Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.978
Filtrar
1.
Nanoscale ; 16(16): 7976-7987, 2024 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-38567463

RESUMO

Collective functionalization of the phytochemicals of medicinal herbs on nanoparticles is emerging as a potential cancer therapeutic strategy. This study presents the facile synthesis of surface-functionalized gold nanoparticles using Bacopa monnieri (Brahmi; Bm) phytochemicals and their therapeutically relevant mechanism of action in the colorectal cancer cell line, HT29. The nanoparticles were characterized using UV-visible spectroscopy, TEM-EDAX, zeta potential analysis, TGA, FTIR and 1H NMR spectroscopy, and HR-LC-MS. The particles (Bm-GNPs) were of polygonal shape and were stable against aggregation. They entered the target cells and inhibited the viability and clonogenicity of the cells with eight times more antiproliferative efficacy (25 ± 1.5 µg mL-1) than Bm extract (Bm-EX). In vitro studies revealed that Bm-GNPs bind tubulin (a protein crucial in cell division and a target of anticancer drugs) and disrupt its helical structure without grossly altering its tertiary conformation. Like other antitubulin agents, Bm-GNPs induced G2/M arrest and ultimately killed the cells, as confirmed using flow cytometry analyses. ZVAD-FMK-mediated global pan-caspase inhibition and the apparent absence of cleaved caspase-3 in treated cells indicated that the death did not involve the classic apoptosis pathway. Cellular ultrastructure analyses, western immunoblots, and in situ immunofluorescence visualization of cellular microtubules revealed microtubule-acetylation-independent induction of autophagy as the facilitator of cell death. Together, the data indicate strong antiproliferative efficacy and a possible mechanism of action for these designer nanoparticles. Bm-GNPs, therefore, merit further investigations, including preclinical evaluations, for their therapeutic potential as inducers of non-apoptotic cell death.


Assuntos
Autofagia , Neoplasias Colorretais , Ouro , Nanopartículas Metálicas , Humanos , Ouro/química , Ouro/farmacologia , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Neoplasias Colorretais/tratamento farmacológico , Nanopartículas Metálicas/química , Autofagia/efeitos dos fármacos , Acetilação , Microtúbulos/metabolismo , Microtúbulos/efeitos dos fármacos , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Adenocarcinoma/tratamento farmacológico , Células HT29 , Caspases/metabolismo , Compostos Fitoquímicos/farmacologia , Compostos Fitoquímicos/química , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Antineoplásicos/farmacologia , Antineoplásicos/química , Tubulina (Proteína)/metabolismo , Tubulina (Proteína)/química
2.
Cell Mol Life Sci ; 81(1): 193, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38652325

RESUMO

The acetylation of α-tubulin on lysine 40 is a well-studied post-translational modification which has been associated with the presence of long-lived stable microtubules that are more resistant to mechanical breakdown. The discovery of α-tubulin acetyltransferase 1 (ATAT1), the enzyme responsible for lysine 40 acetylation on α-tubulin in a wide range of species, including protists, nematodes, and mammals, dates to about a decade ago. However, the role of ATAT1 in different cellular activities and molecular pathways has been only recently disclosed. This review comprehensively summarizes the most recent knowledge on ATAT1 structure and substrate binding and analyses the involvement of ATAT1 in a variety of cellular processes such as cell motility, mitosis, cytoskeletal organization, and intracellular trafficking. Finally, the review highlights ATAT1 emerging roles in human diseases and discusses ATAT1 potential enzymatic and non-enzymatic roles and the current efforts in developing ATAT1 inhibitors.


Assuntos
Acetiltransferases , Proteínas dos Microtúbulos , Tubulina (Proteína) , Humanos , Acetiltransferases/metabolismo , Acetiltransferases/química , Tubulina (Proteína)/metabolismo , Tubulina (Proteína)/química , Animais , Processamento de Proteína Pós-Traducional , Acetilação , Microtúbulos/metabolismo , Mitose , Movimento Celular , Neoplasias/patologia , Neoplasias/enzimologia , Neoplasias/metabolismo , Citoesqueleto/metabolismo
3.
Biomacromolecules ; 25(2): 1282-1290, 2024 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-38251876

RESUMO

Studies of proteins from one organism in another organism's cells have shown that such exogenous proteins stick more, pointing toward coevolution of the cytoplasm and protein surface to minimize stickiness. Here we flip this question around by asking whether exogenous proteins can assemble efficiently into their target complexes in a non-native cytoplasm. We use as our model system the assembly of BtubA and BtubB from Prosthecobacter hosted in human U-2 OS cells. BtubA and B evolved from eukaryotic tubulins after horizontal gene transfer, but they have low surface sequence identity with the homologous human tubulins and do not respond to tubulin drugs such as nocodazole. In U-2 OS cells, BtubA and B assemble efficiently into dimers compared to in vitro, and the wild-type BtubA and B proteins subsequently are able to form microtubules as well. We find that generic crowding effects (Ficoll 70 in vitro) contribute significantly to efficient dimer assembly when compared to sticking interactions (U-2 OS cell lysate in vitro), consistent with the notion that a generic mechanism such as crowding can be effective at driving assembly of exogenous proteins, even when protein-cytoplasm quinary structure and sticking have been modified in a non-native cytoplasm. A simple Monte Carlo model of in vitro and in-cell interactions, treating BtubA and B as sticky dipoles in a matrix of sticky or nonsticky crowders, rationalizes all the experimental trends with two adjustable parameters and reveals nucleation as the likely mechanism for the time-scale separation between dimer- and tubule formation in-cell and in vitro.


Assuntos
Proteínas de Bactérias , Tubulina (Proteína) , Humanos , Tubulina (Proteína)/química , Proteínas de Bactérias/química , Microtúbulos/química
4.
Fitoterapia ; 173: 105781, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38128619

RESUMO

Six anthraquinones were isolated from Morinda scabrida Craib, an unexplored species of Morinda found in the tropical forest of Thailand. All six anthraquinones showed cytotoxicity against A549 lung cancer cells, with the most active compound, nordamnacanthal (MS01), exhibiting the IC50 value of 16.3 ± 2.5 µM. The cytotoxic effect was dose-dependent and led to cell morphological changes characteristic of apoptosis. In addition, flow cytometric analysis showed dose-dependent apoptosis induction and the G2/M phase cell cycle arrest, which was in agreement with the tubulin polymerization inhibitory activity of MS01. Molecular docking analysis illustrated the binding between MS01 and the α/ß-tubulin heterodimer at the colchicine binding site, and UV-visible absorption spectroscopy revealed the DNA binding capacity of MS01.


Assuntos
Neoplasias Pulmonares , Morinda , Humanos , Estrutura Molecular , Morinda/química , Proliferação de Células , Linhagem Celular Tumoral , Polimerização , Neoplasias Pulmonares/tratamento farmacológico , Simulação de Acoplamento Molecular , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo , Antraquinonas/farmacologia , Moduladores de Tubulina/farmacologia , Moduladores de Tubulina/química , Moduladores de Tubulina/metabolismo
5.
Plant Physiol Biochem ; 206: 108296, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38141401

RESUMO

The ivermectin is a potent nematocide and insecticide, which has low toxicity for humans and domestic animals, but due to low biotransformation, it can be dangerous for non-target organisms. The recent determination of ivermectin absorption and accumulation in tissues of higher plants and multiple shreds of evidence of its negative impact on plant physiology provide a basis for the search for ivermectin's molecular targets and mechanisms of action in plant cells. In this research, for the first time, the ivermectin effect on microtubules of Arabidopsis thaliana cells was studied. It was revealed that ivermectin (250 µg mL-1) disrupts the microtubule network, induces the loss of microtubule orientation, leads to microtubule curvature and shrinkage, and their longitudinal and cross-linked bundling in various cells of A. thaliana primary roots. Further, the previously proposed binding of ivermectin to the ß1-tubulin taxane site was developed and confirmed using molecular dynamics simulations of ivermectin complexes with Haemonchus contortus and A. thaliana ß1-tubulins. It was predicted that similar to other microtubule stabilizing agents ivermectin binding causes M-loop stabilization in both H. contortus and A. thaliana ß-tubulin, which leads to the enhancement of lateral contacts between subunits of adjacent protofilaments preventing microtubule depolymerization.


Assuntos
Arabidopsis , Tubulina (Proteína) , Humanos , Animais , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo , Ivermectina/farmacologia , Ivermectina/metabolismo , Arabidopsis/metabolismo , Microtúbulos/metabolismo , Sítios de Ligação
6.
Biomacromolecules ; 24(12): 5678-5686, 2023 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-37934694

RESUMO

Cells use dynamic self-assembly to construct functional structures for maintaining cellular homeostasis. However, using a natural biological small molecule to mimic this phenomenon remains challenging. This work reports the dynamic microfiber formation of nucleopeptide driven by guanosine triphosphate, the small molecule that controls microtubule polymerization in living cells. Deactivation of GTP by enzyme dissociates the fibers, which could be reactivated by adding GTP. Molecular dynamic simulation unveils the mystery of microfiber formation of GBM-1 and GTP. Moreover, the microfiber formation can also be controlled by diffusion-driven GTP gradients across a semipermeable membrane in bulk conditions and the microfluidic method in the defined droplets. This study provides a new platform to construct dynamic self-assembly materials of molecular building blocks driven by GTP.


Assuntos
Microtúbulos , Tubulina (Proteína) , Guanosina Trifosfato , Tubulina (Proteína)/química , Hidrólise , Simulação de Dinâmica Molecular
7.
Eur J Cell Biol ; 102(4): 151370, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37922811

RESUMO

A fair amount of research on microtubules since their discovery in 1963 has focused on their dynamic tips. In contrast, the microtubule lattice was long believed to be highly regular and static, and consequently received far less attention. Yet, as it turned out, the microtubule lattice is neither as regular, nor as static as previously believed: structural studies uncovered the remarkable wealth of different conformations the lattice can accommodate. In the last decade, the microtubule lattice was shown to be labile and to spontaneously undergo renovation, a phenomenon that is intimately linked to structural defects and was called "microtubule self-repair". Following this breakthrough discovery, further recent research provided a deeper understanding of the lattice self-repair mechanism, which we review here. Instrumental to these discoveries were in vitro microtubule reconstitution assays, in which microtubules are grown from the minimal components required for their dynamics. In this review, we propose a shift from the term "lattice self-repair" to "lattice dynamics", since this phenomenon is an inherent property of microtubules and can happen without microtubule damage. We focus on how in vitro microtubule reconstitution assays helped us learn (1) which types of structural variations microtubules display, (2) how these structural variations influence lattice dynamics and microtubule damage caused by mechanical stress, (3) how lattice dynamics impact tip dynamics, and (4) how microtubule-associated proteins (MAPs) can play a role in structuring the lattice. Finally, we discuss the unanswered questions about lattice dynamics and how technical advances will help us tackle these questions.


Assuntos
Proteínas Associadas aos Microtúbulos , Microtúbulos , Microtúbulos/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Tubulina (Proteína)/análise , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo
8.
Annu Rev Cell Dev Biol ; 39: 331-361, 2023 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-37843925

RESUMO

Microtubules are essential dynamic polymers composed of α/ß-tubulin heterodimers. They support intracellular trafficking, cell division, cellular motility, and other essential cellular processes. In many species, both α-tubulin and ß-tubulin are encoded by multiple genes with distinct expression profiles and functionality. Microtubules are further diversified through abundant posttranslational modifications, which are added and removed by a suite of enzymes to form complex, stereotyped cellular arrays. The genetic and chemical diversity of tubulin constitute a tubulin code that regulates intrinsic microtubule properties and is read by cellular effectors, such as molecular motors and microtubule-associated proteins, to provide spatial and temporal specificity to microtubules in cells. In this review, we synthesize the rapidly expanding tubulin code literature and highlight limitations and opportunities for the field. As complex microtubule arrays underlie essential physiological processes, a better understanding of how cells employ the tubulin code has important implications for human disease ranging from cancer to neurological disorders.


Assuntos
Microtúbulos , Tubulina (Proteína) , Humanos , Tubulina (Proteína)/genética , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo , Microtúbulos/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Processamento de Proteína Pós-Traducional/genética , Movimento Celular
9.
Eur J Cell Biol ; 102(4): 151366, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37871345

RESUMO

Microtubules are essential cytoskeletal polymers, which exhibit stochastic transitions between assembly and disassembly, known as catastrophes and rescues. Understanding of catastrophes, rescues, and their control by drugs and microtubule associated proteins (MAPs) has been informed by in vitro reconstitutions of microtubule dynamics. In such experiments microtubules are typically observed on a flat surface of the coverslip. In contrast, we have recently proposed a modified setup in which microtubules assemble from stabilized seeds, overhanging from microfabricated pedestals, so that their dynamic extensions are fully isolated from contact with the coverslip. This assay allows to eliminate potential artifacts, which may substantially affect the frequency of microtubule rescues in vitro. Here we use the pedestal assay to study the sensitivity of microtubules to paclitaxel, one of the best-known inhibitors of microtubule dynamics. By comparing observations in the conventional and the pedestal assays, we find that microtubule dynamics are substantially more sensitive to paclitaxel when the polymers can contact the coverslip. We interpret this as a consequence of the coverslip-induced microtubule assembly perturbation, leading to formation of lattice with defects, and thereby enhancing the efficiency of paclitaxel binding to microtubules in the conventional assay. To test this idea, we use vinblastine, another small-molecule inhibitor, which had been previously shown to cause microtubule growth perturbations. We find that in the pedestal assay vinblastine sensitizes microtubules to paclitaxel to the level, observed in the conventional assay. Interestingly, a minimal fragment of MAP called CLASP2, a previously characterized rescue factor, has a strong effect on microtubule rescues, regardless of the type of assay. Overall, our study underscores the role of microtubule damage in promoting rescues and highlights the utility of the in vitro pedestal assay to study microtubule dynamics modulation by tubulin inhibitors and MAPs.


Assuntos
Proteínas Associadas aos Microtúbulos , Tubulina (Proteína) , Tubulina (Proteína)/análise , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Vimblastina/farmacologia , Vimblastina/análise , Vimblastina/metabolismo , Microtúbulos/metabolismo , Paclitaxel/farmacologia , Paclitaxel/análise , Paclitaxel/metabolismo , Polímeros/análise , Polímeros/metabolismo , Polímeros/farmacologia
10.
Mater Horiz ; 10(11): 5298-5306, 2023 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-37750812

RESUMO

Cell function-associated biomolecular condensation has great potential in modulation of molecular activities. We develop a microtubule-trapping peptide that first self-assembles into nanoparticles and then in situ transforms into nanofibers via ligand-receptor interactions when targeted to tubulin. The nanofibers support the increased exposed targets for further adhering to microtubules and induce the self-assembly of microtubules into networks due to multivalent effects. Microtubule condensation with prolonged retention in cells for up to 24 h, which is 6 times longer than that of the non-transformable nanoparticle group, efficiently induces in vitro cell apoptosis and inhibits in vivo tumour growth. These smart transformable peptide materials for targeted protein condensation have the potential for improving retention and inducing cell apoptosis in tumour therapy.


Assuntos
Microtúbulos , Neoplasias , Humanos , Microtúbulos/metabolismo , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo , Proteínas/metabolismo , Peptídeos/farmacologia , Peptídeos/uso terapêutico , Peptídeos/metabolismo
11.
J Med Chem ; 66(16): 11094-11117, 2023 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-37584263

RESUMO

Endocrine resistance remains a significant problem in the clinical treatment of estrogen receptor α-positive (ERα+) breast cancer (BC). In this study, we developed a series of novel dual-functional ERα degraders based on a bridged bicyclic scaffold with selenocyano (SeCN) side chains. These compounds displayed potent ERα degradation and tubulin depolymerization activity. Among them, compounds 35s and 35t exhibited the most promising antiproliferative and ERα degradation activity in multiple ERα+ BC cell lines bearing either wild-type or mutant ERα. Meanwhile, compounds 35s and 35t disrupted the microtubule network by restraining tubulin polymerization, evidenced by 35t inducing cell cycle arrest in the G2/M phase. In MCF-7 and LCC2 xenograft models, compounds 35s and 35t remarkably suppressed tumor growth without noticeable poisonousness. Finally, this study provided guidance for developing new dual-target antitumor drug candidates for the ERα+ BC therapy, especially for the resistant variant.


Assuntos
Antineoplásicos , Neoplasias da Mama , Receptores de Estrogênio , Feminino , Humanos , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Antineoplásicos/química , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Proliferação de Células , Receptor alfa de Estrogênio/metabolismo , Células MCF-7 , Receptores de Estrogênio/antagonistas & inibidores , Tubulina (Proteína)/química , Moduladores de Tubulina/química , Moduladores de Tubulina/farmacologia
12.
Biochem Soc Trans ; 51(4): 1505-1520, 2023 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-37560910

RESUMO

Kinesin motor proteins couple mechanical movements in their motor domain to the binding and hydrolysis of ATP in their nucleotide-binding pocket. Forces produced through this 'mechanochemical' coupling are typically used to mobilize kinesin-mediated transport of cargos along microtubules or microtubule cytoskeleton remodeling. This review discusses the recent high-resolution structures (<4 Å) of kinesins bound to microtubules or tubulin complexes that have resolved outstanding questions about the basis of mechanochemical coupling, and how family-specific modifications of the motor domain can enable its use for motility and/or microtubule depolymerization.


Assuntos
Cinesinas , Tubulina (Proteína) , Cinesinas/metabolismo , Tubulina (Proteína)/análise , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo , Trifosfato de Adenosina/metabolismo , Microtúbulos/metabolismo , Miosinas
13.
Curr Drug Targets ; 24(11): 889-918, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37519203

RESUMO

Microtubules are a well-known target in cancer chemotherapy because of their critical role in cell division. Chromosome segregation during mitosis depends on the establishment of the mitotic spindle apparatus through microtubule dynamics. The disruption of microtubule dynamics through the stabilization or destabilization of microtubules results in the mitotic arrest of the cells. Microtubule-targeted drugs, which interfere with microtubule dynamics, inhibit the growth of cells at the mitotic phase and induce apoptotic cell death. The principle of microtubule-targeted drugs is to arrest the cells at mitosis and reduce their growth because cancer is a disease of unchecked cell proliferation. Many anti-microtubule agents produce significant inhibition of cancer cell growth and are widely used as chemotherapeutic drugs for the treatment of cancer. The drugs that interact with microtubules generally bind at one of the three sites vinblastine site, taxol site, or colchicine site. Colchicine binds to the interface of tubulin heterodimer and induces the depolymerization of microtubules. The colchicine binding site on microtubules is a much sought-after target in the history of anti-microtubule drug discovery. Many colchicine-binding site inhibitors have been discovered, but their use in the treatment of cancer is limited due to their dose-limiting toxicity and resistance in humans. Combination therapy can be a new treatment strategy to overcome these drawbacks of currently available microtubule-targeted anticancer drugs. This review discusses the significance of microtubules as a potential pharmacological target for cancer and stresses the necessity of finding new microtubule inhibitors to fight the disease.


Assuntos
Antineoplásicos , Neoplasias , Humanos , Células HeLa , Microtúbulos/metabolismo , Mitose , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo , Colchicina/metabolismo , Colchicina/farmacologia , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Antineoplásicos/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo
14.
Acta Biochim Biophys Sin (Shanghai) ; 55(10): 1551-1560, 2023 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-37439022

RESUMO

Microtubules are hollow α/ß-tubulin heterodimeric polymers that play critical roles in cells. In vertebrates, both α- and ß-tubulins have multiple isotypes encoded by different genes, which are intrinsic factors in regulating microtubule functions. However, the structures of microtubules composed of different tubulin isotypes, especially α-tubulin isotypes, remain largely unknown. Here, we purify recombinant tubulin heterodimers composed of different mouse α-tubulin isotypes, including α1A, α1C and α4A, with the ß-tubulin isotype ß2A. We further assemble and determine the cryo-electron microscopy (cryo-EM) structures of α1A/ß2A, α1C/ß2A, and α4A/ß2A microtubules. Our structural analysis demonstrates that α4A/ß2A microtubules exhibit longitudinal contraction between tubulin interdimers compared with α1A/ß2A and α1C/ß2A microtubules. Collectively, our findings reveal that α-tubulin isotype composition can tune microtubule structures, and also provide evidence for the "tubulin code" hypothesis.


Assuntos
Microtúbulos , Tubulina (Proteína) , Animais , Camundongos , Tubulina (Proteína)/química , Tubulina (Proteína)/genética , Microscopia Crioeletrônica , Microtúbulos/fisiologia
15.
Nature ; 620(7972): 122-127, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37407813

RESUMO

Possessing only essential genes, a minimal cell can reveal mechanisms and processes that are critical for the persistence and stability of life1,2. Here we report on how an engineered minimal cell3,4 contends with the forces of evolution compared with the Mycoplasma mycoides non-minimal cell from which it was synthetically derived. Mutation rates were the highest among all reported bacteria, but were not affected by genome minimization. Genome streamlining was costly, leading to a decrease in fitness of greater than 50%, but this deficit was regained during 2,000 generations of evolution. Despite selection acting on distinct genetic targets, increases in the maximum growth rate of the synthetic cells were comparable. Moreover, when performance was assessed by relative fitness, the minimal cell evolved 39% faster than the non-minimal cell. The only apparent constraint involved the evolution of cell size. The size of the non-minimal cell increased by 80%, whereas the minimal cell remained the same. This pattern reflected epistatic effects of mutations in ftsZ, which encodes a tubulin-homologue protein that regulates cell division and morphology5,6. Our findings demonstrate that natural selection can rapidly increase the fitness of one of the simplest autonomously growing organisms. Understanding how species with small genomes overcome evolutionary challenges provides critical insights into the persistence of host-associated endosymbionts, the stability of streamlined chassis for biotechnology and the targeted refinement of synthetically engineered cells2,7-9.


Assuntos
Evolução Molecular , Genes Essenciais , Genoma Bacteriano , Mycoplasma mycoides , Biologia Sintética , Biotecnologia/métodos , Biotecnologia/tendências , Divisão Celular , Genoma Bacteriano/genética , Mutação , Mycoplasma mycoides/citologia , Mycoplasma mycoides/genética , Mycoplasma mycoides/crescimento & desenvolvimento , Biologia Sintética/métodos , Tamanho Celular , Epistasia Genética , Seleção Genética , Aptidão Genética , Simbiose , Tubulina (Proteína)/química
16.
J Chem Theory Comput ; 19(16): 5621-5632, 2023 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-37489636

RESUMO

Simulating the conformations and functions of biological macromolecules by using all-atom (AA) models is a challenging task due to expensive computational costs. One possible strategy to solve this problem is to develop hybrid all-atom and ultra-coarse-grained (AA/UCG) models of the biological macromolecules. In the AA/UCG scheme, the interest regions are described by AA models, while the other regions are described in the UCG representation. In this study, we develop the hybrid AA/UCG models and apply them to investigate the conformational changes of microtubule-bound tubulins. The simulation results of the hybrid models elucidated the mechanism of why the taxol molecules selectively bound microtubules but not tubulin dimers. In addition, we also explore the interactions of the microtubules and dyneins. Our study shows that the hybrid AA/UCG model has great application potential in studying the function of complex biological systems.


Assuntos
Dineínas , Paclitaxel , Dineínas/análise , Dineínas/química , Dineínas/metabolismo , Paclitaxel/farmacologia , Microtúbulos/química , Tubulina (Proteína)/análise , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo , Conformação Molecular
17.
Proc Natl Acad Sci U S A ; 120(27): e2305899120, 2023 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-37364095

RESUMO

Microtubules (MTs) are large cytoskeletal polymers, composed of αß-tubulin heterodimers, capable of stochastically converting from polymerizing to depolymerizing states and vice versa. Depolymerization is coupled with hydrolysis of guanosine triphosphate (GTP) within ß-tubulin. Hydrolysis is favored in the MT lattice compared to a free heterodimer with an experimentally observed rate increase of 500- to 700-fold, corresponding to an energetic barrier lowering of 3.8 to 4.0 kcal/mol. Mutagenesis studies have implicated α-tubulin residues, α:E254 and α:D251, as catalytic residues completing the ß-tubulin active site of the lower heterodimer in the MT lattice. The mechanism for GTP hydrolysis in the free heterodimer, however, is not understood. Additionally, there has been debate concerning whether the GTP-state lattice is expanded or compacted relative to the GDP state and whether a "compacted" GDP-state lattice is required for hydrolysis. In this work, extensive quantum mechanics/molecular mechanics simulations with transition-tempered metadynamics free-energy sampling of compacted and expanded interdimer complexes, as well as a free heterodimer, have been carried out to provide clear insight into the GTP hydrolysis mechanism. α:E254 was found to be the catalytic residue in a compacted lattice, while in the expanded lattice, disruption of a key salt bridge interaction renders α:E254 less effective. The simulations reveal a barrier decrease of 3.8 ± 0.5 kcal/mol for the compacted lattice compared to a free heterodimer, in good agreement with experimental kinetic measurements. Additionally, the expanded lattice barrier was found to be 6.3 ± 0.5 kcal/mol higher than compacted, demonstrating that GTP hydrolysis is variable with lattice state and slower at the MT tip.


Assuntos
Microtúbulos , Tubulina (Proteína) , Guanosina Trifosfato , Tubulina (Proteína)/química , Hidrólise , Guanosina Difosfato/química , Microtúbulos/química
18.
Sci Rep ; 13(1): 10165, 2023 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-37349383

RESUMO

This study examines the electrical properties of isolated brain microtubules (MTs), which are long hollow cylinders assembled from αß-tubulin dimers that form cytoskeletal structures engaged in several functions. MTs are implicated in sensory functions in cilia and flagella and cellular activities that range from cell motility, vesicular traffic, and neuronal processes to cell division in the centrosomes and centrioles. We determined the electrical properties of the MTs with the loose patch clamp technique in either the presence or absence of the MT stabilizer Paclitaxel. We observed electrical oscillations at different holding potentials that responded accordingly in amplitude and polarity. At zero mV in symmetrical ionic conditions, a single MT radiated an electrical power of 10-17 W. The spectral analysis of the time records disclosed a single fundamental peak at 39 Hz in the Paclitaxel-stabilized MTs. However, a richer oscillatory response and two mean conductances were observed in the non-Paclitaxel MTs. The findings evidence that the brain MTs are electrical oscillators that behave as "ionic-based" transistors to generate, propagate, and amplify electrical signals.


Assuntos
Microtúbulos , Tubulina (Proteína) , Microtúbulos/química , Tubulina (Proteína)/química , Paclitaxel/química , Polímeros , Eletricidade
19.
Cell Biochem Biophys ; 81(2): 269-283, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37233844

RESUMO

The tubulin-microtubule system is a major target for a variety of small molecules which can interfere in cell cycle progression. Therefore, it serves as a prospective to control the incessant division of cancer cells. To identify novel inhibitors of the tubulin-microtubule system, a group of estrogen derivatives has been tested with tubulin as a target since literature surveys portray coveted behaviour from the same. Out of them, ß-Estradiol-6-one 6- (O-carboxy methyl Oxime) abbreviated as Oxime, disrupts the cytoskeleton network and induces apoptosis with nuclei fragmentation. It has been revealed from the work that Oxime targets the colchicine binding site and binds tubulin in an entropy-driven manner. This suggests that structural variation might play a key role in modulating the anti-mitotic role of estrogen derivatives. Our work reveals that Oxime might serve as a lead molecule to nurture anti-cancer research, having the potential for recovery of the vast cancer population.


Assuntos
Antimitóticos , Antineoplásicos , Tubulina (Proteína)/química , Antimitóticos/metabolismo , Moduladores de Tubulina/farmacologia , Moduladores de Tubulina/química , Moduladores de Tubulina/metabolismo , Mitose , Estudos Prospectivos , Microtúbulos/metabolismo , Estradiol/farmacologia , Estradiol/metabolismo , Sítios de Ligação , Estrogênios/metabolismo , Antineoplásicos/farmacologia , Antineoplásicos/metabolismo
20.
J Mol Graph Model ; 123: 108503, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37209440

RESUMO

The single-point mutation D26E in human ß-tubulin is associated with drug resistance seen with two anti-mitotic taxanes (paclitaxel and docetaxel) when used to treat cancers. The molecular mechanism of this resistance remains elusive. However, docetaxel and a third-generation taxane, cabazitaxel, are thought to overcome this resistance. Here, structural models of both the wildtype (WT) and D26E mutant (MT) human ß-tubulin were constructed based on the crystal structure of pig ß-tubulin in complex with docetaxel (PDB ID: 1TUB). The three taxanes were docked into the WT and MT ß-tubulin, and the resulting complexes were submitted to three independent runs of 200 ns molecular dynamic simulations, which were then averaged. MM/GBSA calculations revealed the binding energy of paclitaxel with WT and MT ß-Tubulin to be -101.5 ± 8.4 and -90.4 ± 8.9 kcal/mol, respectively. The binding energy of docetaxel was estimated to be -104.7 ± 7.0 kcal/mol with the WT and -103.8 ± 5.5 kcal/mol with the MT ß-tubulin. Interestingly, cabazitaxel was found to have a binding energy of -122.8 ± 10.8 kcal/mol against the WT and -106.2 ± 7.0 kcal/mol against the MT ß-tubulin. These results show that paclitaxel and docetaxel bound to the MT less strongly than the WT, suggesting possible drug resistance. Similarly, cabazitaxel displayed a greater binding propensity against WT and MT ß-tubulin than the other two taxanes. Furthermore, the dynamic cross-correlation matrices (DCCM) analysis suggests that the single-point mutation D26E induces a subtle dynamical difference in the ligand-binding domain. Overall, the present study revealed how the single-point mutation D26E may reduce the binding affinity of the taxanes, however, the effect of the mutation does not significantly affect the binding of cabazitaxel.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Tubulina (Proteína) , Humanos , Animais , Suínos , Tubulina (Proteína)/química , Docetaxel/farmacologia , Taxoides/farmacologia , Taxoides/química , Paclitaxel/farmacologia , Paclitaxel/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA