Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
J Mol Biol ; 433(18): 167120, 2021 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-34197833

RESUMEN

Proteins that exhibit intrinsically disordered regions (IDRs) are prevalent in the human proteome and perform diverse biological functions, including signaling and regulation. Due to these important roles, misregulation of intrinsically disordered proteins (IDPs) is associated with myriad human diseases, including neurodegeneration and cancer. The inherent flexibility of IDPs limits the applicability of the traditional structure-based drug design paradigm; therefore, IDPs have long been considered "undruggable". Using NMR spectroscopy and other methods, we previously discovered small, drug-like molecules that bind specifically, albeit weakly, to dynamic clusters of aromatic residues within p27Kip1 (p27), an archetypal disordered protein involved in cell cycle regulation. Here, using synthetic chemistry, NMR spectroscopy and other biophysical methods, we discovered elaborated analogs of our previously reported molecules with 30-fold increased affinity for p27 (apparent Kd = 57 ± 19 µM). Strikingly, using analytical ultracentrifugation methods, we showed that the highest affinity compounds caused p27 to form soluble, disordered oligomers. Based on these observations, we propose that sequestration within soluble oligomers may represent a general strategy for therapeutically targeting disease-associated IDPs in the future.


Asunto(s)
Inhibidor p27 de las Quinasas Dependientes de la Ciclina/química , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Diseño de Fármacos , Descubrimiento de Drogas , Proteínas Intrínsecamente Desordenadas/metabolismo , Bibliotecas de Moléculas Pequeñas/metabolismo , Humanos , Proteínas Intrínsecamente Desordenadas/química , Unión Proteica , Bibliotecas de Moléculas Pequeñas/química
2.
J Mol Biol ; 433(5): 166795, 2021 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-33422522

RESUMEN

The SCFSKP2 ubiquitin ligase relieves G1 checkpoint control of CDK-cyclin complexes by promoting p27KIP1 degradation. We describe reconstitution of stable complexes containing SKP1-SKP2 and CDK1-cyclin B or CDK2-cyclin A/E, mediated by the CDK regulatory subunit CKS1. We further show that a direct interaction between a SKP2 N-terminal motif and cyclin A can stabilize SKP1-SKP2-CDK2-cyclin A complexes in the absence of CKS1. We identify the SKP2 binding site on cyclin A and demonstrate the site is not present in cyclin B or cyclin E. This site is distinct from but overlapping with features that mediate binding of p27KIP1 and other G1 cyclin regulators to cyclin A. We propose that the capacity of SKP2 to engage with CDK2-cyclin A by more than one structural mechanism provides a way to fine tune the degradation of p27KIP1 and distinguishes cyclin A from other G1 cyclins to ensure orderly cell cycle progression.


Asunto(s)
Ciclina A/química , Quinasa 2 Dependiente de la Ciclina/química , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/química , Puntos de Control de la Fase G1 del Ciclo Celular , Proteínas Quinasas Asociadas a Fase-S/química , Sitios de Unión , Quinasas CDC2-CDC28/química , Quinasas CDC2-CDC28/genética , Quinasas CDC2-CDC28/metabolismo , Ciclina A/genética , Ciclina A/metabolismo , Ciclina E/química , Ciclina E/genética , Ciclina E/metabolismo , Quinasa 2 Dependiente de la Ciclina/genética , Quinasa 2 Dependiente de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Humanos , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Proteolisis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Quinasas Asociadas a Fase-S/genética , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Transducción de Señal
3.
J Mol Biol ; 432(9): 2998-3017, 2020 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-32088186

RESUMEN

The protein p27, a prominent regulatory protein in eukaryotes and an intrinsically disordered protein (IDP), regulates cell division by causing cell cycle arrest when bound in ternary complex with cyclin-dependent kinase (Cdk2) and cyclins (e.g., Cdk2/Cyclin A). We present an integrative study of p27 and its binding to Cdk2/Cyclin A complex by performing single-molecule multiparameter fluorescence spectroscopy, stopped-flow experiments, and molecular dynamics simulations. Our results suggest that unbound p27 adopts a compact conformation and undergoes conformational dynamics across several orders of magnitude in time (nano-to milliseconds), reflecting a multi-step mechanism for binding Cdk2/Cyclin A. Mutagenesis studies reveal that the region D1 in p27 plays a significant role in mediating the association kinetics, undergoing conformational rearrangement upon initial binding. Additionally, FRET experiments indicate an expansion of p27 throughout binding. The detected local and long-range structural dynamics suggest that p27 exhibits a limited binding surface in the unbound form, and stochastic conformational changes in D1 facilitate initial binding to Cdk2/Cyclin A complex. Furthermore, the post-kinase inhibitory domain (post-KID) region of p27 exchanges between distinct conformational ensembles: an extended regime exhibiting worm-like chain behavior, and a compact ensemble, which may protect p27 against nonspecific interactions. In summary, the binding interaction involves three steps: (i) D1 initiates binding, (ii) p27 wraps around Cdk2/Cyclin A and D2 binds, and (iii) the fully-formed fuzzy ternary complex is formed concomitantly with an extension of the post-KID region. An understanding of how the IDP nature of p27 underpins its functional interactions with Cdk2/Cyclin A provides insight into the complex binding mechanisms of IDPs and their regulatory mechanisms.


Asunto(s)
Ciclina A/metabolismo , Quinasa 2 Dependiente de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/química , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Imagen Individual de Molécula/métodos , Sitios de Unión , Ciclina A/química , Quinasa 2 Dependiente de la Ciclina/química , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Transferencia Resonante de Energía de Fluorescencia , Humanos , Modelos Moleculares , Mutación , Unión Proteica , Conformación Proteica , Espectrometría de Fluorescencia , Factores Complejos Ternarios/química
4.
Science ; 366(6471)2019 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-31831640

RESUMEN

The p27 protein is a canonical negative regulator of cell proliferation and acts primarily by inhibiting cyclin-dependent kinases (CDKs). Under some circumstances, p27 is associated with active CDK4, but no mechanism for activation has been described. We found that p27, when phosphorylated by tyrosine kinases, allosterically activated CDK4 in complex with cyclin D1 (CDK4-CycD1). Structural and biochemical data revealed that binding of phosphorylated p27 (phosp27) to CDK4 altered the kinase adenosine triphosphate site to promote phosphorylation of the retinoblastoma tumor suppressor protein (Rb) and other substrates. Surprisingly, purified and endogenous phosp27-CDK4-CycD1 complexes were insensitive to the CDK4-targeting drug palbociclib. Palbociclib instead primarily targeted monomeric CDK4 and CDK6 (CDK4/6) in breast tumor cells. Our data characterize phosp27-CDK4-CycD1 as an active Rb kinase that is refractory to clinically relevant CDK4/6 inhibitors.


Asunto(s)
Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 4 Dependiente de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Piperazinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Regulación Alostérica , Antineoplásicos/farmacología , Biocatálisis , Línea Celular Tumoral , Cristalografía por Rayos X , Ciclina D1/química , Quinasa 4 Dependiente de la Ciclina/química , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/química , Activación Enzimática , Humanos , Fosforilación , Conformación Proteica , Proteína de Retinoblastoma/metabolismo
5.
Cell Death Dis ; 10(6): 422, 2019 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-31142732

RESUMEN

The role of M1 macrophages (M1M)-derived exosomes in the progression of neointimal hyperplasia remains unclear now. Using a transwell co-culture system, we demonstrated that M1M contributed to functional change of vascular smooth muscle cell (VSMC). We further stimulated VSMCs with exosomes isolated from M1M. Our results demonstrated that these exosomes could be taken up by VSMCs through macropinocytosis. Using a microRNA array assay, we identified that miR-222 originated from M1M-derived exosomes triggered the functional changes of VSMCs. In addition, we confirmed that miR-222 played a key role in promoting VSMCs proliferation and migration by targeting Cyclin Dependent Kinase Inhibitor 1B (CDKN1B) and Cyclin Dependent Kinase Inhibitor 1C (CDKN1C) in vitro. In vivo, M1M-derived exosomes significantly aggravated neointima formation following carotid artery ligation injury and wire injury and these effects were partly abolished by miR-222 inhibitor 2'OMe-miR-222. Our findings thus suggest that exosomes derived from M1M could aggravate neointimal hyperplasia through delivering miR-222 into VSMCs. Future studies are warranted to validate if the post-injury vascular neointimal hyperplasia and restenosis could be attenuated by inhibiting miR-222.


Asunto(s)
Traumatismos de las Arterias Carótidas/patología , Exosomas/metabolismo , Hiperplasia/patología , Transducción de Señal , Regiones no Traducidas 3' , Compuestos de Anilina/farmacología , Animales , Antagomirs/metabolismo , Compuestos de Bencilideno/farmacología , Traumatismos de las Arterias Carótidas/veterinaria , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/química , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/química , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/metabolismo , Exosomas/trasplante , Hiperplasia/veterinaria , Macrófagos/citología , Macrófagos/metabolismo , Ratones , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , MicroARNs/metabolismo , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Neointima/patología , Células RAW 264.7
6.
Mol Cancer Ther ; 18(2): 482-493, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30425132

RESUMEN

The CDK inhibitor, p27kip1, encoded by the Cdkn1b gene can negatively modulate cell proliferation. The control of p27 activity during the cell cycle is regulated at multiple levels, including transcription, translation, and protein stability. The last residue of p27 (threonine 198 in human, threonine 197 in mouse) is involved in the control of protein stability. We have generated a murine knock-in model (Cdkn1b T197A) in which threonine 197 is replaced by alanine, which renders p27 protein highly unstable due to a high rate of proteasomal degradation. Expectedly, Cdkn1b T197A/T197A mice present with increased body size and weight, organomegaly, and multiple organ hyperplasia, similar to what is observed in Cdkn1b KO/KO mice. We investigated the effects exerted by the restoration of normal levels of p27 protein in the tissue of Cdkn1b T197A/T197A mice. We found that proteasome inhibition with bortezomib rescues the hyperplasia induced by the lack of p27 expression in Cdkn1b T197A/T197A but not in Cdkn1b KO/KO mice. However, BAY 11-7082, a proteasome inhibitor that stabilizes IκB but not p27, fails to rescue hyperplasia in Cdkn1b T197A/T197A mice. Bortezomib increases p27 half-life and reduces the proliferation in MEFs derived from Cdkn1b T197A/T197A but not from Cdkn1b WT/WT mice, whereas BAY 11-7082 had no effect on the protein levels of p27 and on the proliferation rate of Cdkn1b T197A/T197A MEFs.The results presented here demonstrate that Cdkn1b T197A/T197A mice represent an attractive in vivo model to investigate whether the targeting of p27 degradation machinery might prove beneficial in the treatment of a variety of human proliferative disorders caused by increased turnover of p27 protein.


Asunto(s)
Sustitución de Aminoácidos , Bortezomib/farmacología , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/química , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Modelos Animales , Animales , Técnicas de Sustitución del Gen , Hiperplasia , Ratones , Nitrilos/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Estabilidad Proteica , Proteolisis , Sulfonas/farmacología
7.
Angew Chem Int Ed Engl ; 58(10): 3114-3118, 2019 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-30570821

RESUMEN

Intrinsically disordered proteins have been reported to undergo disorder-to-order transitions upon binding to their partners in the cell. The extent of the ordering upon binding and the lack of order prior to binding is difficult to visualize with classical structure determination methods. Binding of p27 to the Cdk2/cyclin A complex is accompanied by partial folding of p27 in the KID domain, with the retention of dynamic behavior for function, particularly in the C-terminal half of the protein. Herein, native ion mobility mass spectrometry (IM-MS) is employed to measure the intrinsic dynamic properties of p27, both in isolation and within the trimeric complex with Cdk2/cyclin A. The trimeric Cdk2/cyclin A/p27-KID complex possesses significant structural heterogeneity compared to Cdk2/cyclin A. These findings support the formation of a fuzzy complex in which both the N- and C-termini of p27 interact with Cdk2/cyclin A in multiple, closely associated states.


Asunto(s)
Ciclina A/metabolismo , Quinasa 2 Dependiente de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Ciclina A/química , Quinasa 2 Dependiente de la Ciclina/química , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/química , Humanos , Proteínas Intrínsecamente Desordenadas/metabolismo , Espectrometría de Movilidad Iónica , Espectrometría de Masas , Unión Proteica , Conformación Proteica , Pliegue de Proteína , Dominios y Motivos de Interacción de Proteínas , Mapas de Interacción de Proteínas , Multimerización de Proteína
8.
Cell Physiol Biochem ; 51(2): 610-629, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30458449

RESUMEN

BACKGROUND/AIMS: MicroRNAs (miRNAs) or exosomes have recently been shown to play vital regulatory or communication roles in cancer biology. However, the roles and mechanisms of exosomal miRNAs in pancreatic ductal adenocarcinoma (PDAC) remain unknown. We aimed to investigate the detailed roles and mechanisms of tumor-generated exosomal miRNAs in progression of PDAC. METHODS: miR-222 was identified by miRNA microarray studies in exosomes of PDAC cells, and further analyzed in plasma exosomes of PDAC patients. The regulatory mechanisms of miR-222 were explored by qRT-PCR, WB, dual-luciferase assays and immunofluorescence or confocal analysis. Other biological assays include transwell, xenograft models and so on. RESULTS: miR-222 is significantly high in tumor exosomes or highly invasive PDAC cells. miR-222 could directly regulate p27 to promote cell invasion and proliferation. miR-222 could also activate AKT by inhibiting PPP2R2A expression, thus inducing p27 phosphorylation and cytoplasmic p27 expression to promote cell survival, invasion and metastasis. Expressions of miR-222 and p27 were significantly inversely correlated, and cytoplasmic p27, instead of nuclear p27, was associated with tumor malignancy. miR-222 could be transmitted between PDAC cells via exosome communication, and the exosomal miR-222 communication is functional. Plasma exosomal miR-222 in PDAC patients was high and significantly correlated to tumor size and TNM stage, and was an independent risk factor for PDAC patient survival. CONCLUSION: Tumor-generated exosomes could promote invasion and proliferation of neighboring tumor cells via miR-222 transmission, the plasma exosomal miR-222 plays important roles and may be a useful prognostic maker in PDAC.


Asunto(s)
Exosomas/metabolismo , MicroARNs/metabolismo , Neoplasias Pancreáticas/patología , Regiones no Traducidas 3' , Anciano , Animales , Antagomirs/metabolismo , Línea Celular Tumoral , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/química , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Progresión de la Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Desnudos , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/mortalidad , Fosforilación , Proteína Fosfatasa 2/metabolismo , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factores de Riesgo , Tasa de Supervivencia , Neoplasias Pancreáticas
9.
BMC Mol Biol ; 18(1): 23, 2017 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-28882106

RESUMEN

BACKGROUND: Cholangiocarcinoma is one of the deadly disease with poor 5-year survival and poor response to conventional therapies. Previously, we found that p27kip1 nuclear-cytoplasmic translocation confers proliferation potential to cholangiocarcinoma cell line QBC939 and this process is mediated by crm-1. However, no other post-transcriptional regulation was found in this process including sumoylation in cholangiocarcinoma. RESULTS: In this study, we explored the role of sumoylation in the nuclear-cytoplasmic translocation of p27kip1 and its involvement of QBC939 cells' proliferation. First, we identified K73 as the sumoylation site in p27kip1. By utilizing plasmid flag-p27kip1, HA-RanBP2, GST-RanBP2 and His-p27kip1 and immunoprecipitation assay, we validated that p27kip1 can serve as the sumoylation target of RanBP2 in QBC939. Furthermore, we confirmed crm-1's role in promoting nuclear-cytoplasmic translocation of p27kip1 and found that RanBP2's function relies on crm-1. However, K73R mutated p27kip1 can't be identified by crm-1 or RanBP2 in p27kip1 translocation process, suggesting sumoylation of p27kip1 via K73 site is necessary in this process by RanBP2 and crm-1. Phenotypically, the overexpression of either RanBP2 or crm-1 can partially rescue the anti-proliferative effect brought by p27kip1 overexpression in both the MTS and EdU assay. For the first time, we identified and validated the K73 sumoylation site in p27kip1, which is critical to RanBP2 and crm-1 in p27kip1 nuclear-cytoplasmic translocation process. CONCLUSION: Taken together, targeted inhibition of sumoylation of p27kip1 may serve as a potentially potent therapeutic target in the eradication of cholangiocarcinoma development and relapses.


Asunto(s)
Neoplasias de los Conductos Biliares/fisiopatología , Colangiocarcinoma/fisiopatología , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Chaperonas Moleculares/metabolismo , Proteínas de Complejo Poro Nuclear/metabolismo , Sumoilación , Transporte Activo de Núcleo Celular/genética , Neoplasias de los Conductos Biliares/genética , Línea Celular Tumoral , Proliferación Celular/genética , Colangiocarcinoma/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/química , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Expresión Génica , Humanos , Carioferinas/metabolismo , Chaperonas Moleculares/genética , Mutación , Proteínas de Complejo Poro Nuclear/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteína Exportina 1
10.
Cancer Lett ; 403: 354-365, 2017 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-28687353

RESUMEN

p27Kip1 is a cell cycle regulator firstly identified as a cyclin-dependent kinase inhibitor. For a long time, its function has been associated to cell cycle progression inhibition at G1/S boundary in response to antiproliferative stimuli. The picture resulted complicated by the discovery that p27Kip1 is an intrinsically unstructured protein, with numerous CDK-dependent and -independent functions and involvement in many cellular processes, such as cytoskeleton dynamics and cell motility control, apoptosis and autophagy activation. Depending on the cell context, these activities might turn to be oncogenic and stimulate cancer progression and metastatization. Nevertheless, p27Kip1 role in cancer biology suppression was underscored by myriad data reporting its down-regulation and/or cytoplasmic relocalization in different tumors, while usually no genetic alterations were found in human cancers, making the protein a non-canonical oncosuppressor. Recently, mostly due to advances in genomic analyses, CDKN1B, p27Kip1 encoding gene, has been found mutated in several cancers, thus leading to a profound reappraisal of CDKN1B role in tumorigenesis. This review summarizes the main p27Kip1 features, with major emphasis to its role in cancer biology and to the importance of CDKN1B mutations in tumor development.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Neoplasias/metabolismo , Animales , Biomarcadores de Tumor/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/química , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad , Humanos , Mutación , Neoplasias/química , Neoplasias/genética , Neoplasias/patología , Fenotipo , Conformación Proteica , Procesamiento Proteico-Postraduccional , Transducción de Señal , Relación Estructura-Actividad
11.
Angew Chem Int Ed Engl ; 56(25): 7070-7073, 2017 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-28508552

RESUMEN

The kinase inhibitory domain of the cell cycle regulatory protein p27Kip1 (p27) was nuclear spin hyperpolarized using dissolution dynamic nuclear polarization (D-DNP). While intrinsically disordered in isolation, p27 adopts secondary structural motifs, including an α-helical structure, upon binding to cyclin-dependent kinase 2 (Cdk2)/cyclin A. The sensitivity gains obtained with hyperpolarization enable the real-time observation of 13 C NMR signals during p27 folding upon binding to Cdk2/cyclin A on a time scale of several seconds. Time-dependent intensity changes are dependent on the extent of folding and binding, as manifested in differential spin relaxation. The analysis of signal decay rates suggests the existence of a partially folded p27 intermediate during the timescale of the D-DNP NMR experiment.


Asunto(s)
Proteínas Intrínsecamente Desordenadas/química , Espectroscopía de Resonancia Magnética/métodos , Pliegue de Proteína , Ciclina A/química , Quinasa 2 Dependiente de la Ciclina/química , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/química , Unión Proteica , Estructura Secundaria de Proteína , Solubilidad , Factores de Tiempo
12.
BMC Bioinformatics ; 18(1): 15, 2017 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-28056778

RESUMEN

BACKGROUND: The cyclin-dependent kinase 2 (CDK2) together with its cyclin E and A partners is a central regulator of cell growth and division. Deregulation of CDK2 activity is associated with diseases such as cancer. The analysis of substrates identified S/T-P-X-R/K/H as the CDK2 consensus sequence. The crystal structure of cyclin A/CDK2 with a short model peptide supports this sequence and identifies key interactions. However, CDKs use additional determinants to recognize substrates, including the RXL motif that is read by the cyclin subunits. We were interested to determine whether additional amino acids beyond the minimal consensus sequence of the well-studied substrate and tumor suppressor p27KIP1 were relevant for catalysis. RESULTS: To address whether additional amino acids, close to the minimal consensus sequence, play a role in binding, we investigate the interaction of cyclin A/CDK2 with an in vivo cellular partner and CDK inhibitor p27KIP1. This protein is an intrinsically unfolded protein and, in particular, the C-terminal half of the protein has not been accessible to structural analysis. This part harbors the CDK2 phosphorylation site. We used bioinformatics tools, including MODELLER, iTASSER and HADDOCK, along with partial structural information to build a model of the C-terminal region of p27KIP1 with cyclin A/CDK2. This revealed novel interactions beyond the consensus sequence with a proline and a basic amino acid at the P + 1 and the P + 3 sites, respectively. We suggest that the lysine at P + 2 might regulate the reversible association of the second counter ion in the active site of CDK2. The arginine at P + 7 interacts with both cyclin A and CDK2 and is important for the catalytic turnover rate. CONCLUSION: Our modeling identifies additional amino acids in p27KIP1 beyond the consensus sequence that contribute to the efficiency of substrate phosphorylation.


Asunto(s)
Quinasa 2 Dependiente de la Ciclina/química , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/química , Secuencia de Aminoácidos , Animales , Biología Computacional , Ciclina A/química , Ciclina E/química , Humanos , Fosforilación , Conformación Proteica , Spodoptera
13.
Sci Rep ; 6: 26450, 2016 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-27211815

RESUMEN

Cell-cycle regulatory protein, CDK2 is active when bound to its complementary partner protein, CyclinA or E. Recent discovery of the Kip/Cip family of proteins has indicated that the activity of CDK2 is also regulated by a member protein, p27. Although, the mechanism of CDK2 inhibition by p27 binding is known from crystal structure, little is known about the mechanism of CDK2 reactivation. Here, we execute classical and accelerated molecular dynamics simulations of unphosphorylated- and phosphorylated-p27 bound CDK2/CyclinA to unravel the CDK2 reactivation mechanism at molecular-to-atomic detail. Results suggest that the phosphorylation of p27 Y88 residue (pY88-p27) first disrupts the p27/CDK2 hybrid ß-sheet and subsequently ejects the p27 310 helix from CDK2 catalytic cleft. The unbinding of p27 from CDK2/CyclinA complex, thus, follows a two-step unfolding mechanism, where the 310 helix ejection constitutes the rate-limiting step. Interestingly, the unfolding of p27 leaves CDK2/CyclinA in an active state, where the prerequisite CDK2-CyclinA interfacial contacts were regained and ATP achieved its native position for smooth transfer of phosphate. Our findings match very well with NMR chemical shift data that indicated the flip-out of p27 310 helix from CDK2 pocket and kinetic experiments that exhibited significant kinase activity of CDK2 when saturated with pY88-p27.


Asunto(s)
Quinasa 2 Dependiente de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/química , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Eucariontes/metabolismo , Animales , Dominio Catalítico , Ciclina A/metabolismo , Activación Enzimática , Eucariontes/química , Humanos , Modelos Moleculares , Simulación de Dinámica Molecular , Fosforilación , Estructura Secundaria de Proteína , Desplegamiento Proteico , Tirosina/química
14.
Nucleic Acids Res ; 44(W1): W367-74, 2016 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-27198219

RESUMEN

Residue interaction networks (RINs) are an alternative way of representing protein structures where nodes are residues and arcs physico-chemical interactions. RINs have been extensively and successfully used for analysing mutation effects, protein folding, domain-domain communication and catalytic activity. Here we present RING 2.0, a new version of the RING software for the identification of covalent and non-covalent bonds in protein structures, including π-π stacking and π-cation interactions. RING 2.0 is extremely fast and generates both intra and inter-chain interactions including solvent and ligand atoms. The generated networks are very accurate and reliable thanks to a complex empirical re-parameterization of distance thresholds performed on the entire Protein Data Bank. By default, RING output is generated with optimal parameters but the web server provides an exhaustive interface to customize the calculation. The network can be visualized directly in the browser or in Cytoscape. Alternatively, the RING-Viz script for Pymol allows visualizing the interactions at atomic level in the structure. The web server and RING-Viz, together with an extensive help and tutorial, are available from URL: http://protein.bio.unipd.it/ring.


Asunto(s)
Algoritmos , Ciclina A/química , Quinasa 2 Dependiente de la Ciclina/química , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/química , Dominios y Motivos de Interacción de Proteínas , Programas Informáticos , Secuencia de Aminoácidos , Dominio Catalítico , Gráficos por Computador , Ciclina A/genética , Quinasa 2 Dependiente de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Bases de Datos Genéticas , Enlace de Hidrógeno , Internet , Ligandos , Aprendizaje Automático , Modelos Moleculares , Unión Proteica , Pliegue de Proteína , Estructura Secundaria de Proteína , Análisis de Secuencia de Proteína , Electricidad Estática
15.
Mol Carcinog ; 55(2): 220-9, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25594310

RESUMEN

We previously found that the small GTPase Rheb regulates the cell-cycle inhibitor p27KIP1 (p27) in colon cancer cells by a mTORC1-independent mechanism. However, the biological function of the Rheb/p27 axis in cancer cells remains unknown. Here, we show that siRNA-mediated depletion of Rheb decreases survival of human colon cancer cells under serum deprivation. As autophagy can support cell survival, we analyzed the effect of Rheb on this process by detecting the modification of the autophagy marker protein LC3 by western blot and imunofluorescence. We found that Rheb promotes autophagy in several human cancer cell lines under serum deprivation. Accordingly, blocking autophagy inhibited the pro-survival effect of Rheb in colon cancer cells. We then analyzed whether p27 was involved in the biological effect of Rheb. Depletion of p27 inhibited colon cancer cell survival, and Rheb induction of autophagy. These results suggest that p27 has an essential role in the effect of Rheb in response to serum deprivation. In addition, we demonstrated that the role of p27 in autophagy stands on the N-terminal portion of the protein, where the CDK-inhibitory domain is located. Our results indicate that a Rheb/p27 axis accounts for the activation of autophagy that supports cancer cell survival. Our work therefore highlights a biological function of Rheb and prompts the need for future studies to address whether the mTORC1-independent Rheb/p27 axis could contribute to tumorigenesis and/or resistance to mTOR inhibitors.


Asunto(s)
Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Neoplasias/metabolismo , Neuropéptidos/metabolismo , Autofagia , Sitios de Unión , Línea Celular Tumoral , Supervivencia Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/química , Humanos , Proteína Homóloga de Ras Enriquecida en el Cerebro , Estrés Fisiológico
16.
Mol Med Rep ; 11(2): 1435-41, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25351256

RESUMEN

MicroRNAs (miRNAs) are closely associated with cell proliferation, invasion and metastasis in various types of cancer, including prostate cancer. In this study, the role of miR-429 in the regulation of cell proliferation was investigated in prostate cancer cells. miR-429 expression levels were measured in the IF11 and IA8 prostate cancer cell lines and normal prostate epithelial tissues by quantitative polymerase chain reaction. miR-429 mimics or an miR-429 inhibitor were then transfected into the human prostate cancer cell lines. MTT and fluorescence-activated cell sorting were used to detect the effect of miR-429 on cell proliferation. A luciferase reporter system was employed to verify the potential target of miR-429. The results revealed that miR-429 was significantly upregulated in the human prostate cancer cell lines, compared with the normal prostate epithelial tissue. Downregulation of miR-429 expression in IF11 and IA8 cells inhibited cell proliferation and arrested the cells in the G1 phase of the cell cycle. The luciferase assay demonstrated that p27Kip1 was a direct target of miR-429. Furthermore, overexpression of p27Kip1 was observed to partially rescue the proliferation­promoting effect of miR-429 on IA8 cells. In conclusion, to the best of our knowledge this study was the first to show that miR-429 is involved in the oncogenesis of prostate cancer and thus may be a novel prognostic biomarker in prostate cancer.


Asunto(s)
Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Regulación hacia Abajo , MicroARNs/metabolismo , Neoplasias de la Próstata/genética , Regiones no Traducidas 3' , Secuencia de Bases , Sitios de Unión , Línea Celular Tumoral , Proliferación Celular/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/química , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Puntos de Control de la Fase G1 del Ciclo Celular , Humanos , Masculino , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Neoplasias de la Próstata/patología , ARN Bicatenario/metabolismo , Alineación de Secuencia , Transfección , Regulación hacia Arriba
17.
Cell Signal ; 26(10): 2071-85, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24949832

RESUMEN

p27 has essential roles in cellular proliferation and migration, and reduced or cytoplasmic p27 is associated with poor clinical outcomes in a variety of human tumours. Jun activation domain-binding protein (Jab1)/constitutive photomorphogenic-9 signalosome 5 (CSN5) directly interacts with p27 promoting its translocation and cytoplasmic degradation. Sec6 is a component of the exocyst complex. Recently, several studies revealed that Sec6 has specific functions in migration, adhesion, and cell differentiation. However, how Sec6 is involved in the regulation of cell cycle progression is unknown. The present study shows that Sec6 regulates cytoplasmic translocation of p27 through p27 phosphorylation at Thr157, thereby promoting p27 degradation in the cytoplasm via interaction with Jab1 and Siah1 and suppressing cell cycle progression.


Asunto(s)
Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Citoplasma/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Nucleares/metabolismo , Péptido Hidrolasas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Proteínas 14-3-3/química , Proteínas 14-3-3/metabolismo , Complejo del Señalosoma COP9 , Puntos de Control del Ciclo Celular , Línea Celular Tumoral , Núcleo Celular/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/química , Regulación de la Expresión Génica , Células HEK293 , Humanos , Inmunoprecipitación , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Nucleares/química , Péptido Hidrolasas/genética , Fosforilación , Unión Proteica , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Translocación Genética , Ubiquitina-Proteína Ligasas/química , Proteínas de Transporte Vesicular/antagonistas & inhibidores , Proteínas de Transporte Vesicular/genética
18.
Mol Med Rep ; 10(2): 855-9, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24859599

RESUMEN

An increasing body of evidence indicates that microRNAs (miRNAs), a class of small non­coding RNAs, are often aberrantly expressed in human osteosarcoma. This study aimed to investigate the effects of miR­25 and to identify its potential target genes in osteosarcoma (OS) cells. First, the expression of miR­25 was detected by reverse transcription­quantitative polymerase chain reaction (RT-qPCR), which revealed a significant upregulation of miR­25 in osteosarcoma tissues compared to the adjacent healthy tissues. To investigate the role of miR­25 in osteosarcoma cell proliferation, the miR­25 precursor was next transfected into Saos­2 and U2OS cells. Overexpression of miR­25 promoted cell proliferation in vitro and tumor growth in a xenograft mouse model. In addition, our results revealed that the protein expression of p27, a cell­cycle inhibitor, is negatively regulated by miR­25. Restoring the p27 level in miR­25­overexpressing cells reversed the enhancing effect of miR­25 on cell proliferation. Therefore, miR­25 may act as an onco­miRNA in osteosarcoma, which provides new perspectives in cancer treatment strategies based on molecular targeting.


Asunto(s)
Neoplasias Óseas/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , MicroARNs/metabolismo , Osteosarcoma/genética , Regiones no Traducidas 3' , Animales , Secuencia de Bases , Neoplasias Óseas/patología , Línea Celular Tumoral , Proliferación Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/química , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/química , Osteosarcoma/patología , Alineación de Secuencia , Trasplante Heterólogo
19.
J Virol ; 87(24): 13226-38, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24067984

RESUMEN

The Kaposi's sarcoma-associated herpesvirus (KSHV) immediate-early gene, replication, and transcription activator (K-Rta) is a key viral protein that serves as the master regulator for viral lytic replication. In this study, we investigated the role of K-Rta in cell cycle regulation and found that the expression of K-Rta in doxycycline (Dox)-inducible BJAB cells induced cell cycle arrest in G0/G1 phase. Western blot analysis of key cell cycle regulators revealed that K-Rta-mediated cell cycle arrest was associated with a decrease in cyclin A and phosphorylated Rb (pS807/pS811) protein levels, both markers of S phase progression, and an increase in protein levels for p27, a cyclin-dependent kinase inhibitor. Further, we found that K-Rta does not affect the transcription of p27 but regulates p27 at the posttranslational level by inhibiting its proteosomal degradation. Immunofluorescence staining and cell fractionation experiments revealed largely nuclear compartmentalization of p27 in K-Rta-expressing cells, demonstrating that K-Rta not only stabilizes p27 but also modulates its cellular localization. Finally, short hairpin RNA knockdown of p27 significantly abrogates cell cycle arrest in K-Rta-expressing cells, supporting its key role in K-Rta-mediated cell cycle arrest. Our findings are consistent with previous studies which showed that expression of immediate-early genes of several herpesviruses, including herpes simplex virus, Epstein-Barr virus, and cytomegalovirus, results in cell cycle arrest at the G0/G1 phase, possibly to avoid competition for resources needed for host cell replication during the S phase.


Asunto(s)
Núcleo Celular/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Puntos de Control de la Fase G1 del Ciclo Celular , Infecciones por Herpesviridae/metabolismo , Infecciones por Herpesviridae/fisiopatología , Herpesvirus Humano 8/metabolismo , Proteínas Inmediatas-Precoces/metabolismo , Transactivadores/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/química , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Regulación Viral de la Expresión Génica , Infecciones por Herpesviridae/genética , Infecciones por Herpesviridae/virología , Herpesvirus Humano 8/genética , Humanos , Proteínas Inmediatas-Precoces/genética , Estabilidad Proteica , Transporte de Proteínas , Fase de Descanso del Ciclo Celular , Transactivadores/genética
20.
Cancer Sci ; 104(11): 1461-7, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23910095

RESUMEN

Dysregulation of p27(Kip1) due to proteolysis that involves the ubiquitin ligase (SCF) complex with S-phase kinase-associated protein 2 (Skp2) as the substrate-recognition component (SCF(Skp2)) frequently results in tumorigenesis. In this report, we developed a high-throughput screening system to identify small-molecule inhibitors of p27(Kip1) degradation. This system was established by tagging Skp2 with fluorescent monomeric Azami Green (mAG) and CDK subunit 1 (Cks1) (mAGSkp2-Cks1) to bind to p27(Kip1) phosphopeptides. We identified two compounds that inhibited the interaction between mAGSkp2-Cks1 and p27(Kip1): linichlorin A and gentian violet. Further studies have shown that the compounds inhibit the ubiquitination of p27(Kip1) in vitro as well as p27(Kip1) degradation in HeLa cells. Notably, both compounds exhibited preferential antiproliferative activity against HeLa and tsFT210 cells compared with NIH3T3 cells and delayed the G1 phase progression in tsFT210 cells. Our approach indicates a potential strategy for restoring p27(Kip1) levels in human cancers.


Asunto(s)
Antineoplásicos/farmacología , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Violeta de Genciana/farmacología , Sesquiterpenos/farmacología , Ubiquitinación/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Antineoplásicos/química , Puntos de Control del Ciclo Celular , Proliferación Celular/efectos de los fármacos , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/química , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Quinasas Ciclina-Dependientes/metabolismo , Ensayos de Selección de Medicamentos Antitumorales , Violeta de Genciana/química , Células HeLa , Ensayos Analíticos de Alto Rendimiento , Humanos , Ratones , Datos de Secuencia Molecular , Células 3T3 NIH , Fragmentos de Péptidos/química , Estabilidad Proteica , Proteolisis/efectos de los fármacos , Sesquiterpenos/química , Bibliotecas de Moléculas Pequeñas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA