cell migration. This represents a novel actionable pathway that could drive metastatic disease progression in EML4-ALK lung cancer.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Carcinoma de Pulmón de Células no Pequeñas/genética , Humanos , Neoplasias Pulmonares/genética , Microtúbulos , Quinasas Relacionadas con NIMA/genética , Proteínas de Fusión Oncogénica/genética , Proteínas Tirosina Quinasas Receptoras
7.
Nat Chem Biol ; 11(7): 496-503, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26030730

RESUMEN

Serine phosphorylation is a key post-translational modification that regulates diverse biological processes. Powerful analytical methods have identified thousands of phosphorylation sites, but many of their functions remain to be deciphered. A key to understanding the function of protein phosphorylation is access to phosphorylated proteins, but this is often challenging or impossible. Here we evolve an orthogonal aminoacyl-tRNA synthetase/tRNACUA pair that directs the efficient incorporation of phosphoserine (pSer (1)) into recombinant proteins in Escherichia coli. Moreover, combining the orthogonal pair with a metabolically engineered E. coli enables the site-specific incorporation of a nonhydrolyzable analog of pSer. Our approach enables quantitative decoding of the amber stop codon as pSer, and we purify, with yields of several milligrams per liter of culture, proteins bearing biologically relevant phosphorylations that were previously challenging or impossible to access--including phosphorylated ubiquitin and the kinase Nek7, which is synthetically activated by a genetically encoded phosphorylation in its activation loop.


Asunto(s)
Aminoacil-ARNt Sintetasas/genética , Proteínas de Escherichia coli/genética , Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica , Fosfoserina/metabolismo , Procesamiento Proteico-Postraduccional , Aminoacil-ARNt Sintetasas/química , Aminoacil-ARNt Sintetasas/metabolismo , Secuencia de Bases , Codón de Terminación/química , Codón de Terminación/metabolismo , Escherichia coli/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Código Genético , Modelos Moleculares , Datos de Secuencia Molecular , Quinasas Relacionadas con NIMA , Conformación de Ácido Nucleico , Fosforilación , Fosfoserina/química , Ingeniería de Proteínas , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Ubiquitina/química , Ubiquitina/genética , Ubiquitina/metabolismo
8.
Mol Cell ; 36(4): 560-70, 2009 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-19941817

RESUMEN

Mitosis is controlled by multiple protein kinases, many of which are abnormally expressed in human cancers. Nek2, Nek6, Nek7, and Nek9 are NIMA-related kinases essential for proper mitotic progression. We determined the atomic structure of Nek7 and discovered an autoinhibited conformation that suggests a regulatory mechanism not previously described in kinases. Additionally, Nek2 adopts the same conformation when bound to a drug-like molecule. In both structures, a tyrosine side chain points into the active site, interacts with the activation loop, and blocks the alphaC helix. Tyrosine mutants of Nek7 and the related kinase Nek6 are constitutively active. The activity of Nek6 and Nek7, but not the tyrosine mutant, is increased by interaction with the Nek9 noncatalytic C-terminal domain, suggesting a mechanism in which the tyrosine is released from its autoinhibitory position. The autoinhibitory conformation is common to three Neks and provides a potential target for selective kinase inhibitors.


Asunto(s)
Ciclo Celular , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/metabolismo , Tirosina/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Dominio Catalítico , Ciclo Celular/efectos de los fármacos , Línea Celular , Cristalografía por Rayos X , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Mutación/genética , Quinasas Relacionadas con NIMA , Unión Proteica/efectos de los fármacos , Relación Estructura-Actividad
9.
Cell Mol Life Sci ; 73(6): 1209-24, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26755435

RESUMEN

A fusion between the EML4 (echinoderm microtubule-associated protein-like) and ALK (anaplastic lymphoma kinase) genes was identified in non-small cell lung cancer (NSCLC) in 2007 and there has been rapid progress in applying this knowledge to the benefit of patients. However, we have a poor understanding of EML4 and ALK biology and there are many challenges to devising the optimal strategy for treating EML4-ALK NSCLC patients. In this review, we describe the biology of EML4 and ALK, explain the main features of EML4-ALK fusion proteins and outline the therapies that target EML4-ALK. In particular, we highlight the recent advances in our understanding of the structures of EML proteins, describe the molecular mechanisms of resistance to ALK inhibitors and assess current thinking about combinations of ALK drugs with inhibitors that target other kinases or Hsp90.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Proteínas de Ciclo Celular/genética , Neoplasias Pulmonares/genética , Pulmón/patología , Proteínas Asociadas a Microtúbulos/genética , Proteínas de Fusión Oncogénica/genética , Proteínas Tirosina Quinasas Receptoras/genética , Serina Endopeptidasas/genética , Quinasa de Linfoma Anaplásico , Animales , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Proteínas de Ciclo Celular/análisis , Proteínas de Ciclo Celular/metabolismo , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Humanos , Pulmón/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Proteínas Asociadas a Microtúbulos/análisis , Proteínas Asociadas a Microtúbulos/metabolismo , Modelos Moleculares , Terapia Molecular Dirigida , Proteínas de Fusión Oncogénica/análisis , Proteínas de Fusión Oncogénica/metabolismo , Conformación Proteica , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Tirosina Quinasas Receptoras/análisis , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/metabolismo , Serina Endopeptidasas/análisis , Serina Endopeptidasas/metabolismo
10.
Proc Natl Acad Sci U S A ; 111(14): 5195-200, 2014 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-24706829

RESUMEN

Proteins of the echinoderm microtubule-associated protein (EMAP)-like (EML) family contribute to formation of the mitotic spindle and interphase microtubule network. They contain a unique hydrophobic EML protein (HELP) motif and a variable number of WD40 repeats. Recurrent gene rearrangements in nonsmall cell lung cancer fuse EML4 to anaplastic lymphoma kinase (ALK), causing expression of several fusion oncoprotein variants. We have determined a 2.6-Å crystal structure of the representative ∼70-kDa core of EML1, revealing an intimately associated pair of ß-propellers, which we term a TAPE (tandem atypical propeller in EMLs) domain. One propeller is highly atypical, having a discontinuous subdomain unrelated to a WD40 motif in place of one of its blades. This unexpected feature shows how a propeller structure can be assembled from subdomains with distinct folds. The HELP motif is not an independent domain but forms part of the hydrophobic core that joins the two ß-propellers. The TAPE domain binds α/ß-tubulin via its conserved, concave surface, including part of the atypical blade. Mapping the characteristic breakpoints of each EML4-ALK variant onto our structure indicates that the EML4 TAPE domain is truncated in many variants in a manner likely to make the fusion protein structurally unstable. We found that the heat shock protein 90 (Hsp90) inhibitor ganetespib induced degradation of these variants whereas others lacking a partial TAPE domain were resistant in both overexpression models and patient-derived cell lines. The Hsp90-sensitive EML4-ALK variants are exceptions to the rule that oncogenic fusion proteins involve breakpoints in disordered regions of both partners.


Asunto(s)
Proteínas de Ciclo Celular/química , Proteínas HSP90 de Choque Térmico/metabolismo , Proteínas Asociadas a Microtúbulos/química , Proteínas Tirosina Quinasas Receptoras/metabolismo , Serina Endopeptidasas/química , Secuencia de Aminoácidos , Quinasa de Linfoma Anaplásico , Proteínas de Ciclo Celular/metabolismo , Cristalografía por Rayos X , Humanos , Proteínas Asociadas a Microtúbulos/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica , Homología de Secuencia de Aminoácido , Serina Endopeptidasas/metabolismo
11.
Proc Natl Acad Sci U S A ; 111(27): 9840-5, 2014 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-24958871

RESUMEN

Histone deacetylases 1 and 2 (HDAC1/2) form the core catalytic components of corepressor complexes that modulate gene expression. In most cell types, deletion of both Hdac1 and Hdac2 is required to generate a discernible phenotype, suggesting their activity is largely redundant. We have therefore generated an ES cell line in which Hdac1 and Hdac2 can be inactivated simultaneously. Loss of HDAC1/2 resulted in a 60% reduction in total HDAC activity and a loss of cell viability. Cell death is dependent upon cell cycle progression, because differentiated, nonproliferating cells retain their viability. Furthermore, we observe increased mitotic defects, chromatin bridges, and micronuclei, suggesting HDAC1/2 are necessary for accurate chromosome segregation. Consistent with a critical role in the regulation of gene expression, microarray analysis of Hdac1/2-deleted cells reveals 1,708 differentially expressed genes. Significantly for the maintenance of stem cell self-renewal, we detected a reduction in the expression of the pluripotent transcription factors, Oct4, Nanog, Esrrb, and Rex1. HDAC1/2 activity is regulated through binding of an inositol tetraphosphate molecule (IP4) sandwiched between the HDAC and its cognate corepressor. This raises the important question of whether IP4 regulates the activity of the complex in cells. By rescuing the viability of double-knockout cells, we demonstrate for the first time (to our knowledge) that mutations that abolish IP4 binding reduce the activity of HDAC1/2 in vivo. Our data indicate that HDAC1/2 have essential and pleiotropic roles in cellular proliferation and regulate stem cell self-renewal by maintaining expression of key pluripotent transcription factors.


Asunto(s)
División Celular/fisiología , Células Madre Embrionarias/enzimología , Histona Desacetilasa 1/fisiología , Histona Desacetilasa 2/fisiología , Células Madre Pluripotentes/enzimología , Acetilación , Animales , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Regulación de la Expresión Génica , Histonas/metabolismo , Ratones , Ratones Noqueados , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Factores de Transcripción/metabolismo
12.
J Cell Sci ; 127(Pt 11): 2493-506, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24695856

RESUMEN

During mitotic entry, centrosomes separate to establish the bipolar spindle. Delays in centrosome separation can perturb chromosome segregation and promote genetic instability. However, interphase centrosomes are physically tethered by a proteinaceous linker composed of C-Nap1 (also known as CEP250) and the filamentous protein rootletin. Linker disassembly occurs at the onset of mitosis in a process known as centrosome disjunction and is triggered by the Nek2-dependent phosphorylation of C-Nap1. However, the mechanistic consequences of C-Nap1 phosphorylation are unknown. Here, we demonstrate that Nek2 phosphorylates multiple residues within the C-terminal domain of C-Nap1 and, collectively, these phosphorylation events lead to loss of oligomerization and centrosome association. Mutations in non-phosphorylatable residues that make the domain more acidic are sufficient to release C-Nap1 from the centrosome, suggesting that it is an increase in overall negative charge that is required for this process. Importantly, phosphorylation of C-Nap1 also perturbs interaction with the core centriolar protein, Cep135, and interaction of endogenous C-Nap1 and Cep135 proteins is specifically lost in mitosis. We therefore propose that multisite phosphorylation of C-Nap1 by Nek2 perturbs both oligomerization and Cep135 interaction, and this precipitates centrosome disjunction at the onset of mitosis.


Asunto(s)
Autoantígenos/metabolismo , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/metabolismo , Centriolos/metabolismo , Centrosoma/fisiología , Huso Acromático/metabolismo , Autoantígenos/genética , Proteínas de Ciclo Celular/genética , Segregación Cromosómica/genética , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Inestabilidad Genómica , Células HeLa , Humanos , Mitosis , Mutación/genética , Quinasas Relacionadas con NIMA , Fosforilación , Unión Proteica/genética , Ingeniería de Proteínas , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Interferente Pequeño/genética
13.
Biochem Soc Trans ; 44(5): 1281-1288, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27911710

RESUMEN

The EMLs are a conserved family of microtubule-associated proteins (MAPs). The founding member was discovered in sea urchins as a 77-kDa polypeptide that co-purified with microtubules. This protein, termed EMAP for echinoderm MAP, was the major non-tubulin component present in purified microtubule preparations made from unfertilized sea urchin eggs [J. Cell Sci. (1993) 104: , 445-450; J. Cell Sci. (1987) 87: (Pt 1), 71-84]. Orthologues of EMAP were subsequently identified in other echinoderms, such as starfish and sand dollar, and then in more distant eukaryotes, including flies, worms and vertebrates, where the name of ELP or EML (both for EMAP-like protein) has been adopted [BMC Dev. Biol. (2008) 8: , 110; Dev. Genes Evol. (2000) 210: , 2-10]. The common property of these proteins is their ability to decorate microtubules. However, whether they are associated with particular microtubule populations or exercise specific functions in different microtubule-dependent processes remains unknown. Furthermore, although there is limited evidence that they regulate microtubule dynamics, the biochemical mechanisms of their molecular activity have yet to be explored. Nevertheless, interest in these proteins has grown substantially because of the identification of EML mutations in neuronal disorders and oncogenic fusions in human cancers. Here, we summarize our current knowledge of the expression, localization and structure of what is proving to be an interesting and important class of MAPs. We also speculate about their function in microtubule regulation and highlight how the studies of EMLs in human diseases may open up novel avenues for patient therapy.


Asunto(s)
Ciclo Celular/genética , Enfermedad/genética , Proteínas Asociadas a Microtúbulos/genética , Microtúbulos/genética , Animales , Diferenciación Celular/genética , Proliferación Celular/genética , Humanos , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Modelos Genéticos , Erizos de Mar/genética , Erizos de Mar/metabolismo
14.
Malar J ; 15(1): 535, 2016 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-27821169

RESUMEN

BACKGROUND: Examining essential biochemical pathways in Plasmodium falciparum presents serious challenges, as standard molecular techniques such as siRNA cannot be employed in this organism, and generating gene knock-outs of essential proteins requires specialized conditional approaches. In the study of protein kinases, pharmacological inhibition presents a feasible alternative option. However, as in mammalian systems, inhibitors often lack the desired selectivity. Described here is a chemical genetic approach to selectively inhibit Pfnek-2 in P. falciparum, a member of the NIMA-related kinase family that is essential for completion of the sexual development of the parasite. RESULTS: Introduction of a valine to cysteine mutation at position 24 in the glycine rich loop of Pfnek-2 does not affect kinase activity but confers sensitivity to the protein kinase inhibitor 4-(6-ethynyl-9H-purin-2-ylamino) benzene sulfonamide (NCL-00016066). Using a combination of in vitro kinase assays and mass spectrometry, (including phosphoproteomics) the study shows that this compound acts as an irreversible inhibitor to the mutant Pfnek2 likely through a covalent link with the introduced cysteine residue. In particular, this was shown by analysis of total protein mass using mass spectrometry which showed a shift in molecular weight of the mutant kinase in the presence of the inhibitor to be precisely equivalent to the molecular weight of NCL-00016066. A similar molecular weight shift was not observed in the wild type kinase. Importantly, this inhibitor has little activity towards the wild type Pfnek-2 and, therefore, has all the properties of an effective chemical genetic tool that could be employed to determine the cellular targets for Pfnek-2. CONCLUSIONS: Allelic replacement of wild-type Pfnek-2 with the mutated kinase will allow for targeted inhibition of Pfnek-2 with NCL-00016066 and hence pave the way for comparative studies aimed at understanding the biological role and transmission-blocking potential of Pfnek-2.


Asunto(s)
Inhibidores Enzimáticos/metabolismo , Proteínas Mutantes/metabolismo , Quinasas Relacionadas con NIMA/metabolismo , Plasmodium falciparum/enzimología , Purinas/metabolismo , Sulfonamidas/metabolismo , Espectrometría de Masas , Proteínas Mutantes/genética , Quinasas Relacionadas con NIMA/genética
15.
Mol Cell ; 32(4): 576-83, 2008 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-19026787

RESUMEN

The Fizzy/Cdc20 family of proteins are essential activators of the anaphase-promoting complex/cyclosome (APC/C), a multisubunit E3 ubiquitin ligase. However, apart from the well-established role of the C-terminal WD40 domain in substrate recognition, the precise roles of the activators remain elusive. Here we show that Nek2A, which directly binds the APC/C, can be ubiquitylated and destroyed in Fizzy/Cdc20-depleted Xenopus egg extracts when only the N-terminal domain of Fizzy/Cdc20 (N-Cdc20) is added. This activity is dependent upon the C box and is conserved in the alternative activator, Fizzy-related/Cdh1. In contrast, canonical substrates such as cyclin B and securin require both the N-terminal and WD40 domains, unless N-Cdc20 is fused to substrates when the WD40 domain becomes dispensable. Furthermore, in Cdc20-depleted cells, N-Cdc20 can facilitate Nek2A destruction in a C box-dependent manner. Our results reveal a role for the N-terminal domain of the Fizzy/Cdc20 family of activators in triggering substrate ubiquitylation by the APC/C.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas/metabolismo , Complejos de Ubiquitina-Proteína Ligasa/metabolismo , Proteínas de Xenopus/metabolismo , Ciclosoma-Complejo Promotor de la Anafase , Animales , Proteínas Cdc20 , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Modelos Biológicos , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Terciaria de Proteína , Proteínas/genética , Especificidad por Sustrato , Ubiquitinación , Xenopus/genética , Xenopus/metabolismo , Proteínas de Xenopus/química , Proteínas de Xenopus/genética
16.
Biochem J ; 467(3): 529-36, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25740311

RESUMEN

Proteins of the echinoderm microtubule (MT)-associated protein (EMAP)-like (EML) family contribute to formation of the mitotic spindle and interphase MT network. EML1-4 consist of Trp-Asp 40 (WD40) repeats and an N-terminal region containing a putative coiled-coil. Recurrent gene rearrangements in non-small cell lung cancer (NSCLC) fuse EML4 to anaplastic lymphoma kinase (ALK) causing expression of several oncogenic fusion variants. The fusions have constitutive ALK activity due to self-association through the EML4 coiled-coil. We have determined crystal structures of the coiled-coils from EML2 and EML4, which describe the structural basis of both EML self-association and oncogenic EML4-ALK activation. The structures reveal a trimeric oligomerization state directed by a conserved pattern of hydrophobic residues and salt bridges. We show that the trimerization domain (TD) of EML1 is necessary and sufficient for self-association. The TD is also essential for MT binding; however, this property requires an adjacent basic region. These observations prompted us to investigate MT association of EML4-ALK and EML1-ABL1 (Abelson 1) fusions in which variable portions of the EML component are present. Uniquely, EML4-ALK variant 3, which includes the TD and basic region of EML4 but none of the WD40 repeats, was localized to MTs, both when expressed recombinantly and when expressed in a patient-derived NSCLC cell line (H2228). This raises the question of whether the mislocalization of ALK activity to MTs might influence downstream signalling and malignant properties of cells. Furthermore, the structure of EML4 TD may enable the development of protein-protein interaction inhibitors targeting the trimerization interface, providing a possible avenue towards therapeutic intervention in EML4-ALK NSCLC.


Asunto(s)
Proteínas Asociadas a Microtúbulos/química , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Proteínas de Fusión Oncogénica/química , Proteínas de Fusión Oncogénica/metabolismo , Secuencia de Aminoácidos , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Línea Celular Tumoral , Secuencia Conservada , Cristalografía por Rayos X , Células HEK293 , Células HeLa , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Proteínas de Fusión Oncogénica/genética , Dominios y Motivos de Interacción de Proteínas , Estructura Cuaternaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido
17.
J Cell Sci ; 126(Pt 1): 163-75, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23015594

RESUMEN

Proteomic studies in unicellular eukaryotes identified a set of centriolar proteins that included proteome of centriole 1 (Poc1). Functional studies in these organisms implicated Poc1 in centriole duplication and length control, as well as ciliogenesis. Using isoform-specific antibodies and RNAi depletion, we have examined the function of the two related human proteins, Poc1A and Poc1B. We find that Poc1A and Poc1B each localize to centrioles and spindle poles, but do so independently and with different dynamics. However, although loss of one or other Poc1 protein does not obviously disrupt mitosis, depletion of both proteins leads to defects in spindle organization with the generation of unequal or monopolar spindles. Our data indicate that, once incorporated, a fraction of Poc1A and Poc1B remains stably associated with parental centrioles, but that depletion prevents incorporation into nascent centrioles. Nascent centrioles lacking both Poc1A and Poc1B exhibit loss of integrity and maturation, and fail to undergo duplication. Thus, when Poc1A and Poc1B are co-depleted, new centrosomes capable of maturation cannot assemble and unequal spindles result. Interestingly, Poc1B, but not Poc1A, is phosphorylated in mitosis, and depletion of Poc1B alone was sufficient to perturb cell proliferation. Hence, Poc1A and Poc1B play redundant, but essential, roles in generation of stable centrioles, but Poc1B may have additional independent functions during cell cycle progression.


Asunto(s)
Centriolos/metabolismo , Proteínas/metabolismo , Huso Acromático/metabolismo , Western Blotting , Ciclo Celular/genética , Ciclo Celular/fisiología , Proteínas de Ciclo Celular , Línea Celular , Línea Celular Tumoral , Proteínas del Citoesqueleto , Electroforesis en Gel de Poliacrilamida , Citometría de Flujo , Células HeLa , Humanos , Inmunoprecipitación , Mitosis/genética , Mitosis/fisiología , Proteínas/genética
18.
Hum Mutat ; 35(10): 1153-62, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25044745

RESUMEN

We describe a consanguineous Iraqi family with Leber congenital amaurosis (LCA), Joubert syndrome (JBTS), and polycystic kidney disease (PKD). Targeted next-generation sequencing for excluding mutations in known LCA and JBTS genes, homozygosity mapping, and whole-exome sequencing identified a homozygous missense variant, c.317G>C (p.Arg106Pro), in POC1B, a gene essential for ciliogenesis, basal body, and centrosome integrity. In silico modeling suggested a requirement of p.Arg106 for the formation of the third WD40 repeat and a protein interaction interface. In human and mouse retina, POC1B localized to the basal body and centriole adjacent to the connecting cilium of photoreceptors and in synapses of the outer plexiform layer. Knockdown of Poc1b in zebrafish caused cystic kidneys and retinal degeneration with shortened and reduced photoreceptor connecting cilia, compatible with the human syndromic ciliopathy. A recent study describes homozygosity for p.Arg106ProPOC1B in a family with nonsyndromic cone-rod dystrophy. The phenotype associated with homozygous p.Arg106ProPOC1B may thus be highly variable, analogous to homozygous p.Leu710Ser in WDR19 causing either isolated retinitis pigmentosa or Jeune syndrome. Our study indicates that POC1B is required for retinal integrity, and we propose POC1B mutations as a probable cause for JBTS with severe PKD.


Asunto(s)
Proteínas de Ciclo Celular/genética , Enfermedades Cerebelosas/genética , Anomalías del Ojo/genética , Enfermedades Renales Quísticas/genética , Mutación , Retina/anomalías , Anomalías Múltiples , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Proteínas de Ciclo Celular/metabolismo , Enfermedades Cerebelosas/metabolismo , Enfermedades Cerebelosas/patología , Cerebelo/anomalías , Niño , Cilios/metabolismo , Cilios/ultraestructura , Anomalías del Ojo/metabolismo , Anomalías del Ojo/patología , Técnicas de Silenciamiento del Gen , Humanos , Irak , Riñón/patología , Enfermedades Renales Quísticas/metabolismo , Enfermedades Renales Quísticas/patología , Amaurosis Congénita de Leber/genética , Amaurosis Congénita de Leber/metabolismo , Masculino , Ratones , Datos de Secuencia Molecular , Linaje , Retina/metabolismo , Retina/patología , Pez Cebra
19.
Hum Mol Genet ; 21(5): 1155-71, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22106379

RESUMEN

Mutations in the never-in-mitosis A-related kinase, Nek8, are associated with cystic kidney disease in both humans and mice, with Nek8 being the NPHP9 gene in the human juvenile cystic kidney disease, nephronophthisis. Human Nek8/NPHP9 localizes to centrosomes and the proximal region of cilia in dividing and ciliated cells, respectively. However, the regulation of Nek8 kinase activity, as well as its role in ciliogenesis, remains to be defined. Here, by establishing Nek8 kinase assays, we first demonstrate that the localization of Nek8 to centrosomes and cilia is dependent on both kinase activity and the C-terminal non-catalytic RCC1 domain. The kinase domain alone is active, but does not localize correctly, while the RCC1 domain localizes correctly and can be phosphorylated by Nek8. We propose that centrosome recruitment is mediated by the RCC1 domain, but requires a conformational change in the full-length protein that is promoted by autophosphorylation. Interestingly, three human NPHP9-associated mutants retain full kinase activity. However, only two of these, L330F and A497P, localize correctly, suggesting that the third mutant, H425Y, disrupts a centrosome targeting sequence in the RCC1 domain. Importantly, we find that induction of ciliogenesis upon cell cycle exit is accompanied by both activation and proteasomal degradation of Nek8, and that activation is dependent upon phosphorylation within the catalytic domain. Taken together, these findings reveal important insights into the mechanisms through which Nek8 activity and localization are regulated during ciliogenesis.


Asunto(s)
Cilios/metabolismo , Enfermedades Renales Quísticas/genética , Proteínas Quinasas/metabolismo , Animales , Dominio Catalítico , Ciclo Celular , Línea Celular , Núcleo Celular/metabolismo , Centrosoma/metabolismo , Cilios/ultraestructura , Activación Enzimática , Humanos , Enfermedades Renales Quísticas/metabolismo , Ratones , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Mutación , Quinasas Relacionadas con NIMA , Fosforilación , Complejo de la Endopetidasa Proteasomal/metabolismo , Conformación Proteica , Proteínas Quinasas/química , Proteínas Quinasas/genética , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/metabolismo , Fase de Descanso del Ciclo Celular
20.
J Cell Sci ; 125(Pt 19): 4423-33, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23132929

RESUMEN

Genetic screens for cell division cycle mutants in the filamentous fungus Aspergillus nidulans led to the discovery of never-in-mitosis A (NIMA), a serine/threonine kinase that is required for mitotic entry. Since that discovery, NIMA-related kinases, or NEKs, have been identified in most eukaryotes, including humans where eleven genetically distinct proteins named NEK1 to NEK11 are expressed. Although there is no evidence that human NEKs are essential for mitotic entry, it is clear that several NEK family members have important roles in cell cycle control. In particular, NEK2, NEK6, NEK7 and NEK9 contribute to the establishment of the microtubule-based mitotic spindle, whereas NEK1, NEK10 and NEK11 have been implicated in the DNA damage response. Roles for NEKs in other aspects of mitotic progression, such as chromatin condensation, nuclear envelope breakdown, spindle assembly checkpoint signalling and cytokinesis have also been proposed. Interestingly, NEK1 and NEK8 also function within cilia, the microtubule-based structures that are nucleated from basal bodies. This has led to the current hypothesis that NEKs have evolved to coordinate microtubule-dependent processes in both dividing and non-dividing cells. Here, we review the functions of the human NEKs, with particular emphasis on those family members that are involved in cell cycle control, and consider their potential as therapeutic targets in cancer.


Asunto(s)
Puntos de Control del Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Familia de Multigenes , Proteínas Serina-Treonina Quinasas/metabolismo , Regulación Alostérica/efectos de los fármacos , Animales , Antineoplásicos/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/química , Humanos , Mitosis/efectos de los fármacos , Quinasa 1 Relacionada con NIMA , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA