Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.470
Filtrar
1.
J Cell Sci ; 137(2)2024 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-38149663

RESUMO

The microtubule network is formed from polymerised tubulin subunits and associating proteins, which govern microtubule dynamics and a diverse array of functions. To identify novel microtubule-binding proteins, we have developed an unbiased biochemical assay, which relies on the selective extraction of cytosolic proteins from U2OS cells, while leaving behind the microtubule network. Candidate proteins are linked to microtubules by their sensitivities to the depolymerising drug nocodazole or the microtubule-stabilising drug taxol, which is quantitated by mass spectrometry. Our approach is benchmarked by co-segregation of tubulin and previously established microtubule-binding proteins. We then identify several novel candidate microtubule-binding proteins, from which we have selected the ubiquitin E3 ligase tripartite motif-containing protein 3 (TRIM3) for further characterisation. We map TRIM3 microtubule binding to its C-terminal NHL-repeat region. We show that TRIM3 is required for the accumulation of acetylated tubulin, following treatment with taxol. Furthermore, loss of TRIM3 partially recapitulates the reduction in nocodazole-resistant microtubules characteristic of α-tubulin acetyltransferase 1 (ATAT1) depletion. These results can be explained by a decrease in ATAT1 following depletion of TRIM3 that is independent of transcription.


Assuntos
Proteômica , Tubulina (Proteína) , Tubulina (Proteína)/metabolismo , Nocodazol/farmacologia , Microtúbulos/metabolismo , Paclitaxel/farmacologia , Proteínas/metabolismo , Proteínas de Transporte/metabolismo
2.
Mol Cell ; 64(6): 1127-1134, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27984746

RESUMO

Human cancers are characterized by the presence of oncogene-induced DNA replication stress (DRS), making them dependent on repair pathways such as break-induced replication (BIR) for damaged DNA replication forks. To better understand BIR, we performed a targeted siRNA screen for genes whose depletion inhibited G1 to S phase progression when oncogenic cyclin E was overexpressed. RAD52, a gene dispensable for normal development in mice, was among the top hits. In cells in which fork collapse was induced by oncogenes or chemicals, the Rad52 protein localized to DRS foci. Depletion of Rad52 by siRNA or knockout of the gene by CRISPR/Cas9 compromised restart of collapsed forks and led to DNA damage in cells experiencing DRS. Furthermore, in cancer-prone, heterozygous APC mutant mice, homozygous deletion of the Rad52 gene suppressed tumor growth and prolonged lifespan. We therefore propose that mammalian RAD52 facilitates repair of collapsed DNA replication forks in cancer cells.


Assuntos
Proteína da Polipose Adenomatosa do Colo/genética , Ciclina E/genética , Quebras de DNA de Cadeia Dupla , DNA/genética , Osteossarcoma/genética , Proteína Rad52 de Recombinação e Reparo de DNA/genética , Reparo de DNA por Recombinação , Proteína da Polipose Adenomatosa do Colo/deficiência , Animais , Sistemas CRISPR-Cas , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ciclina E/metabolismo , DNA/metabolismo , Fase G1 , Expressão Gênica , Instabilidade Genômica , Humanos , Camundongos , Camundongos Knockout , Nocodazol/farmacologia , Osteossarcoma/metabolismo , Osteossarcoma/mortalidade , Osteossarcoma/patologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteína Rad52 de Recombinação e Reparo de DNA/antagonistas & inibidores , Proteína Rad52 de Recombinação e Reparo de DNA/metabolismo , Fase S , Estresse Fisiológico , Análise de Sobrevida
3.
Proc Natl Acad Sci U S A ; 118(7)2021 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-33558236

RESUMO

The number and activity of Cav1.2 channels in the cardiomyocyte sarcolemma tunes the magnitude of Ca2+-induced Ca2+ release and myocardial contraction. ß-Adrenergic receptor (ßAR) activation stimulates sarcolemmal insertion of CaV1.2. This supplements the preexisting sarcolemmal CaV1.2 population, forming large "superclusters" wherein neighboring channels undergo enhanced cooperative-gating behavior, amplifying Ca2+ influx and myocardial contractility. Here, we determine this stimulated insertion is fueled by an internal reserve of early and recycling endosome-localized, presynthesized CaV1.2 channels. ßAR-activation decreased CaV1.2/endosome colocalization in ventricular myocytes, as it triggered "emptying" of endosomal CaV1.2 cargo into the t-tubule sarcolemma. We examined the rapid dynamics of this stimulated insertion process with live-myocyte imaging of channel trafficking, and discovered that CaV1.2 are often inserted into the sarcolemma as preformed, multichannel clusters. Similarly, entire clusters were removed from the sarcolemma during endocytosis, while in other cases, a more incremental process suggested removal of individual channels. The amplitude of the stimulated insertion response was doubled by coexpression of constitutively active Rab4a, halved by coexpression of dominant-negative Rab11a, and abolished by coexpression of dominant-negative mutant Rab4a. In ventricular myocytes, ßAR-stimulated recycling of CaV1.2 was diminished by both nocodazole and latrunculin-A, suggesting an essential role of the cytoskeleton in this process. Functionally, cytoskeletal disruptors prevented ßAR-activated Ca2+ current augmentation. Moreover, ßAR-regulation of CaV1.2 was abolished when recycling was halted by coapplication of nocodazole and latrunculin-A. These findings reveal that ßAR-stimulation triggers an on-demand boost in sarcolemmal CaV1.2 abundance via targeted Rab4a- and Rab11a-dependent insertion of channels that is essential for ßAR-regulation of cardiac CaV1.2.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Miócitos Cardíacos/metabolismo , Receptores Adrenérgicos beta/metabolismo , Sarcolema/metabolismo , Proteínas rab4 de Ligação ao GTP/metabolismo , Animais , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Linhagem Celular , Células Cultivadas , Endossomos/metabolismo , Feminino , Ventrículos do Coração/citologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Nocodazol/farmacologia , Transporte Proteico , Tiazolidinas/farmacologia
4.
J Chem Inf Model ; 63(22): 7228-7238, 2023 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-37947759

RESUMO

Carbendazim derivatives, commonly used as antiparasitic drugs, have shown potential as anticancer agents due to their ability to induce cell cycle arrest and apoptosis in human cancer cells by inhibiting tubulin polymerization. Crystallographic structures of α/ß-tubulin multimers complexed with nocodazole and mebendazole, two carbendazim derivatives with potent anticancer activity, highlighted the possibility of designing compounds that occupy both benzimidazole- and colchicine-binding sites. In addition, previous studies have demonstrated that the incorporation of a phenoxy group at position 5/6 of carbendazim increases the antiproliferative activity in cancer cell lines. Despite the significant progress made in identifying new tubulin-targeting anticancer compounds, further modifications are needed to enhance their potency and safety. In this study, we explored the impact of modifying the phenoxy substitution pattern on antiproliferative activity. Alchemical free energy calculations were used to predict the binding free energy difference upon ligand modification and define the most viable path for structure optimization. Based on these calculations, seven compounds were synthesized and evaluated against lung and colon cancer cell lines. Our results showed that compound 5a, which incorporates an α-naphthyloxy substitution, exhibits the highest antiproliferative activity against both cancer lines (SK-LU-1 and SW620, IC50 < 100 nM) and induces morphological changes in the cells associated with mitotic arrest and mitotic catastrophe. Nevertheless, the tubulin polymerization assay showed that 5a has a lower inhibitory potency than nocodazole. Molecular dynamics simulations suggested that this low antitubulin activity could be associated with the loss of the key H-bond interaction with V236. This study provides insights into the design of novel carbendazim derivatives with anticancer activity.


Assuntos
Antineoplásicos , Moduladores de Tubulina , Humanos , Moduladores de Tubulina/química , Estrutura Molecular , Relação Estrutura-Atividade , Nocodazol/farmacologia , Tubulina (Proteína)/metabolismo , Proliferação de Células , Simulação de Acoplamento Molecular , Antineoplásicos/farmacologia , Antineoplásicos/química , Polimerização , Ensaios de Seleção de Medicamentos Antitumorais
5.
Mol Cell ; 57(5): 824-835, 2015 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-25661489

RESUMO

Regulated recruitment of the kinase-adaptor complex BUB1/BUB3 to kinetochores is crucial for correcting faulty chromosome-spindle attachments and for spindle assembly checkpoint (SAC) signaling. BUB1/BUB3 localizes to kinetochores by binding phosphorylated MELT motifs (MELpT) in the kinetochore scaffold KNL1. Human KNL1 has 19 repeats that contain a MELT-like sequence. The repeats are, however, larger than MELT, and repeat sequences can vary significantly. Using systematic screening, we show that only a limited number of repeats is "active." Repeat activity correlates with the presence of a vertebrate-specific SHT motif C-terminal to the MELT sequence. SHT motifs are phosphorylated by MPS1 in a manner that requires prior phosphorylation of MELT. Phospho-SHT (SHpT) synergizes with MELpT in BUB3/BUB1 binding in vitro and in cells, and human BUB3 mutated in a predicted SHpT-binding surface cannot localize to kinetochores. Our data show sequential multisite regulation of the KNL1-BUB1/BUB3 interaction and provide mechanistic insight into evolution of the KNL1-BUB3 interface.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Cinetocoros/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Mitose , Proteínas Serina-Treonina Quinases/metabolismo , Motivos de Aminoácidos/genética , Sequência de Aminoácidos , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Células HeLa , Humanos , Immunoblotting , Cinetocoros/efeitos dos fármacos , Pontos de Checagem da Fase M do Ciclo Celular/genética , Proteínas Associadas aos Microtúbulos/genética , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Nocodazol/farmacologia , Fosforilação , Proteínas de Ligação a Poli-ADP-Ribose , Ligação Proteica , Proteínas Serina-Treonina Quinases/genética , Estrutura Terciária de Proteína , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Interferência de RNA , Sequências Repetitivas de Aminoácidos/genética , Homologia de Sequência de Aminoácidos , Imagem com Lapso de Tempo , Moduladores de Tubulina/farmacologia
6.
J Cell Sci ; 133(18)2020 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-32878943

RESUMO

Human retinal pigment epithelial-1 (RPE-1) cells are increasingly being used as a model to study mitosis because they represent a non-transformed alternative to cancer cell lines, such as HeLa cervical adenocarcinoma cells. However, the lack of an efficient method to synchronize RPE-1 cells in mitosis precludes their application for large-scale biochemical and proteomics assays. Here, we report a protocol to synchronize RPE-1 cells based on sequential treatments with the Cdk4 and Cdk6 inhibitor PD 0332991 (palbociclib) and the microtubule-depolymerizing drug nocodazole. With this method, the vast majority (80-90%) of RPE-1 cells arrested at prometaphase and exited mitosis synchronously after release from nocodazole. Moreover, the cells fully recovered and re-entered the cell cycle after the palbociclib-nocodazole block. Finally, we show that this protocol could be successfully employed for the characterization of the protein-protein interaction network of the kinetochore protein Ndc80 by immunoprecipitation coupled with mass spectrometry. This synchronization method significantly expands the versatility and applicability of RPE-1 cells to the study of cell division and might be applied to other cell lines that do not respond to treatments with DNA synthesis inhibitors.


Assuntos
Cinetocoros , Mitose , Humanos , Nocodazol/farmacologia , Prometáfase , Pigmentos da Retina
7.
Nat Chem Biol ; 16(5): 546-555, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32152539

RESUMO

The anaphase-promoting complex/cyclosome (APC/C) is a ubiquitin ligase that initiates anaphase and mitotic exit. APC/C is activated by Cdc20 and inhibited by the mitotic checkpoint complex (MCC), which delays mitotic exit when the spindle assembly checkpoint (SAC) is activated. We previously identified apcin as a small molecule ligand of Cdc20 that inhibits APC/CCdc20 and prolongs mitosis. Here we find that apcin paradoxically shortens mitosis when SAC activity is high. These opposing effects of apcin arise from targeting of a common binding site in Cdc20 required for both substrate ubiquitination and MCC-dependent APC/C inhibition. Furthermore, we found that apcin cooperates with p31comet to relieve MCC-dependent inhibition of APC/C. Apcin therefore causes either net APC/C inhibition, prolonging mitosis when SAC activity is low, or net APC/C activation, shortening mitosis when SAC activity is high, demonstrating that a small molecule can produce opposing biological effects depending on regulatory context.


Assuntos
Ciclossomo-Complexo Promotor de Anáfase/antagonistas & inibidores , Carbamatos/farmacologia , Proteínas Cdc20/antagonistas & inibidores , Diaminas/farmacologia , Mitose/efeitos dos fármacos , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Ciclossomo-Complexo Promotor de Anáfase/metabolismo , Sítios de Ligação , Proteínas de Ciclo Celular/metabolismo , Ciclina B1/metabolismo , Células HCT116 , Células HeLa , Humanos , Nocodazol/farmacologia , Proteínas Nucleares/metabolismo , Fuso Acromático/efeitos dos fármacos , Fuso Acromático/metabolismo , Telomerase/genética , Telomerase/metabolismo , Imagem com Lapso de Tempo , Ubiquitinação
8.
Nucleic Acids Res ; 48(14): e83, 2020 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-32526036

RESUMO

Mass spectrometry (MS)-based quantitative proteomics experiments frequently generate data with missing values, which may profoundly affect downstream analyses. A wide variety of imputation methods have been established to deal with the missing-value issue. To date, however, there is a scarcity of efficient, systematic, and easy-to-handle tools that are tailored for proteomics community. Herein, we developed a user-friendly and powerful stand-alone software, NAguideR, to enable implementation and evaluation of different missing value methods offered by 23 widely used missing-value imputation algorithms. NAguideR further evaluates data imputation results through classic computational criteria and, unprecedentedly, proteomic empirical criteria, such as quantitative consistency between different charge-states of the same peptide, different peptides belonging to the same proteins, and individual proteins participating protein complexes and functional interactions. We applied NAguideR into three label-free proteomic datasets featuring peptide-level, protein-level, and phosphoproteomic variables respectively, all generated by data independent acquisition mass spectrometry (DIA-MS) with substantial biological replicates. The results indicate that NAguideR is able to discriminate the optimal imputation methods that are facilitating DIA-MS experiments over those sub-optimal and low-performance algorithms. NAguideR further provides downloadable tables and figures supporting flexible data analysis and interpretation. NAguideR is freely available at http://www.omicsolution.org/wukong/NAguideR/ and the source code: https://github.com/wangshisheng/NAguideR/.


Assuntos
Proteômica/métodos , Software , Células/efeitos dos fármacos , Simulação por Computador , Conjuntos de Dados como Assunto , Formaldeído/farmacologia , Humanos , Espectrometria de Massas , Microtúbulos/efeitos dos fármacos , Nocodazol/farmacologia , Precursores de Proteínas/química
9.
Int J Mol Sci ; 23(8)2022 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-35456926

RESUMO

The connection between cytoskeleton alterations and diseases is well known and has stimulated research on cell mechanics, aiming to develop reliable biomarkers. In this study, we present results on rheological, adhesion, and morphological properties of primary rat cardiac fibroblasts, the cytoskeleton of which was altered by treatment with cytochalasin D (Cyt-D) and nocodazole (Noc), respectively. We used two complementary techniques: quartz crystal microbalance (QCM) and digital holographic microscopy (DHM). Qualitative data on cell viscoelasticity and adhesion changes at the cell-substrate near-interface layer were obtained with QCM, while DHM allowed the measurement of morphological changes due to the cytoskeletal alterations. A rapid effect of Cyt-D was observed, leading to a reduction in cell viscosity, loss of adhesion, and cell rounding, often followed by detachment from the surface. Noc treatment, instead, induced slower but continuous variations in the rheological behavior for four hours of treatment. The higher vibrational energy dissipation reflected the cell's ability to maintain a stable attachment to the substrate, while a cytoskeletal rearrangement occurs. In fact, along with the complete disaggregation of microtubules at prolonged drug exposure, a compensatory effect of actin polymerization emerged, with increased stress fiber formation.


Assuntos
Microscopia , Técnicas de Microbalança de Cristal de Quartzo , Animais , Citocalasina D/farmacologia , Citoesqueleto/metabolismo , Microtúbulos , Nocodazol/farmacologia , Técnicas de Microbalança de Cristal de Quartzo/métodos , Ratos , Viscosidade
10.
Int J Mol Sci ; 23(2)2022 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-35054928

RESUMO

The brain capillary endothelium is highly regulatory, maintaining the chemical stability of the brain's microenvironment. The role of cytoskeletal proteins in tethering nanotubules (TENTs) during barrier-genesis was investigated using the established immortalized mouse brain endothelial cell line (bEnd5) as an in vitro blood-brain barrier (BBB) model. The morphology of bEnd5 cells was evaluated using both high-resolution scanning electron microscopy and immunofluorescence to evaluate treatment with depolymerizing agents Cytochalasin D for F-actin filaments and Nocodazole for α-tubulin microtubules. The effects of the depolymerizing agents were investigated on bEnd5 monolayer permeability by measuring the transendothelial electrical resistance (TEER). The data endorsed that during barrier-genesis, F-actin and α-tubulin play a cytoarchitectural role in providing both cell shape dynamics and cytoskeletal structure to TENTs forming across the paracellular space to provide cell-cell engagement. Western blot analysis of the treatments suggested a reduced expression of both proteins, coinciding with a reduction in the rates of cellular proliferation and decreased TEER. The findings endorsed that TENTs provide alignment of the paracellular (PC) spaces and tight junction (TJ) zones to occlude bEnd5 PC spaces. The identification of specific cytoskeletal structures in TENTs endorsed the postulate of their indispensable role in barrier-genesis and the maintenance of regulatory permeability across the BBB.


Assuntos
Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/ultraestrutura , Proteínas do Citoesqueleto/metabolismo , Actinas/metabolismo , Animais , Biomarcadores , Linhagem Celular , Proteínas do Citoesqueleto/antagonistas & inibidores , Proteínas do Citoesqueleto/química , Proteínas do Citoesqueleto/genética , Células Endoteliais/metabolismo , Células Endoteliais/ultraestrutura , Endotélio Vascular/metabolismo , Endotélio Vascular/ultraestrutura , Imunofluorescência , Expressão Gênica , Camundongos , Nocodazol/farmacologia , Permeabilidade/efeitos dos fármacos
11.
J Cell Sci ; 132(3)2019 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-30659115

RESUMO

The pathological significance of Tau (encoded by MAPT) in mechanisms driving cell migration in glioblastoma is unclear. By using an shRNA approach to deplete microtubule-stabilizing Tau in U87 cells, we determined its impact on cytoskeletal coordination during migration. We demonstrated here that the motility of these Tau-knockdown cells (shTau cells) was significantly (36%) lower than that of control cells. The shTau cells displayed a slightly changed motility in the presence of nocodazole, which inhibits microtubule formation. Such reduced motility of shTau cells was characterized by a 28% lower number of microtubule bundles at the non-adhesive edges of the tails. In accordance with Tau-stabilized microtubules being required for cell movement, measurements of the front, body and rear section displacements of cells showed inefficient tail retraction in shTau cells. The tail retraction was restored by treatment with Y27632, an inhibitor of Rho-ROCK signaling. Moreover, we clearly identified that shTau cells displayed relocation of the active phosphorylated form of p190-RhoGAP (also known as ARHGAP35), which inhibits Rho-ROCK signaling, and focal adhesion kinase (FAK, also known as PTK2) in cell bodies. In conclusion, our findings indicate that Tau governs the remodeling of microtubule and actin networks for the retraction of the tail of cells, which is necessary for effective migration.


Assuntos
Citoesqueleto de Actina/metabolismo , Fatores de Troca do Nucleotídeo Guanina/genética , Microtúbulos/metabolismo , Neuroglia/metabolismo , Proteínas Repressoras/genética , Quinases Associadas a rho/genética , Proteínas tau/genética , Citoesqueleto de Actina/efeitos dos fármacos , Citoesqueleto de Actina/ultraestrutura , Actinas/genética , Actinas/metabolismo , Amidas/farmacologia , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Quinase 1 de Adesão Focal/genética , Quinase 1 de Adesão Focal/metabolismo , Regulação da Expressão Gênica , Fatores de Troca do Nucleotídeo Guanina/antagonistas & inibidores , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Microtúbulos/efeitos dos fármacos , Microtúbulos/ultraestrutura , Neuroglia/efeitos dos fármacos , Neuroglia/patologia , Nocodazol/farmacologia , Piridinas/farmacologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteínas Repressoras/antagonistas & inibidores , Proteínas Repressoras/metabolismo , Transdução de Sinais , Quinases Associadas a rho/antagonistas & inibidores , Quinases Associadas a rho/metabolismo , Proteínas tau/antagonistas & inibidores , Proteínas tau/metabolismo
12.
Am J Physiol Heart Circ Physiol ; 320(4): H1403-H1410, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33577432

RESUMO

Excessive vascular permeability occurs in inflammatory disease processes. Vascular endothelial cadherin (VE-cadherin) is an adhesion protein that controls vascular permeability. We identified monoclonal antibodies (mAbs) to human VE-cadherin that activate cell adhesion and inhibit the increased permeability of endothelial cell monolayers induced by thrombin receptor activator peptide-6 (TRAP-6). Two mAbs, 8A12c and 3A5a, reduce permeability, whereas an inhibitory mAb, 2E11d, enhances permeability. Activating mAbs also reduce permeability induced by tumor necrosis factor-α (TNF-α) and vascular endothelial cell growth factor (VEGF). The activating mAbs also stabilize the organization of the adherens junctions that are disrupted by TRAP-6, VEGF, or TNF-α. The activating mAbs act directly on the adhesive function of VE-cadherin because they did not block the accumulation of actin filaments stimulated by TRAP-6 and enhance physical cell-cell adhesion of VE-cadherin-expressing tissue culture cells. Therefore, VE-cadherin function can be regulated at the cell surface to control endothelial permeability.NEW & NOTEWORTHY Excessive vascular permeability is a serious complication of many inflammatory disease conditions. We have developed monoclonal antibodies that inhibit increases in endothelial monolayer permeability induced by several signaling factors by activating VE-cadherin mediated adhesion and stabilizing cell junctions. These antibodies and/or the mechanisms they reveal may lead to important therapeutics to treat vascular leakiness and inflammation.


Assuntos
Junções Aderentes/efeitos dos fármacos , Anticorpos Monoclonais/farmacologia , Caderinas/agonistas , Permeabilidade Capilar/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Junções Aderentes/metabolismo , Antígenos CD/metabolismo , Caderinas/antagonistas & inibidores , Caderinas/metabolismo , Linhagem Celular Tumoral , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Nocodazol/farmacologia , Oligopeptídeos/farmacologia , Receptores de Trombina/agonistas , Receptores de Trombina/metabolismo , Transdução de Sinais , Moduladores de Tubulina/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Fator A de Crescimento do Endotélio Vascular/farmacologia
13.
Neurobiol Learn Mem ; 183: 107468, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34058346

RESUMO

Accurate and precise timing is crucial for complex and purposeful behaviors, such as foraging for food or playing a musical instrument. The brain is capable of processing temporal information in a coordinated manner, as if it contains an 'internal clock'. Similar to the need for the brain to orient itself in space in order to understand its surroundings, temporal orientation and tracking is an essential component of cognition as well. While there have been multiple models explaining the neural correlates of timing, independent lines of research appear to converge on the conclusion that populations of neurons in the dorsal striatum encode information relating to where a subject is in time relative to an anticipated goal. Similar to other learning processes, acquisition and maintenance of this temporal information is dependent on synaptic plasticity. Microtubules are cytoskeletal proteins that have been implicated in synaptic plasticity mechanisms and therefore are considered key elements in learning and memory. In this study, we investigated the role of microtubule dynamics in temporal learning by local infusions of microtubule stabilizing and destabilizing agents into the dorsolateral striatum. Our results suggested a bidirectional role for microtubules in timing, such that microtubule stabilization improves the maintenance of learned target durations, but impairs the acquisition of a novel duration. On the other hand, microtubule destabilization enhances the acquisition of novel target durations, while compromising the maintenance of previously learned durations. These findings suggest that microtubule dynamics plays an important role in synaptic plasticity mechanisms in the dorsolateral striatum, which in turn modulates temporal learning and time perception.


Assuntos
Aprendizagem/efeitos dos fármacos , Microtúbulos/efeitos dos fármacos , Neostriado/efeitos dos fármacos , Plasticidade Neuronal/efeitos dos fármacos , Percepção do Tempo/efeitos dos fármacos , Moduladores de Tubulina/farmacologia , Animais , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/fisiologia , Aprendizagem/fisiologia , Proteínas dos Microtúbulos/efeitos dos fármacos , Proteínas dos Microtúbulos/fisiologia , Microtúbulos/fisiologia , Neostriado/fisiologia , Nocodazol/farmacologia , Paclitaxel/farmacologia , Ratos
14.
Toxicol Appl Pharmacol ; 426: 115607, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34089742

RESUMO

Sertoli cells (SC) structurally support and transport nutrients to germ cells during spermatogenesis facilitated by an active cytoskeleton. Chemical perturbation of SC microtubule (MT) dynamics instability leads to premature germ cell exfoliation demonstrating that this process is essential for male fertility, yet the effects of MT damaging drugs on SC lipid metabolism have been less explored. The aim of this study was to advance our understanding of how adequate SC MT dynamicity is needed to finely tune lipid homeostasis. To elucidate the role of MT dynamics instability on the latter, we suppressed MT dynamicity by long-term exposures to 10 nM of nocodazole (NCZ) on TM4-SC cultures. Inhibition of MT dynamics instability affected the distribution of [3H] arachidonate on TM4-SC. Triacylglycerols (TAG) exhibited a higher proportion of the [3H] label, with significantly lower percentages in the mitochondrial phospholipid cardiolipin, and notably, also in phosphatidylethanolamine. A noteworthy and progressive accumulation of lipid droplets during the period of exposure to NCZ was accompanied by increased TAG levels but not cholesterol levels in TM4-SC. NCZ-exposed cells reduced their mitochondrial membrane potential and increased ROS production without triggering apoptosis, had a compromised autophagic flux, and lost their transferrin expression. Although SC morphology was preserved, the NCZ-exposed cells displayed alteration of the normal organization of microfilaments (f-actin) and intermediate filaments (vimentin). Our findings suggest that a preserved MT dynamicity is essential in the maintenance of lipid and fatty acids homeostasis in SC, and thus highlights a novel target in these cells for drugs that impair MT dynamicity.


Assuntos
Metabolismo dos Lipídeos , Microtúbulos/metabolismo , Células de Sertoli/metabolismo , Animais , Antineoplásicos/farmacologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Proteínas do Citoesqueleto/metabolismo , Homeostase/efeitos dos fármacos , Gotículas Lipídicas/metabolismo , Metabolismo dos Lipídeos/efeitos dos fármacos , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Microtúbulos/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/fisiologia , Nocodazol/farmacologia , Células de Sertoli/efeitos dos fármacos , Moduladores de Tubulina/farmacologia
15.
PLoS Comput Biol ; 16(2): e1007649, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32084125

RESUMO

In multi-ciliated cells, directed and synchronous ciliary beating in the apical membrane occurs through appropriate configuration of basal bodies (BBs, roots of cilia). Although it has been experimentally shown that the position and orientation of BBs are coordinated by apical cytoskeletons (CSKs), such as microtubules (MTs), and planar cell polarity (PCP), the underlying mechanism for achieving the patterning of BBs is not yet understood. In this study, we propose that polarity in bundles of apical MTs play a crucial role in the patterning of BBs. First, the necessity of the polarity was discussed by theoretical consideration on the symmetry of the system. The existence of the polarity was investigated by measuring relative angles between the MTs and BBs using published experimental data. Next, a mathematical model for BB patterning was derived by combining the polarity and self-organizational ability of CSKs. In the model, BBs were treated as finite-size particles in the medium of CSKs and excluded volume effects between BBs and CSKs were taken into account. The model reproduces the various experimental observations, including normal and drug-treated phenotypes. Our model with polarity provides a coherent and testable mechanism for apical BB pattern formation. We have also discussed the implication of our study on cell chirality.


Assuntos
Corpos Basais/fisiologia , Cílios/fisiologia , Citoesqueleto/fisiologia , Animais , Membrana Celular , Polaridade Celular , Simulação por Computador , Elasticidade , Células Epiteliais/citologia , Camundongos , Microtúbulos/fisiologia , Modelos Teóricos , Nocodazol/farmacologia , Fenótipo , Traqueia/fisiologia
16.
Nucleic Acids Res ; 47(1): 103-121, 2019 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-30321399

RESUMO

The oncogenic transcription factor B-Myb is an essential regulator of late cell cycle genes whose activation by phosphorylation is still poorly understood. We describe a stepwise phosphorylation mechanism of B-Myb, which involves sequential phosphorylations mediated by cyclin-dependent kinase (Cdk) and Polo-like kinase 1 (Plk1) and Pin1-facilitated peptidyl-prolyl cis/trans isomerization. Our data suggest a model in which initial Cdk-dependent phosphorylation of B-Myb enables subsequent Pin1 binding and Pin1-induced conformational changes of B-Myb. This, in turn, initiates further phosphorylation of Cdk-phosphosites, enabling Plk1 docking and subsequent Plk1-mediated phosphorylation of B-Myb to finally allow B-Myb to stimulate transcription of late cell cycle genes. Our observations reveal novel mechanistic hierarchies of B-Myb phosphorylation and activation and uncover regulatory principles that might also apply to other Myb family members. Strikingly, overexpression of B-Myb and of factors mediating its activation strongly correlates with adverse prognoses for tumor patients, emphasizing B-Myb's role in tumorigenesis.


Assuntos
Proteínas de Ciclo Celular/genética , Peptidilprolil Isomerase de Interação com NIMA/genética , Neoplasias/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/genética , Transativadores/genética , Carcinogênese/efeitos dos fármacos , Ciclo Celular/genética , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/química , Quinases Ciclina-Dependentes/antagonistas & inibidores , Quinases Ciclina-Dependentes/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HeLa , Células Hep G2 , Humanos , Mitose/efeitos dos fármacos , Peptidilprolil Isomerase de Interação com NIMA/antagonistas & inibidores , Peptidilprolil Isomerase de Interação com NIMA/química , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Nocodazol/farmacologia , Peptidilprolil Isomerase/genética , Fosforilação/efeitos dos fármacos , Conformação Proteica/efeitos dos fármacos , Multimerização Proteica/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/química , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/química , Roscovitina/farmacologia , Timidina/farmacologia , Transativadores/química , Transcrição Gênica/efeitos dos fármacos , Quinase 1 Polo-Like
17.
Proc Natl Acad Sci U S A ; 115(16): 4246-4251, 2018 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-29610346

RESUMO

Flaviviruses enter host cells through the process of clathrin-mediated endocytosis, and the spectrum of host factors required for this process are incompletely understood. Here we found that lymphocyte antigen 6 locus E (LY6E) promotes the internalization of multiple flaviviruses, including West Nile virus, Zika virus, and dengue virus. Perhaps surprisingly, LY6E is dispensable for the internalization of the endogenous cargo transferrin, which is also dependent on clathrin-mediated endocytosis for uptake. Since viruses are substantially larger than transferrin, we reasoned that LY6E may be required for uptake of larger cargoes and tested this using transferrin-coated beads of similar size as flaviviruses. LY6E was indeed required for the internalization of transferrin-coated beads, suggesting that LY6E is selectively required for large cargo. Cell biological studies found that LY6E forms tubules upon viral infection and bead internalization, and we found that tubule formation was dependent on RNASEK, which is also required for flavivirus internalization, but not transferrin uptake. Indeed, we found that RNASEK is also required for the internalization of transferrin-coated beads, suggesting it functions upstream of LY6E. These LY6E tubules resembled microtubules, and we found that microtubule assembly was required for their formation and flavivirus uptake. Since microtubule end-binding proteins link microtubules to downstream activities, we screened the three end-binding proteins and found that EB3 promotes virus uptake and LY6E tubularization. Taken together, these results highlight a specialized pathway required for the uptake of large clathrin-dependent endocytosis cargoes, including flaviviruses.


Assuntos
Flavivirus/fisiologia , Internalização do Vírus , Antígenos de Superfície/genética , Antígenos de Superfície/fisiologia , Linhagem Celular Tumoral , Vírus da Dengue/fisiologia , Endocitose/efeitos dos fármacos , Endocitose/fisiologia , Endorribonucleases/fisiologia , Éteres/farmacologia , Proteínas Ligadas por GPI/deficiência , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/fisiologia , Técnicas de Silenciamento de Genes , Humanos , Microesferas , Proteínas Associadas aos Microtúbulos/fisiologia , Microtúbulos/efeitos dos fármacos , Microtúbulos/fisiologia , Nocodazol/farmacologia , Compostos de Espiro/farmacologia , Transferrina , Vírus do Nilo Ocidental/fisiologia , Zika virus/fisiologia
18.
J Neurosci ; 39(36): 7118-7131, 2019 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-31391261

RESUMO

Complex neural circuits are built from axonal branches that allow each neuron to connect with multiple targets. During development, maturation of nascent branches depends on stabilization of newly assembled or transported microtubules, which are thought to be regulated by microtubule-associated proteins (MAPs). However, because many known MAPs inhibit branch formation, it is not clear which MAP is responsible for regulating microtubule stability during branch development. Here, we show that MAP7, a less-well understood MAP that is localized to branch junctions, provides a key molecular mechanism to regulate microtubule stability during branch formation. In developing rodent sensory neurons of mixed sex, MAP7 is required for branch maturation mainly by preventing branch retraction. This function is mediated by the ability of MAP7 to control microtubule stability, as microtubules are more stable at branch junctions where MAP7 is localized. Consistently, nascent branches depleted of MAP7 have decreased stable microtubules but increased dynamic microtubules. Moreover, MAP7 binds to the acetylated and stable region of individual microtubules and avoids the dynamic plus end, thereby creating a boundary that prevents microtubule depolymerization and rescues microtubule polymerization. This unique binding property, which is not observed for other MAPs, can prevent branch retraction caused by laser-induced severing or nocodazole-induced microtubule depolymerization. Together, our study identifies a novel molecular mechanism mediated by MAP7 to regulate microtubule stability and strengthen branches at different stages of axonal branch morphogenesis.SIGNIFICANCE STATEMENT Development and maintenance of axonal branches rely on microtubule stability, but the underlying molecular mechanisms are not fully understood. Here, we show that MAP7, a unique protein that interacts with both microtubules and the motor protein kinesin-1, plays a key role at branch junctions. MAP7 stabilizes microtubules in nascent branches and prevents branch retraction during branch maturation or after laser-induced injury. MAP7 also binds to the acetylated region of microtubules to prevent depolymerization and rescue polymerization. This unique binding property supports a novel mechanism mediated by MAP7 to cooperate with other MAPs and control microtubule stability during axonal branch development. This mechanism could also impact microtubule regulation in branch regeneration after nerve injury.


Assuntos
Axônios/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Neurogênese , Animais , Axônios/efeitos dos fármacos , Células COS , Células Cultivadas , Chlorocebus aethiops , Feminino , Masculino , Camundongos , Nocodazol/farmacologia , Ligação Proteica , Ratos , Ratos Sprague-Dawley , Células Receptoras Sensoriais/citologia , Células Receptoras Sensoriais/efeitos dos fármacos , Células Receptoras Sensoriais/metabolismo , Moduladores de Tubulina/farmacologia
19.
J Mol Cell Cardiol ; 143: 107-119, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32353354

RESUMO

AIMS: Phospholamban (PLB) stoichiometrically regulates the cardiac Ca2+ pump (SERCA2a) in the sarcoplasmic reticulum (SR); but in the nuclear envelope (NE) of cardiomyocytes (CMs), the PLB to SERCA2a molar ratio is higher, which highlights our poor understanding of how SR proteins distribute to their functional subcompartments. By tracking newly made PLB and SERCA2a in CMs, we will elucidate underlying cellular pathways responsible for their unique intracellular distributions. METHODS AND RESULTS: Highly specific monoclonal antibodies were used to compare the subcellular distributions of SERCA2a, PLB, and junctin (JCN) in dog heart tissue. The data supported a view that both non-junctional and junctional SR proteins are all prominently enriched in transverse stretches of SR tubules, along the edges of sarcomeres (SR z-tubules). To understand the genesis of these steady state distributions, we analyzed confocal immunofluorescence images of adult rat CMs after acute expression (12-48 h) of the dog ortholog of PLB (dPLB) or dSERCA2a. Newly made dog proteins in rat CMs were detected using dog-specific monoclonal antibodies. By 12-24 h, dSERCA2a had accumulated within the NE in a punctate pattern, presumably reflecting initial sites of biosynthesis. Over the next 24-48 h, higher levels of dSERCA2a immunofluorescence accumulated in transverse/radial SR tubules, aligned along sarcolemmal transverse (T)-tubules, and extending from NE puncta. The patterns of SR tubules carrying dSERCA2a overlapped with those for newly made JCN, suggesting a common Nuclear Envelope to SR along T-tubules or NEST pathway for SR proteins. In contrast to the SERCA2a distribution pattern, dPLB accumulated uniformly in the NE, without visible puncta. With co-expression of dSERCA2a, however, PLB no longer uniformly filled the NE, but instead moved together with SERCA2a to form bright NE puncta, from which the two proteins then trafficked anterogradely. CONCLUSION: Expression of dog SR protein orthologs (dSERCA2a, dPLB, and dJCN) for as little as 48 h reproduces their characteristic steady state distributions. Detailed analyses of the time courses of protein accumulation suggest a possible mechanism by which PLB distributes to both the NE and SR, unlike SERCA2a. SERCA2a moves in SR z-tubules directly from rough ER, along pathways that are in common with those used by junctional SR proteins. A different trafficking route for PLB away the rough ER/NE led to its accumulation in the NE, a process that may account for its enrichment in NE in situ. Association of SERCA2a with PLB from this NE pool enhanced PLB trafficking along the NEST pathway, contributing to steady state stoichiometry and physiologically regulated SERCA2a.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Miócitos Cardíacos/metabolismo , Membrana Nuclear/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Retículo Sarcoplasmático/metabolismo , Sequência de Aminoácidos , Animais , Biomarcadores , Feminino , Imunofluorescência , Humanos , Nocodazol/farmacologia , Transporte Proteico/efeitos dos fármacos , Ratos , Transdução de Sinais/efeitos dos fármacos
20.
Biochemistry ; 59(28): 2650-2659, 2020 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-32567840

RESUMO

The dynamic cytoskeletal network of microtubules and actin filaments can be disassembled by drugs. Cytoskeletal drugs work by perturbing the monomer-polymer equilibrium, thus changing the size and number of macromolecular crowders inside cells. Changes in both crowding and nonspecific surface interactions ("sticking") following cytoskeleton disassembly can affect the protein stability, structure, and function directly or indirectly by changing the fluidity of the cytoplasm and altering the crowding and sticking of other macromolecules in the cytoplasm. The effect of cytoskeleton disassembly on protein energy landscapes inside cells has yet to be observed. Here we have measured the effect of several cytoskeletal drugs on the folding energy landscape of two FRET-labeled proteins with different in vitro sensitivities to macromolecular crowding. Phosphoglycerate kinase (PGK) was previously shown to be more sensitive to crowding, whereas variable major protein-like sequence expressed (VlsE) was previously shown to be more sensitive to sticking. The in-cell effects of drugs that depolymerize either actin filaments (cytochalasin D and latrunculin B) or microtubules (nocodazole and vinblastine) were compared. The crowding sensor protein CrH2-FRET verified that cytoskeletal drugs decrease the extent of crowding inside cells despite also reducing the overall cell volume. The decreased compactness and folding stability of PGK could be explained by the decreased extent of crowding induced by these drugs. VlsE's opposite response to the drugs shows that depolymerization of the cytoskeleton also changes sticking in the cellular milieu. Our results demonstrate that perturbation of the monomer-polymer cytoskeletal equilibrium, for example, during natural cell migration or stresses from drug treatment, has off-target effects on the energy landscapes of proteins in the cell.


Assuntos
Nocodazol/farmacologia , Dobramento de Proteína/efeitos dos fármacos , Proteínas/química , Moduladores de Tubulina/farmacologia , Vimblastina/farmacologia , Antígenos de Bactérias/química , Proteínas de Bactérias/química , Borrelia burgdorferi/química , Linhagem Celular , Tamanho Celular/efeitos dos fármacos , Citoesqueleto/química , Citoesqueleto/efeitos dos fármacos , Transferência Ressonante de Energia de Fluorescência , Humanos , Lipoproteínas/química , Modelos Moleculares , Fosfoglicerato Quinase/química , Estabilidade Proteica/efeitos dos fármacos , Leveduras/enzimologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA