Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
1.
Int J Mol Sci ; 23(3)2022 Jan 21.
Article in English | MEDLINE | ID: mdl-35163135

ABSTRACT

The cell cycle consists of successive events that lead to the generation of new cells. The cell cycle is regulated by different cyclins, cyclin-dependent kinases (CDKs) and their inhibitors, such as p27Kip1. At the nuclear level, p27Kip1 has the ability to control the evolution of different phases of the cell cycle and oppose cell cycle progression by binding to CDKs. In the cytoplasm, diverse functions have been described for p27Kip1, including microtubule remodeling, axonal transport and phagocytosis. In Alzheimer's disease (AD), alterations to cycle events and a purported increase in neurogenesis have been described in the early disease process before significant pathological changes could be detected. However, most neurons cannot progress to complete their cell division and undergo apoptotic cell death. Increased levels of both the p27Kip1 levels and phosphorylation status have been described in AD. Increased levels of Aß42, tau hyperphosphorylation or even altered insulin signals could lead to alterations in p27Kip1 post-transcriptional modifications, causing a disbalance between the levels and functions of p27Kip1 in the cytoplasm and nucleus, thus inducing an aberrant cell cycle re-entry and alteration of extra cell cycle functions. Further studies are needed to completely understand the role of p27Kip1 in AD and the therapeutic opportunities associated with the modulation of this target.


Subject(s)
Alzheimer Disease/pathology , Cell Cycle , Cyclin-Dependent Kinase Inhibitor p27/chemistry , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Protein Processing, Post-Translational , Alzheimer Disease/metabolism , Animals , Humans
2.
Cells ; 10(9)2021 08 31.
Article in English | MEDLINE | ID: mdl-34571903

ABSTRACT

The Cyclin-dependent kinase (CDK) regulator p27Kip1 is a gatekeeper of G1/S transition. It also regulates G2/M progression and cytokinesis completion, via CDK-dependent or -independent mechanisms. Recently, other important p27Kip1 functions have been described, including the regulation of cell motility and migration, the control of cell differentiation program and the activation of apoptosis/autophagy. Several factors modulate p27Kip1 activities, including its level, cellular localization and post-translational modifications. As a matter of fact, the protein is phosphorylated, ubiquitinated, SUMOylated, O-linked N-acetylglicosylated and acetylated on different residues. p27Kip1 belongs to the family of the intrinsically unstructured proteins and thus it is endowed with a large flexibility and numerous interactors, only partially identified. In this review, we look at p27Kip1 properties and ascribe part of its heterogeneous functions to the ability to act as an anchor or scaffold capable to participate in the construction of different platforms for modulating cell response to extracellular signals and allowing adaptation to environmental changes.


Subject(s)
Cell Cycle , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Intrinsically Disordered Proteins/metabolism , Protein Processing, Post-Translational , Tissue Scaffolds/chemistry , Cyclin-Dependent Kinase Inhibitor p27/chemistry , Humans , Intrinsically Disordered Proteins/chemistry
3.
J Mol Biol ; 433(18): 167120, 2021 09 03.
Article in English | MEDLINE | ID: mdl-34197833

ABSTRACT

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.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/chemistry , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Drug Design , Drug Discovery , Intrinsically Disordered Proteins/metabolism , Small Molecule Libraries/metabolism , Humans , Intrinsically Disordered Proteins/chemistry , Protein Binding , Small Molecule Libraries/chemistry
4.
J Mol Biol ; 433(5): 166795, 2021 03 05.
Article in English | MEDLINE | ID: mdl-33422522

ABSTRACT

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.


Subject(s)
Cyclin A/chemistry , Cyclin-Dependent Kinase 2/chemistry , Cyclin-Dependent Kinase Inhibitor p27/chemistry , G1 Phase Cell Cycle Checkpoints , S-Phase Kinase-Associated Proteins/chemistry , Binding Sites , CDC2-CDC28 Kinases/chemistry , CDC2-CDC28 Kinases/genetics , CDC2-CDC28 Kinases/metabolism , Cyclin A/genetics , Cyclin A/metabolism , Cyclin E/chemistry , Cyclin E/genetics , Cyclin E/metabolism , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Gene Expression Regulation , HEK293 Cells , Humans , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Proteolysis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , S-Phase Kinase-Associated Proteins/genetics , S-Phase Kinase-Associated Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Signal Transduction
5.
J Mol Biol ; 432(9): 2998-3017, 2020 04 17.
Article in English | MEDLINE | ID: mdl-32088186

ABSTRACT

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.


Subject(s)
Cyclin A/metabolism , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase Inhibitor p27/chemistry , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Single Molecule Imaging/methods , Binding Sites , Cyclin A/chemistry , Cyclin-Dependent Kinase 2/chemistry , Cyclin-Dependent Kinase Inhibitor p27/genetics , Fluorescence Resonance Energy Transfer , Humans , Models, Molecular , Mutation , Protein Binding , Protein Conformation , Spectrometry, Fluorescence , Ternary Complex Factors/chemistry
6.
Science ; 366(6471)2019 12 13.
Article in English | MEDLINE | ID: mdl-31831640

ABSTRACT

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.


Subject(s)
Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Allosteric Regulation , Antineoplastic Agents/pharmacology , Biocatalysis , Cell Line, Tumor , Crystallography, X-Ray , Cyclin D1/chemistry , Cyclin-Dependent Kinase 4/chemistry , Cyclin-Dependent Kinase Inhibitor p27/chemistry , Enzyme Activation , Humans , Phosphorylation , Protein Conformation , Retinoblastoma Protein/metabolism
7.
Zygote ; 27(3): 187-189, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31182173

ABSTRACT

SummaryWe report here the existence of bands of higher molecular weight after western blot analysis in three proteins - Skp1, p27 and IκBα in bovine preimplantation embryos. This finding is specific to preimplantation embryos (from the 2-cell stage to the blastocyst stage) and not differentiated fibroblast cells in which these bands were of expected molecular weight. We suggest that these bands of higher molecular weight represent a complex of proteins that are characteristic of preimplantation embryos.


Subject(s)
Blastocyst/metabolism , Embryonic Development , Proteins/metabolism , Animals , Blastocyst/cytology , Blotting, Western , Cattle , Cyclin-Dependent Kinase Inhibitor p27/chemistry , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Female , Molecular Weight , NF-KappaB Inhibitor alpha/chemistry , NF-KappaB Inhibitor alpha/metabolism , Proteins/chemistry , S-Phase Kinase-Associated Proteins/chemistry , S-Phase Kinase-Associated Proteins/metabolism
8.
Cell Death Dis ; 10(6): 422, 2019 05 29.
Article in English | MEDLINE | ID: mdl-31142732

ABSTRACT

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.


Subject(s)
Carotid Artery Injuries/pathology , Exosomes/metabolism , Hyperplasia/pathology , Signal Transduction , 3' Untranslated Regions , Aniline Compounds/pharmacology , Animals , Antagomirs/metabolism , Benzylidene Compounds/pharmacology , Carotid Artery Injuries/veterinary , Cell Movement/drug effects , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p27/chemistry , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Cyclin-Dependent Kinase Inhibitor p57/chemistry , Cyclin-Dependent Kinase Inhibitor p57/genetics , Cyclin-Dependent Kinase Inhibitor p57/metabolism , Exosomes/transplantation , Hyperplasia/veterinary , Macrophages/cytology , Macrophages/metabolism , Mice , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , MicroRNAs/metabolism , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Neointima/pathology , RAW 264.7 Cells
9.
J Am Chem Soc ; 141(12): 4908-4918, 2019 03 27.
Article in English | MEDLINE | ID: mdl-30823702

ABSTRACT

The global dimensions and amplitudes of conformational fluctuations of intrinsically disordered proteins are governed, in part, by the linear segregation versus clustering of oppositely charged residues within the primary sequence. Ion mobility-mass spectrometry (IM-MS) affords unique advantages for probing the conformational consequences of the linear patterning of oppositely charged residues because it measures and separates proteins electrosprayed from solution on the basis of charge and shape. Here, we use IM-MS to measure the conformational consequences of charge patterning on the C-terminal intrinsically disordered region (p27 IDR) of the cell cycle inhibitory protein p27Kip1. We report the range of charge states and accompanying collisional cross section distributions for wild-type p27 IDR and two variants with identical amino acid compositions, κ14 and κ56, distinguished by the extent of linear mixing versus segregation of oppositely charged residues. Wild-type p27 IDR (κ31) and κ14, where the oppositely charged residues are more evenly distributed, exhibit a broad distribution of charge states. This is concordant with high degrees of conformational heterogeneity in solution. By contrast, κ56 with linear segregation of oppositely charged residues leads to limited conformational heterogeneity and a narrow distribution of charged states. Gas-phase molecular dynamics simulations demonstrate that the interplay between chain solvation and intrachain interactions (self-solvation) leads to conformational distributions that are modulated by salt concentration, with the wild-type sequence showing the most sensitivity to changes in salt concentration. These results suggest that the charge patterning within the wild-type p27 IDR may be optimized to sample both highly solvated and self-solvated conformational states.


Subject(s)
Intrinsically Disordered Proteins/chemistry , Mass Spectrometry , Cyclin-Dependent Kinase Inhibitor p27/chemistry , Molecular Dynamics Simulation , Protein Conformation
10.
Mol Cancer Ther ; 18(2): 482-493, 2019 02.
Article in English | MEDLINE | ID: mdl-30425132

ABSTRACT

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.


Subject(s)
Amino Acid Substitution , Bortezomib/pharmacology , Cyclin-Dependent Kinase Inhibitor p27/chemistry , Cyclin-Dependent Kinase Inhibitor p27/genetics , Models, Animal , Animals , Gene Knock-In Techniques , Hyperplasia , Mice , Nitriles/pharmacology , Proteasome Endopeptidase Complex/metabolism , Protein Stability , Proteolysis , Sulfones/pharmacology
11.
Angew Chem Int Ed Engl ; 58(10): 3114-3118, 2019 03 04.
Article in English | MEDLINE | ID: mdl-30570821

ABSTRACT

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.


Subject(s)
Cyclin A/metabolism , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Cyclin A/chemistry , Cyclin-Dependent Kinase 2/chemistry , Cyclin-Dependent Kinase Inhibitor p27/chemistry , Humans , Intrinsically Disordered Proteins/metabolism , Ion Mobility Spectrometry , Mass Spectrometry , Protein Binding , Protein Conformation , Protein Folding , Protein Interaction Domains and Motifs , Protein Interaction Maps , Protein Multimerization
12.
Cell Physiol Biochem ; 51(2): 610-629, 2018.
Article in English | MEDLINE | ID: mdl-30458449

ABSTRACT

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.


Subject(s)
Exosomes/metabolism , MicroRNAs/metabolism , Pancreatic Neoplasms/pathology , 3' Untranslated Regions , Aged , Animals , Antagomirs/metabolism , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p27/chemistry , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Disease Progression , Female , Humans , Male , Mice , Mice, Nude , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/mortality , Phosphorylation , Protein Phosphatase 2/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Risk Factors , Survival Rate , Pancreatic Neoplasms
13.
BMC Mol Biol ; 18(1): 23, 2017 09 07.
Article in English | MEDLINE | ID: mdl-28882106

ABSTRACT

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.


Subject(s)
Bile Duct Neoplasms/physiopathology , Cholangiocarcinoma/physiopathology , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Molecular Chaperones/metabolism , Nuclear Pore Complex Proteins/metabolism , Sumoylation , Active Transport, Cell Nucleus/genetics , Bile Duct Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cholangiocarcinoma/genetics , Cyclin-Dependent Kinase Inhibitor p27/chemistry , Cyclin-Dependent Kinase Inhibitor p27/genetics , Gene Expression , Humans , Karyopherins/metabolism , Molecular Chaperones/genetics , Mutation , Nuclear Pore Complex Proteins/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Exportin 1 Protein
14.
Cancer Lett ; 403: 354-365, 2017 09 10.
Article in English | MEDLINE | ID: mdl-28687353

ABSTRACT

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.


Subject(s)
Biomarkers, Tumor/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Neoplasms/metabolism , Animals , Biomarkers, Tumor/genetics , Cyclin-Dependent Kinase Inhibitor p27/chemistry , Cyclin-Dependent Kinase Inhibitor p27/genetics , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Humans , Mutation , Neoplasms/chemistry , Neoplasms/genetics , Neoplasms/pathology , Phenotype , Protein Conformation , Protein Processing, Post-Translational , Signal Transduction , Structure-Activity Relationship
15.
Angew Chem Int Ed Engl ; 56(25): 7070-7073, 2017 06 12.
Article in English | MEDLINE | ID: mdl-28508552

ABSTRACT

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.


Subject(s)
Intrinsically Disordered Proteins/chemistry , Magnetic Resonance Spectroscopy/methods , Protein Folding , Cyclin A/chemistry , Cyclin-Dependent Kinase 2/chemistry , Cyclin-Dependent Kinase Inhibitor p27/chemistry , Protein Binding , Protein Structure, Secondary , Solubility , Time Factors
16.
BMC Bioinformatics ; 18(1): 15, 2017 Jan 05.
Article in English | MEDLINE | ID: mdl-28056778

ABSTRACT

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.


Subject(s)
Cyclin-Dependent Kinase 2/chemistry , Cyclin-Dependent Kinase Inhibitor p27/chemistry , Amino Acid Sequence , Animals , Computational Biology , Cyclin A/chemistry , Cyclin E/chemistry , Humans , Phosphorylation , Protein Conformation , Spodoptera
17.
Nucleic Acids Res ; 44(W1): W367-74, 2016 07 08.
Article in English | MEDLINE | ID: mdl-27198219

ABSTRACT

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.


Subject(s)
Algorithms , Cyclin A/chemistry , Cyclin-Dependent Kinase 2/chemistry , Cyclin-Dependent Kinase Inhibitor p27/chemistry , Protein Interaction Domains and Motifs , Software , Amino Acid Sequence , Catalytic Domain , Computer Graphics , Cyclin A/genetics , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase Inhibitor p27/genetics , Databases, Genetic , Hydrogen Bonding , Internet , Ligands , Machine Learning , Models, Molecular , Protein Binding , Protein Folding , Protein Structure, Secondary , Sequence Analysis, Protein , Static Electricity
18.
Sci Rep ; 6: 26450, 2016 05 23.
Article in English | MEDLINE | ID: mdl-27211815

ABSTRACT

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.


Subject(s)
Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase Inhibitor p27/chemistry , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Eukaryota/metabolism , Animals , Catalytic Domain , Cyclin A/metabolism , Enzyme Activation , Eukaryota/chemistry , Humans , Models, Molecular , Molecular Dynamics Simulation , Phosphorylation , Protein Structure, Secondary , Protein Unfolding , Tyrosine/chemistry
19.
Proc Natl Acad Sci U S A ; 113(20): 5616-21, 2016 May 17.
Article in English | MEDLINE | ID: mdl-27140628

ABSTRACT

Peptide motifs embedded within intrinsically disordered regions (IDRs) of proteins are often the sites of posttranslational modifications that control cell-signaling pathways. How do IDR sequences modulate the functionalities of motifs? We answer this question using the polyampholytic C-terminal IDR of the cell cycle inhibitory protein p27(Kip1) (p27). Phosphorylation of Thr-187 (T187) within the p27 IDR controls entry into S phase of the cell division cycle. Additionally, the conformational properties of polyampholytic sequences are predicted to be influenced by the linear patterning of oppositely charged residues. Therefore, we designed sequence variants of the p27 IDR to alter charge patterning outside the primary substrate motif containing T187. Computer simulations and biophysical measurements confirm predictions regarding the impact of charge patterning on the global dimensions of IDRs. Through functional studies, we uncover cryptic sequence features within the p27 IDR that influence the efficiency of T187 phosphorylation. Specifically, we find a positive correlation between T187 phosphorylation efficiency and the weighted net charge per residue of an auxiliary motif. We also find that accumulation of positive charges within the auxiliary motif can diminish the efficiency of T187 phosphorylation because this increases the likelihood of long-range intra-IDR interactions that involve both the primary and auxiliary motifs and inhibit their contributions to function. Importantly, our findings suggest that the cryptic sequence features of the WT p27 IDR negatively regulate T187 phosphorylation signaling. Our approaches provide a generalizable strategy for uncovering the influence of sequence contexts on the functionalities of primary motifs in other IDRs.


Subject(s)
Cell Cycle/physiology , Cyclin-Dependent Kinase Inhibitor p27/physiology , Signal Transduction/physiology , Amino Acid Motifs , Amino Acid Sequence , Cyclin-Dependent Kinase Inhibitor p27/chemistry , Humans , Phosphorylation , Protein Conformation
20.
Proteins ; 84(4): 544-60, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26994550

ABSTRACT

Conformational ensembles of intrinsically disordered peptide chains are not fully determined by experimental observations. Uncertainty due to lack of experimental restraints and due to intrinsic disorder can be distinguished if distance distributions restraints are available. Such restraints can be obtained from pulsed dipolar electron paramagnetic resonance (EPR) spectroscopy applied to pairs of spin labels. Here, we introduce a Monte Carlo approach for generating conformational ensembles that are consistent with a set of distance distribution restraints, backbone dihedral angle statistics in known protein structures, and optionally, secondary structure propensities or membrane immersion depths. The approach is tested with simulated restraints for a terminal and an internal loop and for a protein with 69 residues by using sets of sparse restraints for underlying well-defined conformations and for published ensembles of a premolten globule-like and a coil-like intrinsically disordered protein.


Subject(s)
Algorithms , Cyclin-Dependent Kinase 2/chemistry , Cyclin-Dependent Kinase Inhibitor p27/chemistry , Intrinsically Disordered Proteins/chemistry , Models, Molecular , Binding Sites , Electron Spin Resonance Spectroscopy , Monte Carlo Method , Protein Binding , Protein Domains , Protein Folding , Protein Structure, Secondary , Spin Labels
SELECTION OF CITATIONS
SEARCH DETAIL