Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Oncol ; 12: 965455, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36313676

RESUMO

The mitotic kinesin Eg5 has emerged as a potential anti-mitotic target for the purposes of cancer chemotherapy. Whether clinical resistance to these inhibitors can arise is unclear. We exploited HCT116 cancer cell line to select resistant clones to S-trityl-L-cysteine (STLC), an extensively studied Eg5 loop-L5 binding inhibitor. The STLC resistant clones differed in their resistance to other loop-L5 binding inhibitors but remained sensitive to the ATP class of competitive Eg5 specific inhibitors. Eg5 is still necessary for bipolar spindle formation in the resistant clones since the cells were sensitive to RNAi mediated depletion of Eg5. One clone expressing Eg5(T107N), a dominant point mutation in the P-loop of the ATP binding domain of the motor, appeared to be not only resistant but also dependent on the presence of STLC. Eg5(T107N) expression was associated also with resistance to the clinical relevant loop-L5 Eg5 inhibitors, Arry-520 and ispinesib. Ectopic expression of the Eg5(T107N) mutant in the absence of STLC was associated with strong non-exchangeable binding to microtubules causing them to bundle. Biochemical assays showed that in contrast to the wild type Eg5-STLC complex, the ATP binding site of the Eg5(T107N) is accessible for nucleotide exchange only when the inhibitor is present. We predict that resistance can be overcome by inhibitors that bind to other than the Eg5 loop-L5 binding site having different chemical scaffolds, and that allostery-dependent resistance to Eg5 inhibitors may also occur in cells and may have positive implications in chemotherapy since once diagnosed may be beneficial following cessation of the chemotherapeutic regimen.

2.
Biochem Pharmacol ; 184: 114364, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33310050

RESUMO

Eg5, the product of Kif11 gene, also known as kinesin spindle protein, is a motor protein involved in the proper establishment of a bipolar mitotic spindle. Eg5 is one of the 45 different kinesins coded in the human genome of the kinesin motor protein superfamily. Over the last three decades Eg5 has attracted great interest as a promising new mitotic target. The identification of monastrol as specific inhibitor of the ATPase activity of the motor domain of Eg5 inhibiting the Eg5 microtubule motility in vitro and in cellulo sparked an intense interest in academia and industry to pursue the identification of novel small molecules that target Eg5 in order to be used in cancer chemotherapy based on the anti-mitotic strategy. Several Eg5 inhibitors entered clinical trials. Currently the field is faced with the problem that most of the inhibitors tested exhibited only limited efficacy. However, one Eg5 inhibitor, Arry-520 (clinical name filanesib), has demonstrated clinical efficacy in patients with multiple myeloma and is scheduled to enter phase III clinical trials. At the same time, new trends in Eg5 inhibitor research are emerging, including an increased interest in novel inhibitor binding sites and a focus on drug synergy with established antitumor agents to improve chemotherapeutic efficacy. This review presents an updated view of the structure and function of Eg5-inhibitor complexes, traces the possible development of resistance to Eg5 inhibitors and their potential therapeutic applications, and surveys the current challenges and future directions of this active field in drug discovery.


Assuntos
Antimitóticos/farmacologia , Antineoplásicos/farmacologia , Cinesinas/antagonistas & inibidores , Cinesinas/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Antimitóticos/química , Antimitóticos/farmacocinética , Antineoplásicos/farmacocinética , Sítios de Ligação , Produtos Biológicos/química , Produtos Biológicos/farmacologia , Ensaios Clínicos como Assunto , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/fisiologia , Humanos , Cinesinas/química , Terapia de Alvo Molecular/métodos
3.
Mol Cancer Ther ; 18(12): 2394-2406, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31488701

RESUMO

Arry-520 is an advanced drug candidate from the Eg5 inhibitor class undergoing clinical evaluation in patients with relapsed or refractory multiple myeloma. Here, we show by structural analysis that Arry-520 binds stoichiometrically to the motor domain of Eg5 in the conventional allosteric loop L5 pocket in a complex that suggests the same structural mechanism as other Eg5 inhibitors. We have previously shown that acquired resistance through mutations in the allosteric-binding site located at loop L5 in the Eg5 structure appears to be independent of the inhibitors' scaffold, which suggests that Arry-520 will ultimately have the same fate. When Arry-520 was assessed in two cell lines selected for the expression of either Eg5(D130A) or Eg5(L214A) STLC-resistant alleles, mutations previously shown to convey resistance to this class of inhibitors, it was inactive in both. Surprisingly, when the cells were challenged with ispinesib, another Eg5 inhibitor, the Eg5(D130A) cells were resistant, but those expressing Eg5(L214A) were strikingly sensitive. Molecular dynamics simulations suggest that subtle differences in ligand binding and flexibility in both compound and protein may alter allosteric transmission from the loop L5 site that do not necessarily result in reduced inhibitory activity in mutated Eg5 structures. Although we predict that cells challenged with Arry-520 in the clinical setting are likely to acquire resistance through point mutations in the Eg5-binding site, the data for ispinesib suggest that this resistance mechanism is not scaffold independent as previously thought, and new inhibitors can be designed that retain inhibitory activity in these resistant cells.


Assuntos
Antimitóticos/uso terapêutico , Tiadiazóis/uso terapêutico , Antimitóticos/farmacologia , Técnicas de Cultura de Células , Humanos , Modelos Moleculares , Tiadiazóis/farmacologia
4.
Mol Cell ; 72(5): 902-915.e7, 2018 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-30392928

RESUMO

Chromatin adopts a diversity of regular and irregular fiber structures in vitro and in vivo. However, how an array of nucleosomes folds into and switches between different fiber conformations is poorly understood. We report the 9.7 Å resolution crystal structure of a 6-nucleosome array bound to linker histone H1 determined under ionic conditions that favor incomplete chromatin condensation. The structure reveals a flat two-start helix with uniform nucleosomal stacking interfaces and a nucleosome packing density that is only half that of a twisted 30-nm fiber. Hydroxyl radical footprinting indicates that H1 binds the array in an on-dyad configuration resembling that observed for mononucleosomes. Biophysical, cryo-EM, and crosslinking data validate the crystal structure and reveal that a minor change in ionic environment shifts the conformational landscape to a more compact, twisted form. These findings provide insights into the structural plasticity of chromatin and suggest a possible assembly pathway for a 30-nm fiber.


Assuntos
DNA/química , Histonas/química , Proteína 1 de Modelagem do Nucleossomo/química , Nucleossomos/ultraestrutura , Animais , Sítios de Ligação , Clonagem Molecular , Microscopia Crioeletrônica , Cristalografia por Raios X , DNA/genética , DNA/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Radical Hidroxila/química , Modelos Moleculares , Proteína 1 de Modelagem do Nucleossomo/genética , Proteína 1 de Modelagem do Nucleossomo/metabolismo , Nucleossomos/química , Nucleossomos/metabolismo , Concentração Osmolar , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Xenopus laevis
6.
Mol Cell ; 66(3): 384-397.e8, 2017 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-28475873

RESUMO

Linker histones associate with nucleosomes to promote the formation of higher-order chromatin structure, but the underlying molecular details are unclear. We investigated the structure of a 197 bp nucleosome bearing symmetric 25 bp linker DNA arms in complex with vertebrate linker histone H1. We determined electron cryo-microscopy (cryo-EM) and crystal structures of unbound and H1-bound nucleosomes and validated these structures by site-directed protein cross-linking and hydroxyl radical footprinting experiments. Histone H1 shifts the conformational landscape of the nucleosome by drawing the two linkers together and reducing their flexibility. The H1 C-terminal domain (CTD) localizes primarily to a single linker, while the H1 globular domain contacts the nucleosome dyad and both linkers, associating more closely with the CTD-distal linker. These findings reveal that H1 imparts a strong degree of asymmetry to the nucleosome, which is likely to influence the assembly and architecture of higher-order structures.


Assuntos
Montagem e Desmontagem da Cromatina , Cromatina/metabolismo , DNA/metabolismo , Histonas/metabolismo , Nucleossomos/metabolismo , Animais , Pareamento de Bases , Sítios de Ligação , Cromatina/química , Cromatina/genética , Cromatina/ultraestrutura , Microscopia Crioeletrônica , DNA/química , DNA/genética , Histonas/química , Humanos , Modelos Moleculares , Nucleossomos/química , Nucleossomos/genética , Nucleossomos/ultraestrutura , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Relação Estrutura-Atividade , Fatores de Tempo , Xenopus laevis/genética , Xenopus laevis/metabolismo
7.
Sci Rep ; 5: 13242, 2015 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-26289831

RESUMO

TPPP/p25 is a microtubule-associated protein, detected in protein inclusions associated with various neurodegenerative diseases. Deletion analysis data show that TPPP/p25 has two microtubule binding sites, both located in intrinsically disordered domains, one at the N-terminal and the other in the C-terminal domain. In copolymerization assays the full-length protein exhibits microtubule stimulation and bundling activity. In contrast, at the same ratio relative to tubulin, truncated forms of TPPP/p25 exhibit either lower or no microtubule stimulation and no bundling activity, suggesting a cooperative phenomenon which is enhanced by the presence of the two binding sites. The binding characteristics of the N- and C-terminally truncated proteins to taxol-stabilized microtubules are similar to the full-length protein. However, the C-terminally truncated TPPP/p25 shows a lower Bmax for microtubule binding, suggesting that it may bind to a site of tubulin that is masked in microtubules. Bimolecular fluorescent complementation assays in cells expressing combinations of various TPPP/p25 fragments, but not that of the central folded domain, resulted in the generation of a fluorescence signal colocalized with perinuclear microtubule bundles insensitive to microtubule inhibitors. The data suggest that the central folded domain of TPPP/p25 following binding to microtubules can drive s homotypic protein-protein interactions leading to bundled microtubules.


Assuntos
Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Animais , Bovinos , Fluorescência , Humanos , Microtúbulos/ultraestrutura , Paclitaxel/farmacologia , Polimerização/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Deleção de Sequência
8.
Oncotarget ; 6(16): 14669-86, 2015 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-26036640

RESUMO

We previously identified 1-(2,4-dimethoxyphenyl)-3-(1-methylindolyl) propenone (IPP51), a new chalcone derivative that is capable of inducing prometaphase arrest and subsequent apoptosis of bladder cancer cells. Here, we demonstrate that IPP51 selectively inhibits proliferation of tumor-derived cells versus normal non-tumor cells. IPP51 interfered with spindle formation and mitotic chromosome alignment. Accumulation of cyclin B1 and mitotic checkpoint proteins Bub1 and BubR1 on chromosomes in IPP51 treated cells indicated the activation of spindle-assembly checkpoint, which is consistent with the mitotic arrest. The antimitotic actions of other chalcones are often associated with microtubule disruption. Indeed, IPP51 inhibited tubulin polymerization in an in vitro assay with purified tubulin. In cells, IPP51 induced an increase in soluble tubulin. Furthermore, IPP51 inhibited in vitro capillary-like tube formation by endothelial cells, indicating that it has anti-angiogenic activity. Molecular docking showed that the indol group of IPP51 can be accommodated in the colchicine binding site of tubulin. This characteristic was confirmed by an in vitro competition assay demonstrating that IPP51 can compete for colchicine binding to soluble tubulin. Finally, in a human bladder xenograft mouse model, IPP51 inhibited tumor growth without signs of toxicity. Altogether, these findings suggest that IPP51 is an attractive new microtubule-targeting agent with potential chemotherapeutic value.


Assuntos
Microtúbulos/genética , Neoplasias da Bexiga Urinária/genética , Animais , Proliferação de Células , Humanos , Camundongos , Microtúbulos/metabolismo , Simulação de Acoplamento Molecular , Estrutura Molecular , Neoplasias da Bexiga Urinária/tratamento farmacológico , Neoplasias da Bexiga Urinária/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Biochem Pharmacol ; 86(10): 1441-51, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-24041742

RESUMO

Determining the mechanism of action of drugs and their target specificity in cells remains a major challenge. Here we describe the use of cell lines expressing two point mutations in the allosteric inhibitor binding pocket of the mitotic kinesin Eg5 (D130A, in the loop L5 region and L214A in helix α3), which following transfection, were selected for their ability to proliferate normally in the presence of STLC, a well known Eg5 inhibitor. The cell lines were used to discriminate the mechanism of action of other chemically distinct small molecule inhibitors of Eg5 that differ in their mode of action. The STLC resistant cells were capable of continuous proliferation in the presence of ATP uncompetitive inhibitors, such as K858 and dimethylenastron, but were still sensitive to ATP competitive inhibitors that are thought to bind to a distinct site on Eg5 than the allosteric binding pocket. The STLC resistant cell lines can therefore be used as a filter to distinguish Eg5 loop L5 binding drugs from drugs binding to other pockets without prior structural information. Additionally, the cells can be used to analyze whether inhibitors of Eg5 are specific to this potential drug target or whether they have additional targets in dividing cells.


Assuntos
Antineoplásicos/farmacologia , Linhagem Celular Tumoral/efeitos dos fármacos , Cisteína/análogos & derivados , Resistencia a Medicamentos Antineoplásicos , Cinesinas/metabolismo , Adenosina Trifosfatases/metabolismo , Trifosfato de Adenosina/metabolismo , Sítio Alostérico , Sítios de Ligação , Linhagem Celular Tumoral/metabolismo , Proliferação de Células/efeitos dos fármacos , Cisteína/farmacologia , Humanos , Cinesinas/antagonistas & inibidores , Cinesinas/genética , Pontos de Checagem da Fase M do Ciclo Celular/efeitos dos fármacos , Mutação Puntual , Quinazolinas/farmacologia , Tiadiazóis/farmacologia , Tionas/farmacologia
10.
Proc Natl Acad Sci U S A ; 110(21): 8579-84, 2013 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-23657009

RESUMO

The role of the mitotic phosphorylation of the amino (NH2) terminus of Centromere Protein A (CENP-A), the histone variant epigenetic centromeric marker, remains elusive. Here, we show that the NH2 terminus of human CENP-A is essential for mitotic progression and that localization of CENP-C, another key centromeric protein, requires only phosphorylation of the CENP-A NH2 terminus, and is independent of the CENP-A NH2 terminus length and amino acid sequence. Mitotic CENP-A nucleosomal complexes contain CENP-C and phosphobinding 14-3-3 proteins. In contrast, mitotic nucleosomal complexes carrying nonphosphorylatable CENP-A-S7A contained only low levels of CENP-C and no detectable 14-3-3 proteins. Direct interactions between the phosphorylated form of CENP-A and 14-3-3 proteins as well as between 14-3-3 proteins and CENP-C were demonstrated. Taken together, our results reveal that 14-3-3 proteins could act as specific mitotic "bridges," linking phosphorylated CENP-A and CENP-C, which are necessary for the platform function of CENP-A centromeric chromatin in the assembly and maintenance of active kinetochores.


Assuntos
Autoantígenos/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Cinetocoros/metabolismo , Mitose/fisiologia , Nucleossomos/metabolismo , Proteínas 14-3-3/genética , Proteínas 14-3-3/metabolismo , Substituição de Aminoácidos , Autoantígenos/genética , Proteína Centromérica A , Proteínas Cromossômicas não Histona/genética , Células HeLa , Humanos , Mutação de Sentido Incorreto , Nucleossomos/genética , Fosforilação , Estrutura Terciária de Proteína
11.
PLoS Pathog ; 8(4): e1002637, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22496657

RESUMO

The type III secretion system (T3SS) is a complex macromolecular machinery employed by a number of Gram-negative pathogens to inject effectors directly into the cytoplasm of eukaryotic cells. ExoU from the opportunistic pathogen Pseudomonas aeruginosa is one of the most aggressive toxins injected by a T3SS, leading to rapid cell necrosis. Here we report the crystal structure of ExoU in complex with its chaperone, SpcU. ExoU folds into membrane-binding, bridging, and phospholipase domains. SpcU maintains the N-terminus of ExoU in an unfolded state, required for secretion. The phospholipase domain carries an embedded catalytic site whose position within ExoU does not permit direct interaction with the bilayer, which suggests that ExoU must undergo a conformational rearrangement in order to access lipids within the target membrane. The bridging domain connects catalytic domain and membrane-binding domains, the latter of which displays specificity to PI(4,5)P2. Both transfection experiments and infection of eukaryotic cells with ExoU-secreting bacteria show that ExoU ubiquitination results in its co-localization with endosomal markers. This could reflect an attempt of the infected cell to target ExoU for degradation in order to protect itself from its aggressive cytotoxic action.


Assuntos
Proteínas de Bactérias , Sistemas de Secreção Bacterianos , Toxinas Bacterianas , Dobramento de Proteína , Infecções por Pseudomonas/metabolismo , Pseudomonas aeruginosa , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/química , Toxinas Bacterianas/metabolismo , Células HeLa , Humanos , Chaperonas Moleculares/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Estrutura Terciária de Proteína , Pseudomonas aeruginosa/química , Pseudomonas aeruginosa/metabolismo , Relação Estrutura-Atividade , Ubiquitinação
12.
J Med Chem ; 55(6): 2561-73, 2012 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-22309208

RESUMO

Eg5/KSP is a promising mitotic spindle target for drug discovery in cancer chemotherapy and the development of agents against fungal diseases. A range of Eg5 targeting compounds identified by in vitro or cell-based screening is currently in development. We employed structure-based virtual screening of a database of 700, 000 compounds to identify three novel Eg5 inhibitors bearing quinazoline (24) or thioxoimidazolidine (30 and 37) scaffolds. The new compounds inhibit Eg5 ATPase activity, show growth inhibition in proliferation assays, and induce monoastral spindles in cells, the characteristic phenotype for Eg5 inhibiting agents. This is the first successful reported procedure for the identification of Eg5 inhibitors via receptor-ligand interaction-based virtual screening.


Assuntos
Antineoplásicos/síntese química , Bases de Dados Factuais , Imidazolidinas/síntese química , Cinesinas/antagonistas & inibidores , Modelos Moleculares , Quinazolinas/síntese química , Antineoplásicos/química , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Imidazolidinas/química , Imidazolidinas/farmacologia , Cinesinas/química , Ligantes , Ligação Proteica , Quinazolinas/química , Quinazolinas/farmacologia , Fuso Acromático/efeitos dos fármacos , Relação Estrutura-Atividade
13.
Semin Cell Dev Biol ; 22(9): 935-45, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22001111

RESUMO

Kinesin motor proteins are ubiquitously involved in multiple fundamental cellular processes, coordinating transport and mediating changes to cellular architecture. Thus, specific small molecule kinesin inhibitors can shed new light on the functions of kinesins and the dynamic roles in which they participate. Here we review the range of known inhibitors, their key characteristics, and specificity, and discuss their potential suitability for chemical genetics as starting points to further investigate complex kinesin-mediated processes.


Assuntos
Inibidores Enzimáticos/farmacologia , Cinesinas/antagonistas & inibidores , Cinesinas/metabolismo , Humanos , Cinesinas/química
16.
Biochem Pharmacol ; 79(6): 864-72, 2010 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-19896928

RESUMO

The kinesin Eg5 plays an essential role in bipolar spindle formation. A variety of structurally diverse inhibitors of the human kinesin Eg5, including monastrol and STLC, share the same binding pocket on Eg5, composed by helix alpha2/loop L5, and helix alpha3 of the Eg5 motor domain. Previous biochemical analysis in the inhibitor binding pocket of Eg5 identified key residues in the inhibitor binding pocket of Eg5 that in the presence of either monastrol or STLC exhibited ATPase activities similar to the untreated wild type Eg5. Here we evaluated the ability of full-length human Eg5 carrying point mutations in the drug binding pocket to confer resistance in HeLa and U2OS cells to either monastrol or STLC, as measured by the formation of bipolar spindles. Both transfected cells expressing wild type Eg5 and untransfected cells were equally sensitive to both inhibitors. Expression of Eg5 single point mutants R119A, D130A, L132A, I136A, L214A and E215A conferred significant resistance to monastrol. Certain mutations inducing monastrol resistance such as R119A, D130A and L214A also conferred significant resistance to STLC. For the first time at a cellular level, the propensity of selected Eg5 point mutants to confer drug resistance confirms the target specificity of monastrol and STLC for Eg5. These data also suggest a possible mechanism by which drug resistance may occur in tumors treated with agents targeting Eg5.


Assuntos
Cisteína/análogos & derivados , Resistência a Medicamentos , Cinesinas/genética , Cinesinas/metabolismo , Pirimidinas/farmacologia , Tionas/farmacologia , Antineoplásicos/farmacologia , Sítios de Ligação , Cisteína/farmacologia , Humanos , Concentração Inibidora 50 , Modelos Moleculares , Mutação , Ligação Proteica
17.
Cell Cycle ; 8(14): 2211-8, 2009 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-19502785

RESUMO

Members of the structural maintenance of chromosome (SMC) protein family have essential functions during mitosis, ensuring chromosome condensation (SMC2/4) and cohesion (SMC1/3). The SMC5/6 complex has been implicated in a variety of DNA maintenance processes but unlike the other SMC proteins, SMC5/6 have not been attributed any role in mitosis. Here, we find that ablation of either SMC5 or the SUMO-ligase MMS21 leads to premature sister chromatid separation prior to anaphase. The failure of normal chromosome alignment activates the spindle assembly checkpoint and blocks mitotic progression. Interestingly, there is no similar mitotic response to ablation of SMC6. Further, we show that mitotic SMC5 co-elutes from column fractions that contain MMS21 but lack SMC6. Our results thus establish that SMC5 is crucial for mitotic progression and maintenance of sister chromatid cohesion during mitosis, and that this role of SMC5 seems to be independent of the SMC5/6 complex.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Segregação de Cromossomos , Ligases/metabolismo , Mitose , Linhagem Celular Tumoral , Cromátides/efeitos dos fármacos , Proteínas Cromossômicas não Histona , Aberrações Cromossômicas , Células HeLa , Humanos , Metáfase , Fenótipo , RNA Interferente Pequeno/metabolismo
18.
Mol Biol Cell ; 20(15): 3491-502, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19494039

RESUMO

Aurora A and Aurora B, paralogue mitotic kinases, share highly similar primary sequence. Both are important to mitotic progression, but their localizations and functions are distinct. We have combined shRNA suppression with overexpression of Aurora mutants to address the cause of the distinction between Aurora A and Aurora B. Aurora A residue glycine 198 (G198), mutated to asparagine to mimic the aligned asparagine 142 (N142) of Aurora B, causes Aurora A to bind the Aurora B binding partner INCENP but not the Aurora A binding partner TPX2. The mutant Aurora A rescues Aurora B mitotic function. We conclude that binding to INCENP is alone critical to the distinct function of Aurora B. Although G198 of Aurora A is required for TPX2 binding, N142G Aurora B retains INCENP binding and Aurora B function. Thus, although a single residue change transforms Aurora A, the reciprocal mutation of Aurora B does not create Aurora A function. An Aurora A-Delta120 N-terminal truncation construct reinforces Aurora A similarity to Aurora B, because it does not associate with centrosomes but instead associates with kinetochores.


Assuntos
Substituição de Aminoácidos , Proteínas Serina-Treonina Quinases/metabolismo , Sequência de Aminoácidos , Animais , Aurora Quinase B , Aurora Quinases , Sítios de Ligação/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Células HCT116 , Células HeLa , Histonas/metabolismo , Humanos , Imunoprecipitação , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Mitose , Modelos Moleculares , Dados de Sequência Molecular , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosforilação , Ligação Proteica , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/genética , Estrutura Terciária de Proteína , Interferência de RNA , Homologia de Sequência de Aminoácidos , Transfecção , Proteínas de Xenopus/química , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Xenopus laevis
19.
Bioorg Med Chem ; 17(9): 3471-8, 2009 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-19349183

RESUMO

Pyrazolo[1,5-a]-1,3,5-triazine myoseverin derivatives 1a-c were prepared from 4-(N-methyl-N-phenylamino)-2-methylsulfanylpyrazolo[1,5-a]-1,3,5-triazine 2. Their cytotoxic activity, inhibition of tubulin polymerization, and cell cycle effects were evaluated. Compounds 1a and 1c are potent tubulin inhibitors and displayed specific antiproliferative activity in colorectal cancer cell lines at micromolar concentrations.


Assuntos
Purinas/síntese química , Purinas/farmacologia , Pirazóis/síntese química , Pirazóis/farmacologia , Triazinas/síntese química , Triazinas/farmacologia , Moduladores de Tubulina/síntese química , Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Células HCT116 , Humanos , Microtúbulos/efeitos dos fármacos , Microtúbulos/metabolismo , Moduladores de Tubulina/metabolismo , Moduladores de Tubulina/farmacologia
20.
J Med Chem ; 51(5): 1115-25, 2008 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-18266314

RESUMO

The human kinesin Eg5 is a potential drug target for cancer chemotherapy. Eg5 specific inhibitors cause cells to block in mitosis with a characteristic monoastral spindle phenotype. Prolonged metaphase block eventually leads to apoptotic cell death. S-trityl-L-cysteine (STLC) is a tight-binding inhibitor of Eg5 that prevents mitotic progression. It has proven antitumor activity as shown in the NCI 60 tumor cell line screen. It is of considerable interest to define the minimum chemical structure that is essential for Eg5 inhibition and to develop more potent STLC analogues. An initial structure-activity relationship study on a series of STLC analogues reveals the minimal skeleton necessary for Eg5 inhibition as well as indications of how to obtain more potent analogues. The most effective compounds investigated with substitutions at the para-position of one phenyl ring have an estimated K i (app) of 100 nM in vitro and induce mitotic arrest with an EC 50 of 200 nM.


Assuntos
Antineoplásicos/síntese química , Cisteína/análogos & derivados , Cinesinas/antagonistas & inibidores , Mitose , Antineoplásicos/química , Antineoplásicos/farmacologia , Sítios de Ligação , Linhagem Celular Tumoral , Cristalografia por Raios X , Cisteína/síntese química , Cisteína/química , Cisteína/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Modelos Moleculares , Ligação Proteica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...