Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
1.
Sci Rep ; 13(1): 18399, 2023 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-37884585

RESUMEN

Inhibiting protein-protein interactions of the Myc family is a viable pharmacological strategy for modulation of the levels of Myc oncoproteins in cancer. Aurora A kinase (AurA) and N-Myc interaction is one of the most attractive targets of this strategy because formation of this complex blocks proteasomal degradation of N-Myc in neuroblastoma. Two crystallization studies have captured this complex (PDB IDs: 5g1x, 7ztl), partially resolving the AurA interaction region (AIR) of N-Myc. Prompted by the missing N-Myc fragment in these crystal structures, we modeled the complete structure between AurA and N-Myc, and comprehensively analyzed how the incomplete and complete N-Myc behave in complex by molecular dynamics simulations. Molecular dynamics simulations of the incomplete PDB complex (5g1x) repeatedly showed partial dissociation of the short N-Myc fragment (61-89) from the kinase. The missing N-Myc (19-60) fragment was modeled utilizing the N-terminal lobe of AurA as the protein-protein interaction surface, wherein TPX2, a well-known partner of AurA, also binds. Binding free energy calculations along with flexibility analysis confirmed that the complete AIR of N-Myc stabilizes the complex, accentuating the N-terminal lobe of AurA as a binding site for the missing N-Myc fragment (19-60). We further generated additional models consisting of only the missing N-Myc (19-60), and the fused form of TPX2 (7-43) and N-Myc (61-89). These partners also formed more stable interactions with the N-terminal lobe of AurA than did the incomplete N-Myc fragment (61-89) in the 5g1x complex. Altogether, this study provides structural insights into the involvement of the N-terminus of the AIR of N-Myc and the N-terminal lobe of AurA in formation of a stable complex, reflecting its potential for effective targeting of N-Myc.


Asunto(s)
Aurora Quinasa A , Epilepsia , Proteína Proto-Oncogénica N-Myc , Neuroblastoma , Humanos , Aurora Quinasa A/química , Sitios de Unión , Simulación de Dinámica Molecular
2.
Proc Natl Acad Sci U S A ; 120(34): e2304611120, 2023 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-37590418

RESUMEN

Selective orthosteric inhibition of kinases has been challenging due to the conserved active site architecture of kinases and emergence of resistance mutants. Simultaneous inhibition of distant orthosteric and allosteric sites, which we refer to as "double-drugging", has recently been shown to be effective in overcoming drug resistance. However, detailed biophysical characterization of the cooperative nature between orthosteric and allosteric modulators has not been undertaken. Here, we provide a quantitative framework for double-drugging of kinases employing isothermal titration calorimetry, Förster resonance energy transfer, coupled-enzyme assays, and X-ray crystallography. We discern positive and negative cooperativity for Aurora A kinase (AurA) and Abelson kinase (Abl) with different combinations of orthosteric and allosteric modulators. We find that a conformational equilibrium shift is the main principle governing cooperativity. Notably, for both kinases, we find a synergistic decrease of the required orthosteric and allosteric drug dosages when used in combination to inhibit kinase activities to clinically relevant inhibition levels. X-ray crystal structures of the double-drugged kinase complexes reveal the molecular principles underlying the cooperative nature of double-drugging AurA and Abl with orthosteric and allosteric inhibitors. Finally, we observe a fully closed conformation of Abl when bound to a pair of positively cooperative orthosteric and allosteric modulators, shedding light on the puzzling abnormality of previously solved closed Abl structures. Collectively, our data provide mechanistic and structural insights into rational design and evaluation of double-drugging strategies.


Asunto(s)
Aurora Quinasa A , Mesilato de Imatinib , Niacinamida , Inhibidores de Proteínas Quinasas , Proteínas Proto-Oncogénicas c-abl , Humanos , Cristalografía por Rayos X , Mesilato de Imatinib/química , Mesilato de Imatinib/farmacología , Niacinamida/química , Niacinamida/farmacología , Proteínas Proto-Oncogénicas c-abl/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-abl/química , Aurora Quinasa A/antagonistas & inhibidores , Aurora Quinasa A/química , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología
3.
Protein Sci ; 32(9): e4750, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37572333

RESUMEN

Control of eukaryotic cellular function is heavily reliant on the phosphorylation of proteins at specific amino acid residues, such as serine, threonine, tyrosine, and histidine. Protein kinases that are responsible for this process comprise one of the largest families of evolutionarily related proteins. Dysregulation of protein kinase signaling pathways is a frequent cause of a large variety of human diseases including cancer, autoimmune, neurodegenerative, and cardiovascular disorders. In this study, we mapped all pathogenic mutations in 497 human protein kinase domains from the ClinVar database to the reference structure of Aurora kinase A (AURKA) and grouped them by the relevance to the disease type. Our study revealed that the majority of mutation hotspots associated with cancer are situated within the catalytic and activation loops of the kinase domain, whereas non-cancer-related hotspots tend to be located outside of these regions. Additionally, we identified a hotspot at residue R371 of the AURKA structure that has the highest number of exclusively non-cancer-related pathogenic mutations (21) and has not been previously discussed.


Asunto(s)
Proteínas Quinasas , Proteínas Serina-Treonina Quinasas , Humanos , Proteínas Quinasas/química , Proteínas Serina-Treonina Quinasas/química , Aurora Quinasa A/genética , Aurora Quinasa A/química , Aurora Quinasa A/metabolismo , Modelos Moleculares , Fosforilación , Mutación
4.
Drug Res (Stuttg) ; 73(1): 30-39, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36138546

RESUMEN

Aurora kinase is a group of enzymes that belongs to a serine-threonine family and plays a critical role in cellular division. Aurora Kinase A is overexpressed and distributed beyond the nucleus and is involved in tumorigenesis. Flavones are a class of flavonoids that are present in plants that show anticancer activity. Similar compounds of 2'Fluoroflavones are retrieved from the PubChem database. Then drug-like filters viz. REOS and PAINS were applied to remove toxic compounds using Canvas software, resulting in 3882 compounds being subjected to Glide docking with Aurora kinase A. The lead compounds were selected on the merit of hydrogen bonding, salt bridge, as well as pi-pi interactions, 4-(6-Fluoro-4-oxychromen-2yl) benzoic acid, has been found one of the best molecules from docking studies. The binding mode of the lead compound with AURKA reveals that the amino acid residues viz, Lys162, Ala213, and His280 are more important for binding with the binding affinity of -11.760 kcal/mol. The molecular dynamics simulations of 100 ns were done, which shows the mean RMSD value of 1.77 Å for all 3 complexes of the protein and Fluoroflavone and its analogs. This shows that Fluoroflavone and its 2 best analogs are tightly attached to the active sites and thus have conformational stability. Our finding suggests that 4-(6-fluoro-4-oxochromen-2-yl)benzoic acid and 4-(4-Oxochromen-2-yl)benzoate can be further used in vitro and in vivo experiments and can probably serve as a novel drug for cancer treatment.


Asunto(s)
Aurora Quinasa A , Simulación de Dinámica Molecular , Aurora Quinasa A/química , Dominio Catalítico , Bases de Datos de Compuestos Químicos , Ácido Benzoico
5.
Int J Mol Sci ; 23(3)2022 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-35163790

RESUMEN

Aurora A kinase (Aurora A) is a serine/threonine kinase regulating control of multiple events during cell-cycle progression. Playing roles in promoting proliferation and inhibiting cell death in cancer cells leads Aurora A to become a target for cancer therapy. It is overexpressed and associated with a poor prognosis in ovarian cancer. Improving cisplatin therapy outcomes remains an important issue for advanced-stage ovarian cancer treatment, and Aurora A inhibitors may improve it. In the present study, we identified natural compounds with higher docking scores than the known Aurora A ligand through structure-based virtual screening, including the natural compound fangchinoline, which has been associated with anticancer activities but not yet investigated in ovarian cancer. The binding and inhibition of Aurora A by fangchinoline were verified using cellular thermal shift and enzyme activity assays. Fangchinoline reduced viability and proliferation in ovarian cancer cell lines. Combination fangchinoline and cisplatin treatment enhanced cisplatin-DNA adduct levels, and the combination index revealed synergistic effects on cell viability. An in vivo study showed that fangchinoline significantly enhanced cisplatin therapeutic effects in OVCAR-3 ovarian cancer-bearing mice. Fangchinoline may inhibit tumor growth and enhance cisplatin therapy in ovarian cancer. This study reveals a novel Aurora A inhibitor, fangchinoline, as a potentially viable adjuvant for ovarian cancer therapy.


Asunto(s)
Aurora Quinasa A/metabolismo , Bencilisoquinolinas/administración & dosificación , Cisplatino/administración & dosificación , Aductos de ADN/efectos de los fármacos , Neoplasias Ováricas/tratamiento farmacológico , Animales , Aurora Quinasa A/química , Bencilisoquinolinas/química , Bencilisoquinolinas/farmacología , Línea Celular Tumoral , Cisplatino/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Sinergismo Farmacológico , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Modelos Moleculares , Simulación del Acoplamiento Molecular , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Conformación Proteica , Ensayos Antitumor por Modelo de Xenoinjerto
6.
J Am Soc Mass Spectrom ; 33(3): 420-435, 2022 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-35099954

RESUMEN

Protein kinase inhibitors are highly effective in treating diseases driven by aberrant kinase signaling and as chemical tools to help dissect the cellular roles of kinase signaling complexes. Evaluating the effects of binding of small molecule inhibitors on kinase conformational dynamics can assist in understanding both inhibition and resistance mechanisms. Using gas-phase ion-mobility mass spectrometry (IM-MS), we characterize changes in the conformational landscape and stability of the protein kinase Aurora A (Aur A) driven by binding of the physiological activator TPX2 or small molecule inhibition. Aided by molecular modeling, we establish three major conformations, the relative abundances of which were dependent on the Aur A activation status: one highly populated compact conformer similar to that observed in most crystal structures, a second highly populated conformer possessing a more open structure infrequently found in crystal structures, and an additional low-abundance conformer not currently represented in the protein databank. Notably, inhibitor binding induces more compact configurations of Aur A, as adopted by the unbound enzyme, with both IM-MS and modeling revealing inhibitor-mediated stabilization of active Aur A.


Asunto(s)
Aurora Quinasa A , Espectrometría de Movilidad Iónica/métodos , Modelos Moleculares , Aurora Quinasa A/análisis , Aurora Quinasa A/química , Humanos , Espectrometría de Masas/métodos , Conformación Proteica , Estabilidad Proteica
7.
Int J Mol Sci ; 22(23)2021 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-34884931

RESUMEN

Neuroblastoma is a severe childhood disease, accounting for ~10% of all infant cancers. The amplification of the MYCN gene, coding for the N-Myc transcription factor, is an essential marker correlated with tumor progression and poor prognosis. In neuroblastoma cells, the mitotic kinase Aurora-A (AURKA), also frequently overexpressed in cancer, prevents N-Myc degradation by directly binding to a highly conserved N-Myc region. As a result, elevated levels of N-Myc are observed. During recent years, it has been demonstrated that some ATP competitive inhibitors of AURKA also cause essential conformational changes in the structure of the activation loop of the kinase that prevents N-Myc binding, thus impairing the formation of the AURKA/N-Myc complex. In this study, starting from a screening of crystal structures of AURKA in complexes with known inhibitors, we identified additional compounds affecting the conformation of the kinase activation loop. We assessed the ability of such compounds to disrupt the interaction between AURKA and N-Myc in vitro, using Surface Plasmon Resonance competition assays, and in tumor cell lines overexpressing MYCN, by performing Proximity Ligation Assays. Finally, their effects on N-Myc cellular levels and cell viability were investigated. Our results identify PHA-680626 as an amphosteric inhibitor both in vitro and in MYCN overexpressing cell lines, thus expanding the repertoire of known conformational disrupting inhibitors of the AURKA/N-Myc complex and confirming that altering the conformation of the activation loop of AURKA with a small molecule is an effective strategy to destabilize the AURKA/N-Myc interaction in neuroblastoma cancer cells.


Asunto(s)
Aurora Quinasa A/metabolismo , Proteína Proto-Oncogénica N-Myc/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/farmacología , Pirroles/farmacología , Adenosina Trifosfato/metabolismo , Antineoplásicos/farmacología , Aurora Quinasa A/antagonistas & inhibidores , Aurora Quinasa A/química , Azepinas/metabolismo , Azepinas/farmacología , Benzazepinas/metabolismo , Benzazepinas/farmacología , Sitios de Unión , Unión Competitiva , Línea Celular , Evaluación Preclínica de Medicamentos/métodos , Humanos , Proteína Proto-Oncogénica N-Myc/química , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/metabolismo , Conformación Proteica , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/metabolismo , Pirazoles/metabolismo , Pirimidinas/metabolismo , Pirimidinas/farmacología , Pirroles/metabolismo , Resonancia por Plasmón de Superficie
8.
Sci Rep ; 11(1): 23681, 2021 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-34880385

RESUMEN

Lung adenocarcinoma (LUAD) belongs to a subgroup of non-small cell lung cancer (NSCLC) with an increasing incidence all over the world. Tanshinone IIA (TSA), an active compound of Salvia miltiorrhiza Bunge., has been found to have anti-tumor effects on many tumors, but its anti-LUAD effect and its mechanism have not been reported yet. In this study, bio-information analysis was applied to characterize the potential mechanism of TSA on LUA, biological experiments were used to verify the mechanisms involved. TCGA, Pubchem, SwissTargetPrediction, Venny2.1.0, STRING, DAVID, Cytoscape 3.7.2, Omicshare, GEPIA, RSCBPDB, Chem Draw, AutoDockTools, and PyMOL were utilized for analysis in the bio-information analysis and network pharmacology. Our experiments in vitro focused on the anti-LUAD effects and mechanisms of TSA on LUAD cells (A549 and NCI-H1975 cells) via MTT, plate cloning, Annexin V-FITC and PI dual staining, flow cytometry, and western blot assays. A total of 64 differentially expressed genes (DEGs) of TSA for treatment of LUAD were screened out. Gene ontology and pathway analysis revealed characteristic of the DEGs network. After GEPIA-based DEGs confirmation, 46 genes were considered having significant differences. Further, 10 key DEGs (BTK, HSD11B1, ADAM33, TNNC1, THRA, CCNA2, AURKA, MIF, PLK1, and SORD) were identified as the most likely relevant genes from overall survival analysis. Molecular Docking results showed that CCNA2, CDK2 and PLK1 had the lowest docking energy. MTT and plate cloning assays results showed that TSA inhibited the proliferation of LUAD cells in a concentration-dependent manner. Annexin V-FITC and PI dual staining and flow cytometry assays results told that TSA promoted the apoptosis of the two LUAD cells in different degrees, and induced cycle arrest in the G1/S phase. Western blot results showed that TSA significantly down-regulated the expression of CCNA2, CDK2, AURKA, PLK1, and p-ERK. In summary, TSA could suppress the progression of LUAD by inducing cell apoptosis and arresting cell cycle, and these were done by regulating CCNA2-CDK2 complex and AURKA/PLK1 pathway. These findings are the first to demonstrate the molecular mechanism of TSA in treatment of LUAD combination of network bio-information analysis and biological experiments in vitro.


Asunto(s)
Abietanos/farmacología , Adenocarcinoma del Pulmón/metabolismo , Antineoplásicos Fitogénicos/farmacología , Aurora Quinasa A/metabolismo , Proteínas de Ciclo Celular/metabolismo , Ciclina A2/metabolismo , Quinasa 2 Dependiente de la Ciclina/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Abietanos/química , Adenocarcinoma del Pulmón/tratamiento farmacológico , Adenocarcinoma del Pulmón/etiología , Adenocarcinoma del Pulmón/patología , Antineoplásicos Fitogénicos/química , Apoptosis/efectos de los fármacos , Aurora Quinasa A/química , Biomarcadores de Tumor , Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/química , Línea Celular Tumoral , Biología Computacional/métodos , Ciclina A2/química , Quinasa 2 Dependiente de la Ciclina/química , Susceptibilidad a Enfermedades , Perfilación de la Expresión Génica , Humanos , Modelos Moleculares , Mapeo de Interacción de Proteínas , Mapas de Interacción de Proteínas , Proteínas Serina-Treonina Quinasas/química , Proteínas Proto-Oncogénicas/química , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad , Transcriptoma , Quinasa Tipo Polo 1
9.
Biomolecules ; 11(4)2021 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-33921540

RESUMEN

The conformational state of the activation loop (A-loop) is pivotal for the activity of most protein kinases. Hence, the characterization of the conformational dynamics of the A-loop is important to increase our understanding of the molecular processes related to diseases and to support the discovery of small molecule kinase inhibitors. Here, we carry out a combination of molecular dynamics (MD) and essential dynamics (ED) analyses to fully map the effects of phosphorylation, ADP, and conformation disrupting (CD) inhibitors (i.e., CD532 and MLN8054) on the dynamics of the A-loop of Aurora-A. MD revealed that the stability of the A-loop in an open conformation is enhanced by single phospho-Thr-288, while paradoxically, the presence of a second phosphorylation at Thr-287 decreases such stability and renders the A-loop more fluctuant in time and space. Moreover, we found that this post-translational modification has a significant effect on the direction of the A-loop motions. ED analysis suggests that the presence of the phosphate moiety induces the dynamics of Aurora-A to sample two distinct energy minima, instead of a single large minimum, as in unphosphorylated Aurora-A states. This observation indicates that the conformational distributions of Aurora-A with both single and double phospho-threonine modifications are remarkably different from the unphosphorylated state. In the closed states, binding of CD532 and MLN8054 inhibitors has the effect of increasing the distance of the N- and C-lobes of the kinase domain of Aurora-A, and the angle analysis between those two lobes during MD simulations showed that the N- and C-lobes are kept more open in presence of CD532, compared to MLN8054. As the A-loop is a common feature of Aurora protein kinases, our studies provide a general description of the conformational dynamics of this structure upon phosphorylation and different ligands binding.


Asunto(s)
Adenosina Difosfato/metabolismo , Aurora Quinasa A/química , Simulación de Dinámica Molecular , Inhibidores de Proteínas Quinasas/farmacología , Adenosina Difosfato/química , Aurora Quinasa A/antagonistas & inhibidores , Aurora Quinasa A/metabolismo , Benzazepinas/química , Benzazepinas/farmacología , Dominio Catalítico , Humanos , Compuestos de Fenilurea/química , Compuestos de Fenilurea/farmacología , Fosforilación , Unión Proteica , Inhibidores de Proteínas Quinasas/química , Pirimidinas/química , Pirimidinas/farmacología
10.
Gen Comp Endocrinol ; 300: 113617, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-32950578

RESUMEN

The mitosis-associated protein aurora kinase A (AURKA) regulates the maturation of germ cells. We have previously reported using transcriptome analysis that AURKA is expressed in yak testes. Although Tibetan sheep possess an immense economic value, their reproductive rate is low. Herein, the expression and functions of AURKA in the hypothalamus-pituitary-testicular (HPT) axis in Tibetan sheep from Tianzhu were investigated. The cDNA sequence of sheep AURKA was cloned and bioinformatics techniques were used to predict its structure. Tissue expression of AURKA was determined by qPCR, immunoblotting, immunostaining, and immunohistochemistry. The AURKA coding sequence was found to be 1218 bp in length, encoding a 405-amino acid polypeptide chain. Furthermore, the highest sequence similarity of AURKA with the corresponding sequence in other species was seen in goat and cattle; the least degree of similarity was seen in the domestic cat. In addition, AURKA expression was elevated in the testes compared to that in the hypothalamus and pituitary (p < 0.01). Moreover, AURKA was mainly localized in the hypothalamic paraventricular nucleus (magnocellular), chromophobe cells of the pituitary, and spermatogenic cells of the testis. These results indicated that AURKA might participate in sheep reproductive regulation, thus providing a reference for the study of AURKA function in the reproductive process of Tibetan sheep from Tianzhu.


Asunto(s)
Aurora Quinasa A/metabolismo , Hipotálamo/enzimología , Hipófisis/enzimología , Ovinos/metabolismo , Testículo/enzimología , Secuencia de Aminoácidos , Animales , Aurora Quinasa A/química , Aurora Quinasa A/genética , Perfilación de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Masculino , Filogenia , Tibet
11.
Sci Signal ; 13(639)2020 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-32636306

RESUMEN

Reactive oxygen species (ROS) are physiological mediators of cellular signaling and play potentially damaging roles in human diseases. In this study, we found that the catalytic activity of the Ser/Thr kinase Aurora A was inhibited by the oxidation of a conserved cysteine residue (Cys290) that lies adjacent to Thr288, a critical phosphorylation site in the activation segment. Cys is present at the equivalent position in ~100 human Ser/Thr kinases, a residue that we found was important not only for the activity of human Aurora A but also for that of fission yeast MAPK-activated kinase (Srk1) and PKA (Pka1). Moreover, the presence of this conserved Cys predicted biochemical redox sensitivity among a cohort of human CAMK, AGC, and AGC-like kinases. Thus, we predict that redox modulation of the conserved Cys290 of Aurora A may be an underappreciated regulatory mechanism that is widespread in eukaryotic Ser/Thr kinases. Given the key biological roles of these enzymes, these findings have implications for understanding physiological and pathological responses to ROS and highlight the importance of protein kinase regulation through multivalent modification of the activation segment.


Asunto(s)
Aurora Quinasa A/química , Aurora Quinasa A/metabolismo , Evolución Molecular , Animales , Aurora Quinasa A/genética , Cisteína/química , Cisteína/genética , Cisteína/metabolismo , Células HeLa , Humanos , Ratones , Oxidación-Reducción
12.
Sci Signal ; 13(641)2020 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-32694171

RESUMEN

Cell cycle-dependent redox changes can mediate transient covalent modifications of cysteine thiols to modulate the activities of regulatory kinases and phosphatases. Our previously reported finding that protein cysteine oxidation is increased during mitosis relative to other cell cycle phases suggests that redox modifications could play prominent roles in regulating mitotic processes. The Aurora family of kinases and their downstream targets are key components of the cellular machinery that ensures the proper execution of mitosis and the accurate segregation of chromosomes to daughter cells. In this study, x-ray crystal structures of the Aurora A kinase domain delineate redox-sensitive cysteine residues that, upon covalent modification, can allosterically regulate kinase activity and oligomerization state. We showed in both Xenopus laevis egg extracts and mammalian cells that a conserved cysteine residue within the Aurora A activation loop is crucial for Aurora A activation by autophosphorylation. We further showed that covalent disulfide adducts of this residue promote autophosphorylation of the Aurora A kinase domain. These findings reveal a potential mechanistic link between Aurora A activation and changes in the intracellular redox state during mitosis and provide insights into how novel small-molecule inhibitors may be developed to target specific subpopulations of Aurora A.


Asunto(s)
Aurora Quinasa A/química , Aurora Quinasa A/metabolismo , Mitosis , Animales , Aurora Quinasa A/genética , Cristalografía por Rayos X , Activación Enzimática/genética , Células HEK293 , Humanos , Oxidación-Reducción , Xenopus laevis
13.
Science ; 367(6480): 912-917, 2020 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-32079772

RESUMEN

A myriad of cellular events are regulated by allostery; therefore, evolution of this process is of fundamental interest. Here, we use ancestral sequence reconstruction to resurrect ancestors of two colocalizing proteins, Aurora A kinase and its allosteric activator TPX2 (targeting protein for Xklp2), to experimentally characterize the evolutionary path of allosteric activation. Autophosphorylation of the activation loop is the most ancient activation mechanism; it is fully developed in the oldest kinase ancestor and has remained stable over 1 billion years of evolution. As the microtubule-associated protein TPX2 appeared, efficient kinase binding to TPX2 evolved, likely owing to increased fitness by virtue of colocalization. Subsequently, TPX2-mediated allosteric kinase regulation gradually evolved. Surprisingly, evolution of this regulation is encoded in the kinase and did not arise by a dominating mechanism of coevolution.


Asunto(s)
Aurora Quinasa A/clasificación , Aurora Quinasa A/metabolismo , Evolución Molecular , Regulación Alostérica , Animales , Aurora Quinasa A/química , Proteínas de Ciclo Celular/metabolismo , Activación Enzimática , Humanos , Proteínas Asociadas a Microtúbulos/metabolismo , Filogenia
14.
Biochem J ; 477(2): 431-444, 2020 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-31904830

RESUMEN

Protein Ser/Thr phosphatase-6 (PP6) regulates pathways for activation of NF-kB, YAP1 and Aurora A kinase (AURKA). PP6 is a heterotrimer comprised of a catalytic subunit, one of three different SAPS subunits and one of three different ankyrin-repeat ANKRD subunits. Here, we show FLAG-PP6C expressed in cells preferentially binds endogenous SAPS3, and the complex is active with the chemical substrate DiFMUP. SAPS3 has multiple acidic sequence motifs recognized by protein kinase CK2 (CK2) and SAPS3 is phosphorylated by purified CK2, without affecting its associated PP6 phosphatase activity. However, HA3-SAPS3-PP6 phosphatase activity using pT288 AURKA as substrate is significantly increased by phosphorylation with CK2. The substitution of Ala in nine putative phosphorylation sites in SAPS3 was required to prevent CK2 activation of the phosphatase. Different CK2 chemical inhibitors equally increased phosphorylation of endogenous AURKA in living cells, consistent with reduction in PP6 activity. CRISPR/Cas9 deletion or siRNA knockdown of SAPS3 resulted in highly activated endogenous AURKA, and a high proportion of cells with abnormal nuclei. Activation of PP6 by CK2 can form a feedback loop with bistable changes in substrates.


Asunto(s)
Aurora Quinasa A/genética , Quinasa de la Caseína II/química , Fosfoproteínas Fosfatasas/genética , Alanina/genética , Sustitución de Aminoácidos/genética , Aurora Quinasa A/química , Sistemas CRISPR-Cas/genética , Quinasa de la Caseína II/genética , Dominio Catalítico/genética , Inhibidores Enzimáticos/farmacología , Células HeLa , Humanos , Fosfoproteínas Fosfatasas/antagonistas & inhibidores , Fosfoproteínas Fosfatasas/química , Fosforilación/genética , Unión Proteica/efectos de los fármacos , ARN Interferente Pequeño/genética , Especificidad por Sustrato/efectos de los fármacos
15.
Redox Biol ; 28: 101318, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31546169

RESUMEN

Aurora A kinase is a master mitotic regulator whose functions are controlled by several regulatory interactions and post-translational modifications. It is frequently dysregulated in cancer, making Aurora A inhibition a very attractive antitumor target. However, recently uncovered links between Aurora A, cellular metabolism and redox regulation are not well understood. In this study, we report a novel mechanism of Aurora A regulation in the cellular response to oxidative stress through CoAlation. A combination of biochemical, biophysical, crystallographic and cell biology approaches revealed a new and, to our knowledge, unique mode of Aurora A inhibition by CoA, involving selective binding of the ADP moiety of CoA to the ATP binding pocket and covalent modification of Cys290 in the activation loop by the thiol group of the pantetheine tail. We provide evidence that covalent CoA modification (CoAlation) of Aurora A is specific, and that it can be induced by oxidative stress in human cells. Oxidising agents, such as diamide, hydrogen peroxide and menadione were found to induce Thr 288 phosphorylation and DTT-dependent dimerization of Aurora A. Moreover, microinjection of CoA into fertilized mouse embryos disrupts bipolar spindle formation and the alignment of chromosomes, consistent with Aurora A inhibition. Altogether, our data reveal CoA as a new, rather selective, inhibitor of Aurora A, which locks this kinase in an inactive state via a "dual anchor" mechanism of inhibition that might also operate in cellular response to oxidative stress. Finally and most importantly, we believe that these novel findings provide a new rationale for developing effective and irreversible inhibitors of Aurora A, and perhaps other protein kinases containing appropriately conserved Cys residues.


Asunto(s)
Aurora Quinasa A/química , Aurora Quinasa A/metabolismo , Coenzima A/administración & dosificación , Animales , Coenzima A/química , Coenzima A/farmacología , Cristalografía por Rayos X , Células HEK293 , Células Hep G2 , Humanos , Ratones , Modelos Moleculares , Estrés Oxidativo , Fosforilación , Conformación Proteica , Huso Acromático/efectos de los fármacos , Huso Acromático/metabolismo
16.
Elife ; 82019 12 24.
Artículo en Inglés | MEDLINE | ID: mdl-31872801

RESUMEN

Deciphering how signaling enzymes operate within discrete microenvironments is fundamental to understanding biological processes. A-kinase anchoring proteins (AKAPs) restrict the range of action of protein kinases within intracellular compartments. We exploited the AKAP targeting concept to create genetically encoded platforms that restrain kinase inhibitor drugs at distinct subcellular locations. Local Kinase Inhibition (LoKI) allows us to ascribe organelle-specific functions to broad specificity kinases. Using chemical genetics, super resolution microscopy, and live-cell imaging we discover that centrosomal delivery of Polo-like kinase 1 (Plk1) and Aurora A (AurA) inhibitors attenuates kinase activity, produces spindle defects, and prolongs mitosis. Targeted inhibition of Plk1 in zebrafish embryos illustrates how centrosomal Plk1 underlies mitotic spindle assembly. Inhibition of kinetochore-associated pools of AurA blocks phosphorylation of microtubule-kinetochore components. This versatile precision pharmacology tool enhances investigation of local kinase biology.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/genética , Aurora Quinasa A/genética , Proteínas de Ciclo Celular/genética , Mitosis/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Animales , Aurora Quinasa A/química , Proteínas de Ciclo Celular/química , Centrosoma/química , Centrosoma/ultraestructura , Desarrollo Embrionario/genética , Regulación del Desarrollo de la Expresión Génica , Humanos , Cinetocoros/química , Microtúbulos/genética , Fosforilación/genética , Inhibidores de Proteínas Quinasas/química , Proteínas Serina-Treonina Quinasas/química , Proteínas Proto-Oncogénicas/química , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo , Quinasa Tipo Polo 1
17.
Anal Chem ; 91(20): 13222-13229, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31525957

RESUMEN

Small-molecule inhibitors of protein kinases attract widespread interest in the field of disease therapy because of their high specificity and ease of administration. However, dissecting the conformational inhibition dynamics of kinase inhibitors is still challenging. Here, simultaneously monitoring the conformational inhibition details and potency of Aurora A kinase inhibitors has been achieved by active isotope dimethyl labeling coupled with mass spectrometry-based quantitative lysine reactivity profiling. The conformational effects of inhibitors on lysine reactivity can be globally quantified to feasibly reveal the regions involved in the kinase dynamic inhibition. The half-maximum disturbance concentrations (DC50 values) of the conformation-specific lysine residues could directly represent the conformational selectivity and potency of kinase inhibitors. Further, K309 is discovered as a novel hotspot contributing to the inhibition of Aurora A kinase via the specific rotation of kinase activation loop. This quantitative lysine reactivity profiling strategy might greatly promote the development of targeted drugs.


Asunto(s)
Aurora Quinasa A/metabolismo , Lisina/química , Inhibidores de Proteínas Quinasas/metabolismo , Aurora Quinasa A/química , Aurora Quinasa A/genética , Humanos , Simulación de Dinámica Molecular , Mutación , Unión Proteica , Conformación Proteica , Inhibidores de Proteínas Quinasas/química
18.
Cell Cycle ; 18(18): 2228-2238, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31357906

RESUMEN

Aurora-A is a serine/threonine kinase, which is overexpressed in multiple human cancers and plays a key role in tumorigenesis and tumor development. In this study, we found that the receptor of activated C-kinase1 (RACK1), an important regulator of biological functions, interacted with Aurora-A and co-localized with Aurora-A at centrosomes. Moreover, RACK1 induces the auto-phosphorylation of Aurora-A in vitro and in vivo. Depletion of RACK1 impaired the activation of Aurora-A in late G2 phase, then inhibited the mitotic entry and leaded to multi-polarity, severe chromosome alignment defects, or centrosome amplification. Taken together, these results suggest that RACK1 is a new partner of Aurora-A and play a critical role in the regulation of the Aurora-A activity during mitosis, which may provide a basis for future anticancer studies targeting Aurora-A.


Asunto(s)
Aurora Quinasa A/metabolismo , Puntos de Control de la Fase G2 del Ciclo Celular/genética , Puntos de Control de la Fase M del Ciclo Celular/genética , Proteínas de Neoplasias/metabolismo , Receptores de Cinasa C Activada/metabolismo , Aurora Quinasa A/química , Aurora Quinasa A/genética , Dominio Catalítico , Centrosoma/metabolismo , Células HeLa , Humanos , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Fosforilación/genética , Unión Proteica , Estabilidad Proteica , Receptores de Cinasa C Activada/química , Receptores de Cinasa C Activada/genética , Transducción de Señal/genética , Huso Acromático/metabolismo , Transfección
19.
Proc Natl Acad Sci U S A ; 116(28): 13937-13942, 2019 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-31239342

RESUMEN

Despite being the subject of intense effort and scrutiny, kinases have proven to be consistently challenging targets in inhibitor drug design. A key obstacle has been promiscuity and consequent adverse effects of drugs targeting the ATP binding site. Here we introduce an approach to controlling kinase activity by using monobodies that bind to the highly specific regulatory allosteric pocket of the oncoprotein Aurora A (AurA) kinase, thereby offering the potential for more specific kinase modulators. Strikingly, we identify a series of highly specific monobodies acting either as strong kinase inhibitors or activators via differential recognition of structural motifs in the allosteric pocket. X-ray crystal structures comparing AurA bound to activating vs inhibiting monobodies reveal the atomistic mechanism underlying allosteric modulation. The results reveal 3 major advantages of targeting allosteric vs orthosteric sites: extreme selectivity, ability to inhibit as well as activate, and avoidance of competing with ATP that is present at high concentrations in the cells. We envision that exploiting allosteric networks for inhibition or activation will provide a general, powerful pathway toward rational drug design.


Asunto(s)
Aurora Quinasa A/química , Aurora Quinasa B/química , Inhibidores de Proteínas Quinasas/química , Proteínas Quinasas/química , Adenosina Trifosfato/química , Adenosina Trifosfato/metabolismo , Regulación Alostérica/genética , Aurora Quinasa A/antagonistas & inhibidores , Aurora Quinasa A/genética , Aurora Quinasa B/antagonistas & inhibidores , Aurora Quinasa B/genética , Sitios de Unión/genética , Proteínas Portadoras/química , Proteínas Portadoras/genética , Cristalografía por Rayos X , Diseño de Fármacos , Dominio de Fibronectina del Tipo III/genética , Humanos , Conformación Proteica , Proteínas Quinasas/genética
20.
Sci Rep ; 9(1): 5550, 2019 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-30944388

RESUMEN

The tumour suppressor protein RASSF1A is phosphorylated by Aurora A kinase, thereby impairing its tumour suppressor function. Consequently, inhibiting the interaction between Aurora A and RASSF1A may be used for anti-tumour therapy. We used recombinant variants of RASSF1A to map the sites of interaction with Aurora A. The phosphorylation kinetics of three truncated RASSF1A variants has been analysed. Compared to the RASSF1A form lacking the 120 residue long N-terminal part, the Km value of the phosphorylation is increased from 10 to 45 µM upon additional deletion of the C-terminal SARAH domain. On the other hand, deletion of the flexible loop (Δ177-197) that precedes the phosphorylation site/s (T202/S203) results in a reduction of the kcat value from about 40 to 7 min-1. Direct physical interaction between the isolated SARAH domain and Aurora A was revealed by SPR. These data demonstrate that the SARAH domain of RASSF1A is involved in the binding to Aurora A kinase. Structural modelling confirms that a novel complex is feasible between the SARAH domain and the kinase domain of Aurora A. In addition, a regulatory role of the loop in the catalytic phosphorylation reaction has been demonstrated both experimentally and by structural modelling.


Asunto(s)
Aurora Quinasa A/metabolismo , Dominios y Motivos de Interacción de Proteínas , Receptores Opioides kappa/metabolismo , Aurora Quinasa A/química , Aurora Quinasa A/genética , Sitios de Unión , Cromatografía en Gel , Modelos Moleculares , Mutación , Fosforilación , Multimerización de Proteína , Receptores Opioides kappa/química , Receptores Opioides kappa/genética , Resonancia por Plasmón de Superficie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...